User login
How do SSRIs cause sexual dysfunction?
Although selective serotonin reuptake inhibitors (SSRIs) are frequently prescribed1 and are better tolerated than older antidepressants, side effects such as sexual dysfunction limit patient acceptance of these medications. DSM-IV-TR categorizes medication-induced sexual dysfunction as a type of substance-induced sexual dysfunction.2 These dysfunctions are characterized by impairment of various sexual response phases (Table 1).2,3
Estimating the true incidence and prevalence of SSRI-related sexual dysfunction can be difficult. Zimmerman et al4 compared psychiatrists’ clinical assessments of depressed patients receiving ongoing treatment with results of a standardized side effects questionnaire and found that even though psychiatrists regularly inquired about sexual side effects, on the questionnaire patients reported higher rates of almost all sexual dysfunctions. The incidence of SSRI-induced sexual dysfunction also can be difficult to ascertain because some sexual dysfunctions frequently accompany a primary psychiatric disorder5 or physical illness. Balon6 suggested that the incidence of SSRI-associated sexual dysfunction is 30% to 50%, although others have reported higher incidences.
Few quality studies have focused on identifying the exact nature and causes of SSRI treatment-emergent sexual dysfunction. This article describes mechanisms that may be fundamental to SSRI-associated sexual dysfunction.
Table 1
Sexual dysfunction and the sexual response cycle
Phase | Description | Dysfunction/disorder |
---|---|---|
Desire | Characterized by sexual fantasies and the desire to have sex | Hypoactive sexual desire disorder |
Sexual aversion disorder | ||
Hypoactive sexual desire disorder due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired desire | ||
Excitement | Subjective sense of sexual pleasure and accompanying physiologic changes | Female sexual arousal disorder |
Erectile disorder | ||
Erectile disorder due to a general medical condition | ||
Dyspareunia due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired arousal | ||
Orgasm | Peaking of sexual pleasure with release of sexual tension | Female orgasmic disorder |
Male orgasmic disorder | ||
Premature ejaculation | ||
Other sexual dysfunction due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired orgasm | ||
Resolution | A sense of general relaxation, well-being, and muscle relaxation | Postcoital dysphoria |
Postcoital headache | ||
Source: References 2,3 |
Not just serotonin
Although SSRIs are relatively selective for the serotonergic system, they affect other neurotransmitter systems as well (Table 2).7 For example, at high dosages paroxetine is believed to block norepinephrine reuptake, and it has a clinically significant anticholinergic effect. Also, sertraline is a potent reuptake inhibitor of dopamine.8 Therefore, our discussion will include these neurotransmitters.
In their dual control model of male sexual response, Bancroft et al9 discuss the interplay between excitatory and inhibitory mechanisms at the central and peripheral levels. For example, they describe the role of norepinephrine mediation in the central arousal system via the disinhibition of dopaminergic and a possible testosterone mechanism. They also point to possible inhibition of central sexual arousal by neuropeptidergic and serotonergic mechanisms.
Evidence linking serotonin to sexual dysfunction is inconclusive because there are no exclusively serotonergic agents. Drugs frequently used to test these hypotheses often affect other neurotransmitters, which means conclusions are not specific to serotonin. Animal studies of the impact of serotonin agonist and antagonist agents on mounting and ejaculation have reported inconsistent results.10 Differential roles of 5-HT1 and 5-HT2 receptor activation on sexual behavior may explain some of these inconsistencies.8 However, 1 study found that antiserotonergic pharmacologic agents enhance sexual excitation in laboratory animals,11 and a separate study showed that severing serotonergic axons in the medial forebrain bundle in male rats facilitated ejaculation.12
Monteiro et al13 found a high incidence of anorgasmia in previously orgasmic patients after they received clomipramine, which may be partially attributed to the drug’s serotonergic action. This prompted researchers to hypothesize that central serotonergic tone inhibits sexual behavior. However, based on current evidence, it would be best to consider serotonin as having a modulating effect10—as opposed to a complete inhibitory effect—on human sexual behavior.
Regarding the parasympathetic system, it was long believed that cholinergic innervations mediate penile erection. However, a more plausible hypothesis may be that parasympathetic cholinergic transmission at best has a modulating effect when other neurotransmitters—primarily the adrenergic system—are affected by concomitant pharmacologic interventions. Segraves10 proposed that cholinergic potentiating of adrenergic activity may be primarily responsible for bethanechol-induced reversal of SSRI-induced sexual dysfunction.
The adrenergic system is believed to play a role in penile erection and ejaculation.10 Adrenergic fibers innervate the vas deferens, seminal vesicles, trigone of the urinary bladder, and proximal urethra.14 Penile contractile and erectile tissue is richly innervated by the adrenergic nerve fibers.10 Ejaculation is mediated by α1-adrenergic receptors.10
Table 2
Neurotransmitters affected by SSRIs
SSRI | Neurotransmitters |
---|---|
Citalopram | 5-HT |
Escitalopram | 5-HT |
Fluoxetine | 5-HT, NE, DA |
Fluvoxamine | 5-HT |
Paroxetine | 5-HT, NE, Ach |
Sertraline | 5-HT, NE, DA |
5-HT: serotonin; Ach: acetylcholine; DA: dopamine; NE: norepinephrine; SSRIs: selective serotonin reuptake inhibitors | |
Source: Reference 7 |
The role of nitric oxide synthase
The advent of sildenafil underscored the importance of nitric oxide-mediated relaxation pathways in treating erectile dysfunction. Nitric oxide plays an important role in mediating the penile vasculature changes essential for erection and also is hypothesized to promote penile smooth muscle relaxation via cyclic guanosine monophosphate, there-by contributing to physiologic erection.15 Paroxetine is known to inhibit nitric oxide synthase, which reduces nitric oxide levels. The exact mechanism of this interaction remains unclear; however, it is hypothesized that 3 nitric oxide synthase isoenzymes are structurally similar to cytochrome P450 (CYP450). Paroxetine is a strong CYP2D6 inhibitor, which contributes to low nitric oxide levels in patients taking the drug.16
SSRIs and sexual response
Because decreased libido is part of depressive psychopathology,5 it is difficult to attribute loss of sexual desire directly to SSRIs. Nonetheless, SSRIs are associated with a risk of clinically significant loss of sexual desire that may resemble moderate to severe hypoactive sexual desire disorder.17 Reduced mesolimbic dopaminergic activity as a result of inhibitory serotonergic midbrain raphe nuclei projections is 1 possible cause.18 This hypothesis has lead investigators to examine drug targets in the CNS for hypoactive sexual desire disorder that would inhibit serotonergic tone and lead to brain dopaminergic system stimulation.
Another putative hypothesis for SSRI-induced loss of sexual desire is the role of 5-HT1A receptor-mediated norepinephrine neurotransmission. Because the sympathetic nervous system is believed to be involved in genital arousal in women, a small study analyzed the effect of sympathetic activation on SSRI-induced sexual dysfunction.19 Women who received paroxetine and sertraline—both are highly selective for 5-HT1A—showed improvement in sexual arousal and orgasm after taking ephedrine before sexual activity.19 Women who took fluoxetine, which is less selective for 5-HT1A, show no change or decreased sexual function.
SSRIs are associated with reduced nocturnal penile erections and severe erectile dysfunction, but the relationship is not robust.17 SSRI-induced suppression of rapid eye movement sleep20 may partially explain reduced nocturnal and early morning erections. Supraspinal areas and preganglionic sacral neurons involved in sexual excitement also are reported to have substantial serotonergic activity, which suggests that serotonin has a direct role in erectile dysfunction at a comparative peripheral level.21 However, a recent study17 found no difference in flaccid and peak systolic velocity when comparing patients taking SSRIs with those who do not. This indicates that SSRIs’ affect on spontaneous and sexually aroused erections may be mediated at both central and peripheral levels.
Delayed ejaculation frequently is associated with SSRIs17 and usually is not caused by depressive psychopathology.22 Animal studies show that increased serotonergic tone predicts ejaculatory latency by acting as an inhibitor at the hypothalamus level.23 In contrast, noradrenergic tone enhances ejaculation.24 Antidepressants that increase noradrenaline levels and serotonin levels—such as serotonin-norepinephrine reuptake inhibitors—induce milder ejaculatory delay.17
A recent study17 found impaired climax and reduced libido in partners of patients using SSRIs. Patients receiving SSRIs report less frequent sexual intercourse and heightened guilt associated with masturbation, and SSRIs are associated with psychosocial factors such as higher stress at work and increased risk of conflicts with partners and other family members.17 In addition to biologic mechanisms, these psychosocial and intra-couple factors might contribute to SSRI-associated sexual dysfunction. However, because the temporal association between SSRI use and psychosocial dysfunction is ambiguous, this hypothesis should be interpreted with caution.
SSRIs have been associated with lower serum levels of luteinizing hormone, follicle-stimulating hormone, and testosterone.25 However, these findings need to be replicated before drawing firm conclusions on intermediary role of hormones in SSRI-emergent sexual dysfunction.
Be aware that other medications may contribute to sexual dysfunction experienced by a patient receiving an SSRI (Table 3).26
Table 3
Other than SSRIs, which medications can cause sexual dysfunction?
Psychotropics |
---|
Amphetamines |
Anticonvulsants |
Antidepressants
|
Antipsychotics |
Benzodiazepines |
Nonpsychotropics |
Antihypertensives
|
Digoxin |
Histamine blockers |
Lipid-lowering agents |
Narcotics |
Oral contraceptives |
SSRIs: selective serotonin reuptake inhibitors |
Source: Reference 26 |
SSRIs for premature ejaculation?
Because SSRIs can cause delayed ejaculation, they have been used off-label to treat premature ejaculation.27 For this purpose, paroxetine and sertraline have been prescribed with daily or on-demand dosing before sexual intercourse28 and daily fluoxetine has been used.29 However, none of these SSRIs is FDA-approved for treating premature ejaculation, daily dosing of SSRIs exposes patients to undesirable side effects, and inconsistent use of paroxetine can lead to discontinuation syndrome.
These concerns have lead researchers to seek an SSRI that could be used on as-needed basis and would not cause some of the deleterious side effects associated with current SSRIs. The short-acting SSRI dapoxetine is in FDA review for treating premature ejaculation; the drug is approved for this use in several countries outside the United States.30
Sexual health education
Because sexual dysfunction can be caused by underlying psychopathology or physical illness, it is essential to obtain a detailed sexual history at your patient’s initial assessment and at every follow-up visit. Patients and providers may be guarded when discussing sexual health, which can be a barrier to providing comprehensive health care. The organizations listed in Related Resources can provide information and materials to help patients and health care providers better understand sexual health. Addressing the importance of sexual health in a comprehensive, culturally sensitive manner can substantially improve our patients’ medication compliance and prognosis.
- American Association of Sexuality Educators, Counselors, and Therapists. www.aasect.org.
- Sexual Medicine and Wellness Center. www.methodistsexualwellness.com.
- The Sexual Health Network. www.sexualhealth.com.
- International Society for the Study of Women’s Sexual Health. www.isswsh.org.
Drug Brand Names
- Bethanechol • Urecholine
- Citalopram • Celexa
- Clomipramine • Anafranil
- Dapoxetine • Priligy
- Digoxin • Lanoxin
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Paroxetine • Paxil
- Sertraline • Zoloft
- Sildenafil • Viagra
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848-856.
2. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
3. Sadock BJ, Sadock VA. Abnormal sexuality and sexual dysfunctions. In Sadock BJ, Sadock VA. Kaplan & Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry. 10th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:689-705.
4. Zimmerman M, Galione JN, Attiullah N, et al. Underrecognition of clinically significant side effects in depressed outpatients. J Clin Psychiatry. 2010;71(4):484-490.
5. Casper RC, Redmont DE, Jr, Katz MM, et al. Somatic symptoms in primary affective disorder: presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42:1098-1104.
6. Balon R. SSRI-associated sexual dysfunction. Am J Psychiatry. 2006;163(9):1504-1509; quiz 1664.
7. Schatzberg AF, Nemeroff CB. eds. The American Psychiatric Publishing textbook of psychopharmacology. 4th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2009.
8. Schatzberg AF, Cole JO, DeBattista C. Antidepressants. In: Schatzberg AF, Cole JO, DeBattista C. Manual of clinical psychopharmacology. 6th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2007:37-168.
9. Bancroft J, Janssen E. The dual control model of male sexual response: a theoretical approach to centrally mediated erectile dysfunction. Neurosci Biobehav Rev. 2000;24(5):571-579.
10. Segraves RT. Effects of psychotropic drugs on human erection and ejaculation. Arch Gen Psychiatry. 1989;46(3):275-284.
11. Tagliamonte A, Tagliamonte P, Gessa GL, et al. Compulsive sexual activity induced by p-chlorophenylalanine in normal and pinealectomized male rats. Science. 1969;166(911):1433-1435.
12. Rodriguez M, Castro R, Hernandez G, et al. Different roles of catecholaminergic and serotoninergic neurons of the medial forebrain bundle on male rat sexual behavior. Physiol Behav. 1984;33(1):5-11.
13. Monteiro WO, Noshirvani HF, Marks IM, et al. Anorgasmia from clomipramine in obsessive-compulsive disorder. A controlled trial. Br J Psychiatry. 1987;151:107-112.
14. Kleeman FJ. The physiology of the internal urinary sphincter. J Urol. 1970;104(4):549-554.
15. Stahl SM. How psychiatrists can build new therapies for impotence. J Clin Psychiatry. 1998;59(2):47-48.
16. Bredt DS, Hwang PM, Glatt CE, et al. Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature. 1991;351(6329):714-718.
17. Corona G, Ricca V, Bandini E, et al. Selective serotonin reuptake inhibitor-induced sexual dysfunction. J Sex Med. 2009;6(5):1259-1269.
18. Pfaus JG. Pathways of sexual desire. J Sex Med. 2009;6(6):1506-1533.
19. Ahrold TK, Meston CM. Effects of SNS activation on SSRI-induced sexual side effects differ by SSRI. J Sex Marital Ther. 2009;35(4):311-319.
20. Hirshkowitz M, Schmidt MH. Sleep-related erections: clinical perspectives and neural mechanisms. Sleep Med Rev. 2005;9(4):311-329.
21. Miner MM, Seftel AD. Centrally acting mechanisms for the treatment of male sexual dysfunction. Urol Clin North Am. 2007;34(4):483-496, v.
22. Labbate LA, Grimes J, Hines A, et al. Sexual dysfunction induced by serotonin reuptake antidepressants. J Sex Marital Ther. 1998;24(1):3-12.
23. Waldinger MD. The neurobiological approach to premature ejaculation. J Urol. 2002;168(6):2359-2367.
24. Meston CM, Frohlich PF. The neurobiology of sexual function. Arch Gen Psychiatry. 2000;57(11):1012-1030.
25. Safarinejad MR. Evaluation of endocrine profile and hypothalamic-pituitary-testis axis in selective serotonin reuptake inhibitor-induced male sexual dysfunction. J Clin Psychopharmacol. 2008;28(4):418-423.
26. Sajith SG, Morgan C, Clarke D. Pharmacological management of inappropriate sexual behaviours: a review of its evidence, rationale and scope in relation to men with intellectual disabilities. J Intellect Disabil Res. 2008;52(12):1078-1090.
27. Giuliano F, Hellstrom WJ. The pharmacological treatment of premature ejaculation. BJU Int. 2008;102(6):668-675.
28. Kim SW, Paick JS. Short-term analysis of the effects of as needed use of sertraline at 5 PM for the treatment of premature ejaculation. Urology. 1999;54(3):544-547.
29. Waldinger MD, Zwinderman AH, Schweitzer DH, et al. Relevance of methodological design for the interpretation of efficacy of drug treatment of premature ejaculation: a systematic review and meta-analysis. Int J Impot Res. 2004;16(4):369-381.
30. Kendirci M, Salem E, Hellstrom WJ. Dapoxetine, a novel selective serotonin transport inhibitor for the treatment of premature ejaculation. Ther Clin Risk Manag. 2007;3(2):277-289.
Although selective serotonin reuptake inhibitors (SSRIs) are frequently prescribed1 and are better tolerated than older antidepressants, side effects such as sexual dysfunction limit patient acceptance of these medications. DSM-IV-TR categorizes medication-induced sexual dysfunction as a type of substance-induced sexual dysfunction.2 These dysfunctions are characterized by impairment of various sexual response phases (Table 1).2,3
Estimating the true incidence and prevalence of SSRI-related sexual dysfunction can be difficult. Zimmerman et al4 compared psychiatrists’ clinical assessments of depressed patients receiving ongoing treatment with results of a standardized side effects questionnaire and found that even though psychiatrists regularly inquired about sexual side effects, on the questionnaire patients reported higher rates of almost all sexual dysfunctions. The incidence of SSRI-induced sexual dysfunction also can be difficult to ascertain because some sexual dysfunctions frequently accompany a primary psychiatric disorder5 or physical illness. Balon6 suggested that the incidence of SSRI-associated sexual dysfunction is 30% to 50%, although others have reported higher incidences.
Few quality studies have focused on identifying the exact nature and causes of SSRI treatment-emergent sexual dysfunction. This article describes mechanisms that may be fundamental to SSRI-associated sexual dysfunction.
Table 1
Sexual dysfunction and the sexual response cycle
Phase | Description | Dysfunction/disorder |
---|---|---|
Desire | Characterized by sexual fantasies and the desire to have sex | Hypoactive sexual desire disorder |
Sexual aversion disorder | ||
Hypoactive sexual desire disorder due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired desire | ||
Excitement | Subjective sense of sexual pleasure and accompanying physiologic changes | Female sexual arousal disorder |
Erectile disorder | ||
Erectile disorder due to a general medical condition | ||
Dyspareunia due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired arousal | ||
Orgasm | Peaking of sexual pleasure with release of sexual tension | Female orgasmic disorder |
Male orgasmic disorder | ||
Premature ejaculation | ||
Other sexual dysfunction due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired orgasm | ||
Resolution | A sense of general relaxation, well-being, and muscle relaxation | Postcoital dysphoria |
Postcoital headache | ||
Source: References 2,3 |
Not just serotonin
Although SSRIs are relatively selective for the serotonergic system, they affect other neurotransmitter systems as well (Table 2).7 For example, at high dosages paroxetine is believed to block norepinephrine reuptake, and it has a clinically significant anticholinergic effect. Also, sertraline is a potent reuptake inhibitor of dopamine.8 Therefore, our discussion will include these neurotransmitters.
In their dual control model of male sexual response, Bancroft et al9 discuss the interplay between excitatory and inhibitory mechanisms at the central and peripheral levels. For example, they describe the role of norepinephrine mediation in the central arousal system via the disinhibition of dopaminergic and a possible testosterone mechanism. They also point to possible inhibition of central sexual arousal by neuropeptidergic and serotonergic mechanisms.
Evidence linking serotonin to sexual dysfunction is inconclusive because there are no exclusively serotonergic agents. Drugs frequently used to test these hypotheses often affect other neurotransmitters, which means conclusions are not specific to serotonin. Animal studies of the impact of serotonin agonist and antagonist agents on mounting and ejaculation have reported inconsistent results.10 Differential roles of 5-HT1 and 5-HT2 receptor activation on sexual behavior may explain some of these inconsistencies.8 However, 1 study found that antiserotonergic pharmacologic agents enhance sexual excitation in laboratory animals,11 and a separate study showed that severing serotonergic axons in the medial forebrain bundle in male rats facilitated ejaculation.12
Monteiro et al13 found a high incidence of anorgasmia in previously orgasmic patients after they received clomipramine, which may be partially attributed to the drug’s serotonergic action. This prompted researchers to hypothesize that central serotonergic tone inhibits sexual behavior. However, based on current evidence, it would be best to consider serotonin as having a modulating effect10—as opposed to a complete inhibitory effect—on human sexual behavior.
Regarding the parasympathetic system, it was long believed that cholinergic innervations mediate penile erection. However, a more plausible hypothesis may be that parasympathetic cholinergic transmission at best has a modulating effect when other neurotransmitters—primarily the adrenergic system—are affected by concomitant pharmacologic interventions. Segraves10 proposed that cholinergic potentiating of adrenergic activity may be primarily responsible for bethanechol-induced reversal of SSRI-induced sexual dysfunction.
The adrenergic system is believed to play a role in penile erection and ejaculation.10 Adrenergic fibers innervate the vas deferens, seminal vesicles, trigone of the urinary bladder, and proximal urethra.14 Penile contractile and erectile tissue is richly innervated by the adrenergic nerve fibers.10 Ejaculation is mediated by α1-adrenergic receptors.10
Table 2
Neurotransmitters affected by SSRIs
SSRI | Neurotransmitters |
---|---|
Citalopram | 5-HT |
Escitalopram | 5-HT |
Fluoxetine | 5-HT, NE, DA |
Fluvoxamine | 5-HT |
Paroxetine | 5-HT, NE, Ach |
Sertraline | 5-HT, NE, DA |
5-HT: serotonin; Ach: acetylcholine; DA: dopamine; NE: norepinephrine; SSRIs: selective serotonin reuptake inhibitors | |
Source: Reference 7 |
The role of nitric oxide synthase
The advent of sildenafil underscored the importance of nitric oxide-mediated relaxation pathways in treating erectile dysfunction. Nitric oxide plays an important role in mediating the penile vasculature changes essential for erection and also is hypothesized to promote penile smooth muscle relaxation via cyclic guanosine monophosphate, there-by contributing to physiologic erection.15 Paroxetine is known to inhibit nitric oxide synthase, which reduces nitric oxide levels. The exact mechanism of this interaction remains unclear; however, it is hypothesized that 3 nitric oxide synthase isoenzymes are structurally similar to cytochrome P450 (CYP450). Paroxetine is a strong CYP2D6 inhibitor, which contributes to low nitric oxide levels in patients taking the drug.16
SSRIs and sexual response
Because decreased libido is part of depressive psychopathology,5 it is difficult to attribute loss of sexual desire directly to SSRIs. Nonetheless, SSRIs are associated with a risk of clinically significant loss of sexual desire that may resemble moderate to severe hypoactive sexual desire disorder.17 Reduced mesolimbic dopaminergic activity as a result of inhibitory serotonergic midbrain raphe nuclei projections is 1 possible cause.18 This hypothesis has lead investigators to examine drug targets in the CNS for hypoactive sexual desire disorder that would inhibit serotonergic tone and lead to brain dopaminergic system stimulation.
Another putative hypothesis for SSRI-induced loss of sexual desire is the role of 5-HT1A receptor-mediated norepinephrine neurotransmission. Because the sympathetic nervous system is believed to be involved in genital arousal in women, a small study analyzed the effect of sympathetic activation on SSRI-induced sexual dysfunction.19 Women who received paroxetine and sertraline—both are highly selective for 5-HT1A—showed improvement in sexual arousal and orgasm after taking ephedrine before sexual activity.19 Women who took fluoxetine, which is less selective for 5-HT1A, show no change or decreased sexual function.
SSRIs are associated with reduced nocturnal penile erections and severe erectile dysfunction, but the relationship is not robust.17 SSRI-induced suppression of rapid eye movement sleep20 may partially explain reduced nocturnal and early morning erections. Supraspinal areas and preganglionic sacral neurons involved in sexual excitement also are reported to have substantial serotonergic activity, which suggests that serotonin has a direct role in erectile dysfunction at a comparative peripheral level.21 However, a recent study17 found no difference in flaccid and peak systolic velocity when comparing patients taking SSRIs with those who do not. This indicates that SSRIs’ affect on spontaneous and sexually aroused erections may be mediated at both central and peripheral levels.
Delayed ejaculation frequently is associated with SSRIs17 and usually is not caused by depressive psychopathology.22 Animal studies show that increased serotonergic tone predicts ejaculatory latency by acting as an inhibitor at the hypothalamus level.23 In contrast, noradrenergic tone enhances ejaculation.24 Antidepressants that increase noradrenaline levels and serotonin levels—such as serotonin-norepinephrine reuptake inhibitors—induce milder ejaculatory delay.17
A recent study17 found impaired climax and reduced libido in partners of patients using SSRIs. Patients receiving SSRIs report less frequent sexual intercourse and heightened guilt associated with masturbation, and SSRIs are associated with psychosocial factors such as higher stress at work and increased risk of conflicts with partners and other family members.17 In addition to biologic mechanisms, these psychosocial and intra-couple factors might contribute to SSRI-associated sexual dysfunction. However, because the temporal association between SSRI use and psychosocial dysfunction is ambiguous, this hypothesis should be interpreted with caution.
SSRIs have been associated with lower serum levels of luteinizing hormone, follicle-stimulating hormone, and testosterone.25 However, these findings need to be replicated before drawing firm conclusions on intermediary role of hormones in SSRI-emergent sexual dysfunction.
Be aware that other medications may contribute to sexual dysfunction experienced by a patient receiving an SSRI (Table 3).26
Table 3
Other than SSRIs, which medications can cause sexual dysfunction?
Psychotropics |
---|
Amphetamines |
Anticonvulsants |
Antidepressants
|
Antipsychotics |
Benzodiazepines |
Nonpsychotropics |
Antihypertensives
|
Digoxin |
Histamine blockers |
Lipid-lowering agents |
Narcotics |
Oral contraceptives |
SSRIs: selective serotonin reuptake inhibitors |
Source: Reference 26 |
SSRIs for premature ejaculation?
Because SSRIs can cause delayed ejaculation, they have been used off-label to treat premature ejaculation.27 For this purpose, paroxetine and sertraline have been prescribed with daily or on-demand dosing before sexual intercourse28 and daily fluoxetine has been used.29 However, none of these SSRIs is FDA-approved for treating premature ejaculation, daily dosing of SSRIs exposes patients to undesirable side effects, and inconsistent use of paroxetine can lead to discontinuation syndrome.
These concerns have lead researchers to seek an SSRI that could be used on as-needed basis and would not cause some of the deleterious side effects associated with current SSRIs. The short-acting SSRI dapoxetine is in FDA review for treating premature ejaculation; the drug is approved for this use in several countries outside the United States.30
Sexual health education
Because sexual dysfunction can be caused by underlying psychopathology or physical illness, it is essential to obtain a detailed sexual history at your patient’s initial assessment and at every follow-up visit. Patients and providers may be guarded when discussing sexual health, which can be a barrier to providing comprehensive health care. The organizations listed in Related Resources can provide information and materials to help patients and health care providers better understand sexual health. Addressing the importance of sexual health in a comprehensive, culturally sensitive manner can substantially improve our patients’ medication compliance and prognosis.
- American Association of Sexuality Educators, Counselors, and Therapists. www.aasect.org.
- Sexual Medicine and Wellness Center. www.methodistsexualwellness.com.
- The Sexual Health Network. www.sexualhealth.com.
- International Society for the Study of Women’s Sexual Health. www.isswsh.org.
Drug Brand Names
- Bethanechol • Urecholine
- Citalopram • Celexa
- Clomipramine • Anafranil
- Dapoxetine • Priligy
- Digoxin • Lanoxin
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Paroxetine • Paxil
- Sertraline • Zoloft
- Sildenafil • Viagra
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Although selective serotonin reuptake inhibitors (SSRIs) are frequently prescribed1 and are better tolerated than older antidepressants, side effects such as sexual dysfunction limit patient acceptance of these medications. DSM-IV-TR categorizes medication-induced sexual dysfunction as a type of substance-induced sexual dysfunction.2 These dysfunctions are characterized by impairment of various sexual response phases (Table 1).2,3
Estimating the true incidence and prevalence of SSRI-related sexual dysfunction can be difficult. Zimmerman et al4 compared psychiatrists’ clinical assessments of depressed patients receiving ongoing treatment with results of a standardized side effects questionnaire and found that even though psychiatrists regularly inquired about sexual side effects, on the questionnaire patients reported higher rates of almost all sexual dysfunctions. The incidence of SSRI-induced sexual dysfunction also can be difficult to ascertain because some sexual dysfunctions frequently accompany a primary psychiatric disorder5 or physical illness. Balon6 suggested that the incidence of SSRI-associated sexual dysfunction is 30% to 50%, although others have reported higher incidences.
Few quality studies have focused on identifying the exact nature and causes of SSRI treatment-emergent sexual dysfunction. This article describes mechanisms that may be fundamental to SSRI-associated sexual dysfunction.
Table 1
Sexual dysfunction and the sexual response cycle
Phase | Description | Dysfunction/disorder |
---|---|---|
Desire | Characterized by sexual fantasies and the desire to have sex | Hypoactive sexual desire disorder |
Sexual aversion disorder | ||
Hypoactive sexual desire disorder due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired desire | ||
Excitement | Subjective sense of sexual pleasure and accompanying physiologic changes | Female sexual arousal disorder |
Erectile disorder | ||
Erectile disorder due to a general medical condition | ||
Dyspareunia due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired arousal | ||
Orgasm | Peaking of sexual pleasure with release of sexual tension | Female orgasmic disorder |
Male orgasmic disorder | ||
Premature ejaculation | ||
Other sexual dysfunction due to a general medical condition | ||
Substance-induced sexual dysfunction with impaired orgasm | ||
Resolution | A sense of general relaxation, well-being, and muscle relaxation | Postcoital dysphoria |
Postcoital headache | ||
Source: References 2,3 |
Not just serotonin
Although SSRIs are relatively selective for the serotonergic system, they affect other neurotransmitter systems as well (Table 2).7 For example, at high dosages paroxetine is believed to block norepinephrine reuptake, and it has a clinically significant anticholinergic effect. Also, sertraline is a potent reuptake inhibitor of dopamine.8 Therefore, our discussion will include these neurotransmitters.
In their dual control model of male sexual response, Bancroft et al9 discuss the interplay between excitatory and inhibitory mechanisms at the central and peripheral levels. For example, they describe the role of norepinephrine mediation in the central arousal system via the disinhibition of dopaminergic and a possible testosterone mechanism. They also point to possible inhibition of central sexual arousal by neuropeptidergic and serotonergic mechanisms.
Evidence linking serotonin to sexual dysfunction is inconclusive because there are no exclusively serotonergic agents. Drugs frequently used to test these hypotheses often affect other neurotransmitters, which means conclusions are not specific to serotonin. Animal studies of the impact of serotonin agonist and antagonist agents on mounting and ejaculation have reported inconsistent results.10 Differential roles of 5-HT1 and 5-HT2 receptor activation on sexual behavior may explain some of these inconsistencies.8 However, 1 study found that antiserotonergic pharmacologic agents enhance sexual excitation in laboratory animals,11 and a separate study showed that severing serotonergic axons in the medial forebrain bundle in male rats facilitated ejaculation.12
Monteiro et al13 found a high incidence of anorgasmia in previously orgasmic patients after they received clomipramine, which may be partially attributed to the drug’s serotonergic action. This prompted researchers to hypothesize that central serotonergic tone inhibits sexual behavior. However, based on current evidence, it would be best to consider serotonin as having a modulating effect10—as opposed to a complete inhibitory effect—on human sexual behavior.
Regarding the parasympathetic system, it was long believed that cholinergic innervations mediate penile erection. However, a more plausible hypothesis may be that parasympathetic cholinergic transmission at best has a modulating effect when other neurotransmitters—primarily the adrenergic system—are affected by concomitant pharmacologic interventions. Segraves10 proposed that cholinergic potentiating of adrenergic activity may be primarily responsible for bethanechol-induced reversal of SSRI-induced sexual dysfunction.
The adrenergic system is believed to play a role in penile erection and ejaculation.10 Adrenergic fibers innervate the vas deferens, seminal vesicles, trigone of the urinary bladder, and proximal urethra.14 Penile contractile and erectile tissue is richly innervated by the adrenergic nerve fibers.10 Ejaculation is mediated by α1-adrenergic receptors.10
Table 2
Neurotransmitters affected by SSRIs
SSRI | Neurotransmitters |
---|---|
Citalopram | 5-HT |
Escitalopram | 5-HT |
Fluoxetine | 5-HT, NE, DA |
Fluvoxamine | 5-HT |
Paroxetine | 5-HT, NE, Ach |
Sertraline | 5-HT, NE, DA |
5-HT: serotonin; Ach: acetylcholine; DA: dopamine; NE: norepinephrine; SSRIs: selective serotonin reuptake inhibitors | |
Source: Reference 7 |
The role of nitric oxide synthase
The advent of sildenafil underscored the importance of nitric oxide-mediated relaxation pathways in treating erectile dysfunction. Nitric oxide plays an important role in mediating the penile vasculature changes essential for erection and also is hypothesized to promote penile smooth muscle relaxation via cyclic guanosine monophosphate, there-by contributing to physiologic erection.15 Paroxetine is known to inhibit nitric oxide synthase, which reduces nitric oxide levels. The exact mechanism of this interaction remains unclear; however, it is hypothesized that 3 nitric oxide synthase isoenzymes are structurally similar to cytochrome P450 (CYP450). Paroxetine is a strong CYP2D6 inhibitor, which contributes to low nitric oxide levels in patients taking the drug.16
SSRIs and sexual response
Because decreased libido is part of depressive psychopathology,5 it is difficult to attribute loss of sexual desire directly to SSRIs. Nonetheless, SSRIs are associated with a risk of clinically significant loss of sexual desire that may resemble moderate to severe hypoactive sexual desire disorder.17 Reduced mesolimbic dopaminergic activity as a result of inhibitory serotonergic midbrain raphe nuclei projections is 1 possible cause.18 This hypothesis has lead investigators to examine drug targets in the CNS for hypoactive sexual desire disorder that would inhibit serotonergic tone and lead to brain dopaminergic system stimulation.
Another putative hypothesis for SSRI-induced loss of sexual desire is the role of 5-HT1A receptor-mediated norepinephrine neurotransmission. Because the sympathetic nervous system is believed to be involved in genital arousal in women, a small study analyzed the effect of sympathetic activation on SSRI-induced sexual dysfunction.19 Women who received paroxetine and sertraline—both are highly selective for 5-HT1A—showed improvement in sexual arousal and orgasm after taking ephedrine before sexual activity.19 Women who took fluoxetine, which is less selective for 5-HT1A, show no change or decreased sexual function.
SSRIs are associated with reduced nocturnal penile erections and severe erectile dysfunction, but the relationship is not robust.17 SSRI-induced suppression of rapid eye movement sleep20 may partially explain reduced nocturnal and early morning erections. Supraspinal areas and preganglionic sacral neurons involved in sexual excitement also are reported to have substantial serotonergic activity, which suggests that serotonin has a direct role in erectile dysfunction at a comparative peripheral level.21 However, a recent study17 found no difference in flaccid and peak systolic velocity when comparing patients taking SSRIs with those who do not. This indicates that SSRIs’ affect on spontaneous and sexually aroused erections may be mediated at both central and peripheral levels.
Delayed ejaculation frequently is associated with SSRIs17 and usually is not caused by depressive psychopathology.22 Animal studies show that increased serotonergic tone predicts ejaculatory latency by acting as an inhibitor at the hypothalamus level.23 In contrast, noradrenergic tone enhances ejaculation.24 Antidepressants that increase noradrenaline levels and serotonin levels—such as serotonin-norepinephrine reuptake inhibitors—induce milder ejaculatory delay.17
A recent study17 found impaired climax and reduced libido in partners of patients using SSRIs. Patients receiving SSRIs report less frequent sexual intercourse and heightened guilt associated with masturbation, and SSRIs are associated with psychosocial factors such as higher stress at work and increased risk of conflicts with partners and other family members.17 In addition to biologic mechanisms, these psychosocial and intra-couple factors might contribute to SSRI-associated sexual dysfunction. However, because the temporal association between SSRI use and psychosocial dysfunction is ambiguous, this hypothesis should be interpreted with caution.
SSRIs have been associated with lower serum levels of luteinizing hormone, follicle-stimulating hormone, and testosterone.25 However, these findings need to be replicated before drawing firm conclusions on intermediary role of hormones in SSRI-emergent sexual dysfunction.
Be aware that other medications may contribute to sexual dysfunction experienced by a patient receiving an SSRI (Table 3).26
Table 3
Other than SSRIs, which medications can cause sexual dysfunction?
Psychotropics |
---|
Amphetamines |
Anticonvulsants |
Antidepressants
|
Antipsychotics |
Benzodiazepines |
Nonpsychotropics |
Antihypertensives
|
Digoxin |
Histamine blockers |
Lipid-lowering agents |
Narcotics |
Oral contraceptives |
SSRIs: selective serotonin reuptake inhibitors |
Source: Reference 26 |
SSRIs for premature ejaculation?
Because SSRIs can cause delayed ejaculation, they have been used off-label to treat premature ejaculation.27 For this purpose, paroxetine and sertraline have been prescribed with daily or on-demand dosing before sexual intercourse28 and daily fluoxetine has been used.29 However, none of these SSRIs is FDA-approved for treating premature ejaculation, daily dosing of SSRIs exposes patients to undesirable side effects, and inconsistent use of paroxetine can lead to discontinuation syndrome.
These concerns have lead researchers to seek an SSRI that could be used on as-needed basis and would not cause some of the deleterious side effects associated with current SSRIs. The short-acting SSRI dapoxetine is in FDA review for treating premature ejaculation; the drug is approved for this use in several countries outside the United States.30
Sexual health education
Because sexual dysfunction can be caused by underlying psychopathology or physical illness, it is essential to obtain a detailed sexual history at your patient’s initial assessment and at every follow-up visit. Patients and providers may be guarded when discussing sexual health, which can be a barrier to providing comprehensive health care. The organizations listed in Related Resources can provide information and materials to help patients and health care providers better understand sexual health. Addressing the importance of sexual health in a comprehensive, culturally sensitive manner can substantially improve our patients’ medication compliance and prognosis.
- American Association of Sexuality Educators, Counselors, and Therapists. www.aasect.org.
- Sexual Medicine and Wellness Center. www.methodistsexualwellness.com.
- The Sexual Health Network. www.sexualhealth.com.
- International Society for the Study of Women’s Sexual Health. www.isswsh.org.
Drug Brand Names
- Bethanechol • Urecholine
- Citalopram • Celexa
- Clomipramine • Anafranil
- Dapoxetine • Priligy
- Digoxin • Lanoxin
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Paroxetine • Paxil
- Sertraline • Zoloft
- Sildenafil • Viagra
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848-856.
2. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
3. Sadock BJ, Sadock VA. Abnormal sexuality and sexual dysfunctions. In Sadock BJ, Sadock VA. Kaplan & Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry. 10th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:689-705.
4. Zimmerman M, Galione JN, Attiullah N, et al. Underrecognition of clinically significant side effects in depressed outpatients. J Clin Psychiatry. 2010;71(4):484-490.
5. Casper RC, Redmont DE, Jr, Katz MM, et al. Somatic symptoms in primary affective disorder: presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42:1098-1104.
6. Balon R. SSRI-associated sexual dysfunction. Am J Psychiatry. 2006;163(9):1504-1509; quiz 1664.
7. Schatzberg AF, Nemeroff CB. eds. The American Psychiatric Publishing textbook of psychopharmacology. 4th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2009.
8. Schatzberg AF, Cole JO, DeBattista C. Antidepressants. In: Schatzberg AF, Cole JO, DeBattista C. Manual of clinical psychopharmacology. 6th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2007:37-168.
9. Bancroft J, Janssen E. The dual control model of male sexual response: a theoretical approach to centrally mediated erectile dysfunction. Neurosci Biobehav Rev. 2000;24(5):571-579.
10. Segraves RT. Effects of psychotropic drugs on human erection and ejaculation. Arch Gen Psychiatry. 1989;46(3):275-284.
11. Tagliamonte A, Tagliamonte P, Gessa GL, et al. Compulsive sexual activity induced by p-chlorophenylalanine in normal and pinealectomized male rats. Science. 1969;166(911):1433-1435.
12. Rodriguez M, Castro R, Hernandez G, et al. Different roles of catecholaminergic and serotoninergic neurons of the medial forebrain bundle on male rat sexual behavior. Physiol Behav. 1984;33(1):5-11.
13. Monteiro WO, Noshirvani HF, Marks IM, et al. Anorgasmia from clomipramine in obsessive-compulsive disorder. A controlled trial. Br J Psychiatry. 1987;151:107-112.
14. Kleeman FJ. The physiology of the internal urinary sphincter. J Urol. 1970;104(4):549-554.
15. Stahl SM. How psychiatrists can build new therapies for impotence. J Clin Psychiatry. 1998;59(2):47-48.
16. Bredt DS, Hwang PM, Glatt CE, et al. Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature. 1991;351(6329):714-718.
17. Corona G, Ricca V, Bandini E, et al. Selective serotonin reuptake inhibitor-induced sexual dysfunction. J Sex Med. 2009;6(5):1259-1269.
18. Pfaus JG. Pathways of sexual desire. J Sex Med. 2009;6(6):1506-1533.
19. Ahrold TK, Meston CM. Effects of SNS activation on SSRI-induced sexual side effects differ by SSRI. J Sex Marital Ther. 2009;35(4):311-319.
20. Hirshkowitz M, Schmidt MH. Sleep-related erections: clinical perspectives and neural mechanisms. Sleep Med Rev. 2005;9(4):311-329.
21. Miner MM, Seftel AD. Centrally acting mechanisms for the treatment of male sexual dysfunction. Urol Clin North Am. 2007;34(4):483-496, v.
22. Labbate LA, Grimes J, Hines A, et al. Sexual dysfunction induced by serotonin reuptake antidepressants. J Sex Marital Ther. 1998;24(1):3-12.
23. Waldinger MD. The neurobiological approach to premature ejaculation. J Urol. 2002;168(6):2359-2367.
24. Meston CM, Frohlich PF. The neurobiology of sexual function. Arch Gen Psychiatry. 2000;57(11):1012-1030.
25. Safarinejad MR. Evaluation of endocrine profile and hypothalamic-pituitary-testis axis in selective serotonin reuptake inhibitor-induced male sexual dysfunction. J Clin Psychopharmacol. 2008;28(4):418-423.
26. Sajith SG, Morgan C, Clarke D. Pharmacological management of inappropriate sexual behaviours: a review of its evidence, rationale and scope in relation to men with intellectual disabilities. J Intellect Disabil Res. 2008;52(12):1078-1090.
27. Giuliano F, Hellstrom WJ. The pharmacological treatment of premature ejaculation. BJU Int. 2008;102(6):668-675.
28. Kim SW, Paick JS. Short-term analysis of the effects of as needed use of sertraline at 5 PM for the treatment of premature ejaculation. Urology. 1999;54(3):544-547.
29. Waldinger MD, Zwinderman AH, Schweitzer DH, et al. Relevance of methodological design for the interpretation of efficacy of drug treatment of premature ejaculation: a systematic review and meta-analysis. Int J Impot Res. 2004;16(4):369-381.
30. Kendirci M, Salem E, Hellstrom WJ. Dapoxetine, a novel selective serotonin transport inhibitor for the treatment of premature ejaculation. Ther Clin Risk Manag. 2007;3(2):277-289.
1. Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848-856.
2. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
3. Sadock BJ, Sadock VA. Abnormal sexuality and sexual dysfunctions. In Sadock BJ, Sadock VA. Kaplan & Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry. 10th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:689-705.
4. Zimmerman M, Galione JN, Attiullah N, et al. Underrecognition of clinically significant side effects in depressed outpatients. J Clin Psychiatry. 2010;71(4):484-490.
5. Casper RC, Redmont DE, Jr, Katz MM, et al. Somatic symptoms in primary affective disorder: presence and relationship to the classification of depression. Arch Gen Psychiatry. 1985;42:1098-1104.
6. Balon R. SSRI-associated sexual dysfunction. Am J Psychiatry. 2006;163(9):1504-1509; quiz 1664.
7. Schatzberg AF, Nemeroff CB. eds. The American Psychiatric Publishing textbook of psychopharmacology. 4th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2009.
8. Schatzberg AF, Cole JO, DeBattista C. Antidepressants. In: Schatzberg AF, Cole JO, DeBattista C. Manual of clinical psychopharmacology. 6th ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2007:37-168.
9. Bancroft J, Janssen E. The dual control model of male sexual response: a theoretical approach to centrally mediated erectile dysfunction. Neurosci Biobehav Rev. 2000;24(5):571-579.
10. Segraves RT. Effects of psychotropic drugs on human erection and ejaculation. Arch Gen Psychiatry. 1989;46(3):275-284.
11. Tagliamonte A, Tagliamonte P, Gessa GL, et al. Compulsive sexual activity induced by p-chlorophenylalanine in normal and pinealectomized male rats. Science. 1969;166(911):1433-1435.
12. Rodriguez M, Castro R, Hernandez G, et al. Different roles of catecholaminergic and serotoninergic neurons of the medial forebrain bundle on male rat sexual behavior. Physiol Behav. 1984;33(1):5-11.
13. Monteiro WO, Noshirvani HF, Marks IM, et al. Anorgasmia from clomipramine in obsessive-compulsive disorder. A controlled trial. Br J Psychiatry. 1987;151:107-112.
14. Kleeman FJ. The physiology of the internal urinary sphincter. J Urol. 1970;104(4):549-554.
15. Stahl SM. How psychiatrists can build new therapies for impotence. J Clin Psychiatry. 1998;59(2):47-48.
16. Bredt DS, Hwang PM, Glatt CE, et al. Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature. 1991;351(6329):714-718.
17. Corona G, Ricca V, Bandini E, et al. Selective serotonin reuptake inhibitor-induced sexual dysfunction. J Sex Med. 2009;6(5):1259-1269.
18. Pfaus JG. Pathways of sexual desire. J Sex Med. 2009;6(6):1506-1533.
19. Ahrold TK, Meston CM. Effects of SNS activation on SSRI-induced sexual side effects differ by SSRI. J Sex Marital Ther. 2009;35(4):311-319.
20. Hirshkowitz M, Schmidt MH. Sleep-related erections: clinical perspectives and neural mechanisms. Sleep Med Rev. 2005;9(4):311-329.
21. Miner MM, Seftel AD. Centrally acting mechanisms for the treatment of male sexual dysfunction. Urol Clin North Am. 2007;34(4):483-496, v.
22. Labbate LA, Grimes J, Hines A, et al. Sexual dysfunction induced by serotonin reuptake antidepressants. J Sex Marital Ther. 1998;24(1):3-12.
23. Waldinger MD. The neurobiological approach to premature ejaculation. J Urol. 2002;168(6):2359-2367.
24. Meston CM, Frohlich PF. The neurobiology of sexual function. Arch Gen Psychiatry. 2000;57(11):1012-1030.
25. Safarinejad MR. Evaluation of endocrine profile and hypothalamic-pituitary-testis axis in selective serotonin reuptake inhibitor-induced male sexual dysfunction. J Clin Psychopharmacol. 2008;28(4):418-423.
26. Sajith SG, Morgan C, Clarke D. Pharmacological management of inappropriate sexual behaviours: a review of its evidence, rationale and scope in relation to men with intellectual disabilities. J Intellect Disabil Res. 2008;52(12):1078-1090.
27. Giuliano F, Hellstrom WJ. The pharmacological treatment of premature ejaculation. BJU Int. 2008;102(6):668-675.
28. Kim SW, Paick JS. Short-term analysis of the effects of as needed use of sertraline at 5 PM for the treatment of premature ejaculation. Urology. 1999;54(3):544-547.
29. Waldinger MD, Zwinderman AH, Schweitzer DH, et al. Relevance of methodological design for the interpretation of efficacy of drug treatment of premature ejaculation: a systematic review and meta-analysis. Int J Impot Res. 2004;16(4):369-381.
30. Kendirci M, Salem E, Hellstrom WJ. Dapoxetine, a novel selective serotonin transport inhibitor for the treatment of premature ejaculation. Ther Clin Risk Manag. 2007;3(2):277-289.
The re-emerging role of therapeutic neuromodulation
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments
The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:
- electroconvulsive therapy (ECT)
- vagus nerve stimulation (VNS)
- transcranial magnetic stimulation (TMS)
- deep brain stimulation (DBS).
It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.
Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.
Table 1
Therapeutic neuromodulation: Categorization based on risk
Noninvasive, nonseizurogenic TMS, tDCS, CES | ||
Noninvasive, seizurogenic ECT, MST, FEAST | ||
Invasive, nonseizurogenic VNS, DBS, EpCS | ||
CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
Table 2
Approved and investigational indications of neuromodulation
Approach | Description | Clinical application |
---|---|---|
CES | Uses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the face | Depression Anxiety Sleep disorders |
DBS | ‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNS | Depression OCD* Parkinson’s disease* Dystonia* |
ECT | Short-term electrical stimulation sufficient to induce a seizure | Depression* Schizophrenia Mania |
EpCS | Uses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortex | Depression Pain |
FEAST | An alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrode | Depression |
MST | Intense, high-frequency magnetic pulses sufficient to induce a seizure | Depression |
tDCS | Sustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalp | Depression |
TMS | Use of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibition | Depression* PTSD OCD Schizophrenia Substance use disorders Tinnitus |
VNS | Use of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleus | Epilepsy* Depression* |
*FDA-approved indications CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
ECT: Oldest and most effective
ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.
ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:
- access and expertise are limited in many areas
- cognition is at least temporarily adversely affected
- relapse rates after acute benefit are high
- cost
- public perception often is negative.
Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).
Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7
Stimulating the vagus nerve
VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.
The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.
With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.
Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.
In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.
A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.
Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.
Table 3
Vagus nerve stimulation treatment parameters
Parameter | Units | Range | Median value at 12 months in pivotal study |
---|---|---|---|
Output current | Milliamps (mA) | 0 to 3.5 | 1 |
Signal frequency | Hertz (Hz) | 1.30 | 20 |
Pulse width | Microseconds (µsec) | 130 to 1,000 | 500 |
Duty cycle: ON time* | Seconds | 7 to 60 | 30 |
Duty cycle: OFF time* | Minutes | 0.2 to 180 | 5 |
*Stimulation cycle is 24 hours per day Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005 |
A newer option: TMS
TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.
TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.
TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14
O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.
Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:
- using a novel somatosensory system that produced similar sensations with sham and active TMS
- assessing the success of maintaining the blind
- establishing a rigorous clinical rating system
- utilizing MRI-guided adjustment of coil placement in a subset of patients.
The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).
Table 4
Treatment parameters of transcranial magnetic stimulation
Parameter | Comment |
---|---|
Motor threshold | Lowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored |
Stimulus coil location | Most common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex |
Stimulus pulse(s) or train | |
Intensity | 80% to 120% of MT |
Frequency | ≤1 to 20 Hz |
Duration | ≤1 millisecond |
Interpulse interval | 50 to 100 milliseconds |
Stimulus train duration | 3 to 6 seconds |
Inter-train interval | 20 to 60 seconds |
Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63 |
Deep brain stimulation
DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).
Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.
Adverse effects of DBS can be:
- surgical-related (eg, seizure, bleeding, infection)
- device-related (eg, lead breakage, malfunction)
- stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).
The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.
Cost and reimbursement
Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.
Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34
Related Resource
- Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.
Drug Brand Names
- Lithium • Eskalith, Lithobid
- Nortriptyline • Aventyl, Pamelor
1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.
2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.
3. Rasmussen KG, Mueller M, Rummans TA, et al. Is baseline medication resistance associated with potential for relapse after successful remission of a depressive episode with ECT? Data from the Consortium for Research on Electroconvulsive Therapy (CORE). J Clin Psychiatry. 2009;70(2):232-237.
4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.
5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.
6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.
7. Sackeim HA, Prudic J, Nobler MS, et al. Effects of pulse width and electrode placement on the efficacy and cognitive effects of electroconvulsive therapy. Brain Stimulat. 2008;1:71-83.
8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.
9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.
10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.
11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.
12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.
13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.
14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.
15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.
16. Lisanby SH, Husain MM, Rosenquist PB, et al. Daily left prefrontal repetitive transcranial magnetic stimulation in the acute treatment of major depression: clinical predictors of outcome in a multisite, randomized controlled clinical trial. Neuropsychopharmacology. 2009;34(2):522-534.
17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.
19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.
20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.
21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.
22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.
23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.
24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.
25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.
26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.
27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.
28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.
29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.
30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.
31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.
32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.
33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.
34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments
The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:
- electroconvulsive therapy (ECT)
- vagus nerve stimulation (VNS)
- transcranial magnetic stimulation (TMS)
- deep brain stimulation (DBS).
It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.
Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.
Table 1
Therapeutic neuromodulation: Categorization based on risk
Noninvasive, nonseizurogenic TMS, tDCS, CES | ||
Noninvasive, seizurogenic ECT, MST, FEAST | ||
Invasive, nonseizurogenic VNS, DBS, EpCS | ||
CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
Table 2
Approved and investigational indications of neuromodulation
Approach | Description | Clinical application |
---|---|---|
CES | Uses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the face | Depression Anxiety Sleep disorders |
DBS | ‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNS | Depression OCD* Parkinson’s disease* Dystonia* |
ECT | Short-term electrical stimulation sufficient to induce a seizure | Depression* Schizophrenia Mania |
EpCS | Uses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortex | Depression Pain |
FEAST | An alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrode | Depression |
MST | Intense, high-frequency magnetic pulses sufficient to induce a seizure | Depression |
tDCS | Sustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalp | Depression |
TMS | Use of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibition | Depression* PTSD OCD Schizophrenia Substance use disorders Tinnitus |
VNS | Use of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleus | Epilepsy* Depression* |
*FDA-approved indications CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
ECT: Oldest and most effective
ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.
ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:
- access and expertise are limited in many areas
- cognition is at least temporarily adversely affected
- relapse rates after acute benefit are high
- cost
- public perception often is negative.
Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).
Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7
Stimulating the vagus nerve
VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.
The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.
With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.
Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.
In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.
A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.
Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.
Table 3
Vagus nerve stimulation treatment parameters
Parameter | Units | Range | Median value at 12 months in pivotal study |
---|---|---|---|
Output current | Milliamps (mA) | 0 to 3.5 | 1 |
Signal frequency | Hertz (Hz) | 1.30 | 20 |
Pulse width | Microseconds (µsec) | 130 to 1,000 | 500 |
Duty cycle: ON time* | Seconds | 7 to 60 | 30 |
Duty cycle: OFF time* | Minutes | 0.2 to 180 | 5 |
*Stimulation cycle is 24 hours per day Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005 |
A newer option: TMS
TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.
TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.
TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14
O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.
Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:
- using a novel somatosensory system that produced similar sensations with sham and active TMS
- assessing the success of maintaining the blind
- establishing a rigorous clinical rating system
- utilizing MRI-guided adjustment of coil placement in a subset of patients.
The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).
Table 4
Treatment parameters of transcranial magnetic stimulation
Parameter | Comment |
---|---|
Motor threshold | Lowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored |
Stimulus coil location | Most common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex |
Stimulus pulse(s) or train | |
Intensity | 80% to 120% of MT |
Frequency | ≤1 to 20 Hz |
Duration | ≤1 millisecond |
Interpulse interval | 50 to 100 milliseconds |
Stimulus train duration | 3 to 6 seconds |
Inter-train interval | 20 to 60 seconds |
Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63 |
Deep brain stimulation
DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).
Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.
Adverse effects of DBS can be:
- surgical-related (eg, seizure, bleeding, infection)
- device-related (eg, lead breakage, malfunction)
- stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).
The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.
Cost and reimbursement
Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.
Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34
Related Resource
- Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.
Drug Brand Names
- Lithium • Eskalith, Lithobid
- Nortriptyline • Aventyl, Pamelor
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments
The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:
- electroconvulsive therapy (ECT)
- vagus nerve stimulation (VNS)
- transcranial magnetic stimulation (TMS)
- deep brain stimulation (DBS).
It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.
Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.
Table 1
Therapeutic neuromodulation: Categorization based on risk
Noninvasive, nonseizurogenic TMS, tDCS, CES | ||
Noninvasive, seizurogenic ECT, MST, FEAST | ||
Invasive, nonseizurogenic VNS, DBS, EpCS | ||
CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
Table 2
Approved and investigational indications of neuromodulation
Approach | Description | Clinical application |
---|---|---|
CES | Uses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the face | Depression Anxiety Sleep disorders |
DBS | ‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNS | Depression OCD* Parkinson’s disease* Dystonia* |
ECT | Short-term electrical stimulation sufficient to induce a seizure | Depression* Schizophrenia Mania |
EpCS | Uses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortex | Depression Pain |
FEAST | An alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrode | Depression |
MST | Intense, high-frequency magnetic pulses sufficient to induce a seizure | Depression |
tDCS | Sustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalp | Depression |
TMS | Use of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibition | Depression* PTSD OCD Schizophrenia Substance use disorders Tinnitus |
VNS | Use of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleus | Epilepsy* Depression* |
*FDA-approved indications CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation |
ECT: Oldest and most effective
ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.
ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:
- access and expertise are limited in many areas
- cognition is at least temporarily adversely affected
- relapse rates after acute benefit are high
- cost
- public perception often is negative.
Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).
Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7
Stimulating the vagus nerve
VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.
The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.
With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.
Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.
In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.
A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.
Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.
Table 3
Vagus nerve stimulation treatment parameters
Parameter | Units | Range | Median value at 12 months in pivotal study |
---|---|---|---|
Output current | Milliamps (mA) | 0 to 3.5 | 1 |
Signal frequency | Hertz (Hz) | 1.30 | 20 |
Pulse width | Microseconds (µsec) | 130 to 1,000 | 500 |
Duty cycle: ON time* | Seconds | 7 to 60 | 30 |
Duty cycle: OFF time* | Minutes | 0.2 to 180 | 5 |
*Stimulation cycle is 24 hours per day Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005 |
A newer option: TMS
TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.
TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.
TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14
O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.
Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:
- using a novel somatosensory system that produced similar sensations with sham and active TMS
- assessing the success of maintaining the blind
- establishing a rigorous clinical rating system
- utilizing MRI-guided adjustment of coil placement in a subset of patients.
The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).
Table 4
Treatment parameters of transcranial magnetic stimulation
Parameter | Comment |
---|---|
Motor threshold | Lowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored |
Stimulus coil location | Most common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex |
Stimulus pulse(s) or train | |
Intensity | 80% to 120% of MT |
Frequency | ≤1 to 20 Hz |
Duration | ≤1 millisecond |
Interpulse interval | 50 to 100 milliseconds |
Stimulus train duration | 3 to 6 seconds |
Inter-train interval | 20 to 60 seconds |
Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63 |
Deep brain stimulation
DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).
Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.
Adverse effects of DBS can be:
- surgical-related (eg, seizure, bleeding, infection)
- device-related (eg, lead breakage, malfunction)
- stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).
The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.
Cost and reimbursement
Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.
Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34
Related Resource
- Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.
Drug Brand Names
- Lithium • Eskalith, Lithobid
- Nortriptyline • Aventyl, Pamelor
1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.
2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.
3. Rasmussen KG, Mueller M, Rummans TA, et al. Is baseline medication resistance associated with potential for relapse after successful remission of a depressive episode with ECT? Data from the Consortium for Research on Electroconvulsive Therapy (CORE). J Clin Psychiatry. 2009;70(2):232-237.
4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.
5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.
6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.
7. Sackeim HA, Prudic J, Nobler MS, et al. Effects of pulse width and electrode placement on the efficacy and cognitive effects of electroconvulsive therapy. Brain Stimulat. 2008;1:71-83.
8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.
9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.
10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.
11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.
12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.
13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.
14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.
15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.
16. Lisanby SH, Husain MM, Rosenquist PB, et al. Daily left prefrontal repetitive transcranial magnetic stimulation in the acute treatment of major depression: clinical predictors of outcome in a multisite, randomized controlled clinical trial. Neuropsychopharmacology. 2009;34(2):522-534.
17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.
19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.
20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.
21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.
22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.
23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.
24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.
25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.
26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.
27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.
28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.
29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.
30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.
31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.
32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.
33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.
34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.
1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.
2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.
3. Rasmussen KG, Mueller M, Rummans TA, et al. Is baseline medication resistance associated with potential for relapse after successful remission of a depressive episode with ECT? Data from the Consortium for Research on Electroconvulsive Therapy (CORE). J Clin Psychiatry. 2009;70(2):232-237.
4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.
5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.
6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.
7. Sackeim HA, Prudic J, Nobler MS, et al. Effects of pulse width and electrode placement on the efficacy and cognitive effects of electroconvulsive therapy. Brain Stimulat. 2008;1:71-83.
8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.
9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.
10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.
11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.
12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.
13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.
14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.
15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.
16. Lisanby SH, Husain MM, Rosenquist PB, et al. Daily left prefrontal repetitive transcranial magnetic stimulation in the acute treatment of major depression: clinical predictors of outcome in a multisite, randomized controlled clinical trial. Neuropsychopharmacology. 2009;34(2):522-534.
17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.
19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.
20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.
21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.
22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.
23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.
24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.
25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.
26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.
27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.
28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.
29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.
30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.
31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.
32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.
33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.
34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.
Tailoring depression treatment for women with breast cancer
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/depression-treatment-for-women-with.html#comments
Psychological distress among patients with breast cancer is common and is linked to worse clinical outcomes. Depressive and anxiety symptoms affect up to 40% of breast cancer patients,1 and depression is associated with a higher relative risk of mortality in individuals with breast cancer.2 Psychotropic medications and psychotherapy used to treat depression in patients without carcinoma also are appropriate and effective for breast cancer patients. However, some patients present distinct challenges to standard treatment. For example, growing evidence suggests that some selective serotonin reuptake inhibitors (SSRIs) may reduce the effectiveness of tamoxifen, a chemotherapeutic agent. This article discusses challenges in diagnosing and treating depression in breast cancer patients and reviews evidence supporting appropriate psychiatric care.
Increased vulnerability
In 10% to 30% of women, a breast cancer diagnosis may lead to increased vulnerability to depressive disorders, including adjustment disorders with depressed mood, major depressive disorder (MDD), and mood disorders related to general medical conditions.3,4 The risk of developing a depressive disorder is highest in the year after receiving the breast cancer diagnosis.4
A woman’s risk of developing a depressive disorder may depend on the type of cancer treatment she receives. For example, breast asymmetry is common after breast conserving surgery. Waljee et al5 found that women with breast asymmetry had increased fears of cancer recurrence and more feelings of self-consciousness. More pronounced asymmetry led to a higher incidence of depressive symptoms. However, among 90 patients undergoing bilateral prophylactic mastectomy, the rate of depression had not changed 1 year after the procedure.6 Chemotherapy, particularly at high doses, is a risk factor for depression.4,7,8
Self-blame for developing breast cancer can affect mood. In 2007, Friedman et al9 determined that higher levels of self-blame correlated with higher levels of depression and decreased quality of life. Women often blamed themselves for various reasons, including:
- poor coping skills
- anxiety about their health and treatments
- inability to express emotions
- delays in medical consultation.
Exacerbated symptoms and side effects. Women with depression often experience increased side effects from cancer treatments, and the subjective experience of these effects—including hot flashes, cognitive impairment, pain, and sexual dysfunction—likely is intensified.4 Somatic symptoms of depression may be exacerbated by cancer treatment side effects or mistaken for effects of the treatment. When somatic symptoms of depression are mistaken for treatment side effects, depression—and the opportunity to treat it—can be overlooked.10
Depression may be a risk factor for poor adherence to cancer treatment. In a quantitative review of studies correlating depression and medical treatment noncompliance, DiMatteo et al11 determined that compared with nondepressed patients, those with depression were 3 times more likely to not adhere to treatment recommendations; this review was not limited to cancer patients. Depressive symptoms—notably poor concentration and amotivation—can create the impression that a patient is poorly adherent. Women with comorbid depression and breast cancer may have difficulty understanding treatment recommendations or remembering daily treatment goals.4
Appropriate screening tools
Factors that may increase a breast cancer patient’s risk for developing a psychiatric disorder are listed in Table 1.10 Many depression screening tools are available; below we describe 3 commonly used for patients with breast cancer.
The National Comprehensive Cancer Center Distress Thermometer allows patients to rate their overall distress level over the past week on a scale from 0 to 10, using a visual analogue.12 The Distress Thermometer has been validated for several cancer populations and in different parts of the world. A score of 7 has both good sensitivity and specificity for detecting depression in breast cancer patients. Consider a complete psychiatric evaluation for patients with scores ≥7.13
The Profile of Mood States questionnaire14 is a reliable, valid 65-item questionnaire often used in studies of mood dysregulation and breast cancer. Subscales include depression-dejection, tension-anxiety, anger-hostility, confusion-bewilderment, vigor-activity, and fatigue-inertia. Using a 5-point Likert scale, patients rate their symptoms over the past week. Subscale scores are then added to a total mood disturbance score.14,15
The Hospital Anxiety and Depression Scale (HADS) is a sensitive, reliable 14-item scale that is commonly used to study depression and anxiety in patients with breast cancer.16 HADS includes two 7-item subscales—anxiety and depression—and answers are scored on a 4-point Likert scale. Patients are asked to respond quickly and avoid thinking too long about their answers.
Table 1
Risk factors for psychiatric distress related to breast cancer
Past psychiatric illness |
Family history of psychiatric illness |
Younger age (<45 years) |
Having young children |
Limited social support |
Substance use |
Single status |
Pain |
Physical disability |
Poor family coherence |
Financial strain |
Source: Adapted from reference 10 |
Psychotherapeutic options
Behavioral therapies can diminish symptoms of depression, according to a review of studies and practice guidelines on managing depression in cancer patients.17 Group interventions, in particular, can be valuable. Anderson et al18 found that group cohesion, member connectedness, and more sessions correlated with decreased psychological distress.
Psychoeducation aims to provide medical information and discuss cancer’s causes, prognosis, and treatment strategies. Group settings can help improve communication and problem-solving skills. In a randomized controlled trial (RCT) of 203 women with breast cancer, psychoeducational group treatment reduced depression, anger, and fatigue.19
Cognitive-behavioral therapy (CBT) helps patients identify and restructure negative thoughts and increase positive, adaptive behaviors. Hunter et al20 noted significant improvement in depressed mood, anxiety, and sleep in 17 women experiencing menopausal symptoms who received group CBT after completing breast cancer treatment. In 1 study, 124 patients with metastatic breast cancer who received 8 weekly sessions of group CBT reported reduced depression and mood disturbance and improved self-esteem compared with a no-therapy control group.21
Supportive-expressive therapy (SET) is a manual-based therapy that focuses on increasing social support, improving symptom control, and enhancing communication between the patient and treatment team. Affective expression helps lead the therapist to issues that should be addressed. Evidence on the effectiveness of SET for patients with breast cancer is mixed. A study of 357 women with breast cancer who were randomly assigned to 12-week group SET or an educational control condition found no evidence that SET reduced distress.22 However, a trial of 485 women with advanced breast cancer who were randomly assigned to group SET plus relaxation therapy or relaxation therapy alone showed that SET improved quality of life, including protection against depression.23
Mindfulness-based stress reduction (MBSR) is a standardized form of meditation and yoga. Clinicians teach patients visualization, breathing exercises, and meditation to help them become aware of the body’s reaction to stress and how to regulate it. In an RCT of 84 female breast cancer survivors, a 6-week MBSR program diminished depressive symptoms, improved physical functioning, and reduced fear of cancer recurrence.24
Evidence for antidepressants
SSRIs. Expert consensus guidelines on treating depression in women recommend an SSRI as a first-line agent.25 In RCTs, fluoxetine, paroxetine, and sertraline were more effective than placebo in treating depression and related symptoms specifically in women with breast cancer (Table 2).26-28
The interaction between SSRIs and chemotherapy agents is a concern. Tamoxifen decreases the rate of death from breast cancer in hormone receptor positive breast cancers.29 Endoxifen, a potent anti-estrogen, is an active metabolite of tamoxifen via cytochrome P450 (CYP) 2D6. Goetz et al30 demonstrated that women with decreased or inhibited metabolism via CYP2D6 had significantly shorter times to breast cancer recurrence, compared with women with extensive CYP2D6 metabolism.
SSRIs can varyingly inhibit CYP2D6. In a prospective trial of 158 breast cancer patients receiving tamoxifen, paroxetine and fluoxetine were found to be strong inhibitors of CYP2D6 and led to low levels of endoxifen.31 In contrast, weaker inhibitors, including sertraline and citalopram, led to intermediate levels of endoxifen. In a retrospective cohort study, Kelly et al32 demonstrated that women treated with paroxetine, in combination with tamoxifen, had an increased risk of death compared with women treated with other SSRIs or venlafaxine and tamoxifen. They estimated that paroxetine use in women treated with tamoxifen would lead to 1 additional breast cancer death per 20 women within 5 years of discontinuing tamoxifen.
According to American Psychiatric Association practice guidelines for treatment of MDD, depressed breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on CYP2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine.33
Serotonin-norepinephrine reuptake inhibitors (SNRIs) may be used to treat depressive disorders. In addition, venlafaxine may be helpful in treating post-mastectomy pain syndrome. Approximately one-half of patients who undergo mastectomy or breast reconstruction may experience a postoperative pain syndrome.34 The most common symptom is a burning, stabbing pain in the axilla, arm, and chest wall of the affected side. This pain is worsened by movement and is poorly responsive to opioids.35
In a 10-week RCT of 13 patients with neuropathic pain after breast cancer treatment, venlafaxine significantly improved pain relief compared with placebo, although the drug did not affect depression or anxiety.36 In a study of 100 patients given venlafaxine or placebo for 2 weeks starting the night before undergoing partial or radical mastectomy with axillary dissection, those receiving venlafaxine had a significantly lower incidence of pain in the chest wall, arm, and axillary region, and scores of pain with movement were decreased.37 There was no difference in opioid usage between groups.
Tricyclic antidepressants have been demonstrated to be effective in breast cancer patients. Side effects—notably anticholinergic effects—limit their use as antidepressants, especially when compared with SSRI treatment. In a study that randomly assigned 179 women with breast cancer to paroxetine, 20 to 40 mg/d, or amitriptyline, 75 to 150 mg/d, anticholinergic effects were almost twice as frequent in the amitriptyline group (19%) compared with paroxetine (11%).38 In a 4-week double-blind, placebo-controlled crossover trial of 15 breast cancer patients, amitriptyline significantly relieved neuropathic pain, but its adverse effects made most patients unwilling to use the medication regularly.39
Table 2
Evidence supporting SSRI use in patients with breast cancer*
Study | Design | Results |
---|---|---|
Navari et al, 200826 | 193 patients with newly diagnosed early-stage breast cancer were randomized to fluoxetine, 20 mg/d, or placebo for 6 months | Fluoxetine reduced depressive symptoms, improved quality of life, and led to higher completion of adjuvant chemotherapy and/or hormone therapy |
Roscoe et al, 200527 | 94 women with breast cancer receiving at least 4 cycles of chemotherapy were randomized to paroxetine, 20 mg/d, or placebo | Paroxetine significantly reduced depression during chemotherapy |
Kimick et al, 200628 | 62 women with early-stage breast cancer receiving the chemotherapy agent tamoxifen who reported hot flashes were randomized to sertraline, 50 mg/d, or placebo for 6 weeks | Sertraline was significantly more effective than placebo at reducing hot flashes |
* Breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on cytochrome P450 2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine SSRIs: selective serotonin reuptake inhibitors |
Related Resources
- American Cancer Society. www.cancer.org.
- National Comprehensive Cancer network. www.nccn.org.
Drug Brand Names
- Amitripyline • Elavil
- Citalopram • Celexa
- Desvenlafaxine • Pristiq
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Paroxetine • Paxil
- Sertraline • Zoloft
- Tamoxifen • Nolvadex
- Venlafaxine • Effexor
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Von Ah D, Kang DH. Correlates of mood disturbance in women with breast cancer: patterns over time. J Adv Nurs. 2008;61(6):676-689.
2. Hjerl K, Andersen EW, Keiding N, et al. Depression as a prognostic factor for breast cancer mortality. Psychosomatics. 2003;44:24-30.
3. Stanton AL. Psychosocial concerns and interventions for cancer survivors. J Clin Oncol. 2006;24(32):5132-5137.
4. Fann JR, Thomas-Rich AM, Katon WJ, et al. Major depression after breast cancer: a review of epidemiology and treatment. Gen Hosp Psychiatry. 2008;30:112-126.
5. Waljee JF, Hu ES, Ubel PA, et al. Effect of esthetic outcome after breast-conserving surgery on psychosocial functioning and quality of life. J Clin Oncol. 2008;26(20):3331-3337.
6. Brandberg Y, Sandelin K, Erikson S, et al. Psychological reactions, quality of life, and body image after bilateral prophylactic mastectomy in women at high risk for breast cancer: a prospective 1-year follow-up study. J Clin Oncol. 2008;26(24):3943-3949.
7. Lee KC, Ray T, Hunkeler E, et al. Tamoxifen treatment and new onset depression in breast cancer patients. Psychosomatics. 2007;48:205-210.
8. Thornton LM, Carson WE, Shapiro CL, et al. Delayed emotional recovery after taxane-based chemotherapy. Cancer. 2008;113(3):638-647.
9. Friedman LC, Romero C, Elledge R, et al. Attribution of blame, self-forgiving attitude and psychological adjustment in women with breast cancer. J Behav Med. 2007;30:351-357.
10. Spiegel D, Riba M. Psychological aspects of cancer. In: DeVita VT, Lawrence TS, Rosenberg SA, eds. Principles and practice of oncology. 8th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2008:2817–2826.
11. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment. Arch Intern Med. 2000;160:2101-2107.
12. National Comprehensive Cancer Network. Practice guidelines in oncology—distress management v.1.2010. 2010. Available at: http://www.nccn.org. Accessed October 5, 2010.
13. Hegel MT, Collins ED, Kearing S. Sensitivity and specificity of the distress thermometer for depression in newly diagnosed breast cancer patients. Psychooncology. 2008;17:556-560.
14. Lorr M, McNair DM, Droppleman LF. POMS profile of mood states. Available at: http://www.mhs.com/product.aspx?gr=cli&prod=poms&id=overview. Accessed September 29, 2010.
15. Classen C, Butler LD, Koopman C. Supportive-expressive group therapy and distress in patients with metastatic breast cancer: a randomized clinical intervention trial. Arch Gen Psychiatry. 2001;58:494-501.
16. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67:361-370.
17. Barsevick AM, Sweeney C, Haney E, et al. A systematic qualitative analysis of psychoeducational interventions for depression in patients with cancer. Oncol Nurs Forum. 2002;29(1):73-84.
18. Andersen BL, Shelby RA, Golden-Kreutz DM. RCT of a psychological intervention for patients with cancer: I. Mechanisms of change. J Consult Clin Psychol. 2007;75(6):927-938.
19. Dolbreault S, Cayrou S, Bredart A, et al. The effectiveness of a psycho-educational group after early-stage breast cancer treatment: results of a randomized French study. Psychooncology. 2009;18:647-656.
20. Hunter MS, Coventry S, Hamed H, et al. Evaluation of a group cognitive behavioural intervention for women suffering from menopausal symptoms following breast cancer treatment. Psychooncology. 2009;18(5):560-563.
21. Edelman S, Bell DR, Kidman AD. A group cognitive behaviour therapy programme with metastatic breast cancer patients. Psychooncology. 1999;8(4):295-305.
22. Classen CC, Kraemer HC, Blasey C, et al. Supportive-expressive group therapy for primary breast cancer patients: a randomized prospective multicenter trial. Psychooncology. 2008;17:438-447.
23. Kissane DW, Grabsch B, Clarke DM, et al. Supportive-expressive group therapy for women with metastatic breast cancer: survival and psychosocial outcome from a randomized control trial. Psychooncology. 2007;16(4):277-286.
24. Lengacher CA, Johnson-Mallard V, Post-White J, et al. Randomized controlled trial of mindfulness-based stress reduction (MBSR) for survivors of breast cancer. Psychooncology. 2009;18:1261-1272.
25. Altshuler LL, Cohen LS, Moline ML, et al. Treatment of depression in women: a summary of expert consensus guidelines. J Psychiatr Pract. 2001;7(3):185-208.
26. Navari RM, Brenner MC, Wilson MN. Treatment of depressive symptoms in patients with early stage breast cancer undergoing adjuvant therapy. Breast Cancer Res Treat. 2008;112(1):197-201.
27. Roscoe JA, Morrow GR, Hickok JT, et al. Effect of paroxetine hydrochloride on fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res Treat. 2005;89(3):243-249.
28. Kimmick GG, Lovato J, McQuellon R, et al. Randomized, double-blind, placebo-controlled crossover study of sertraline (Zoloft) for treatment of hot flashes in women with early stage breast cancer taking tamoxifen. Breast J. 2006;12(2):114-122.
29. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomized trials. Lancet. 2005;365(9472):1687-1717.
30. Goetz MP, Knox SK, Suman VJ, et al. The impact of cytochrome P450 2D6 metabolism in women receiving adjuvant tamoxifen. Breast Cancer Res Treat. 2007;101(1):113-121.
31. Borges S, Desta Z, Li L, et al. Quantitative effect of CYP2D6 genotype and inhibitors on tamoxifen metabolism: implication for optimization of breast cancer treatment. Clin Pharmacol Ther. 2006;80(1):61-74.
32. Kelly CM, Juurlink DN, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population based cohort study. BMJ. 2010;8:340;c693.-
33. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. 3rd ed. Arlington, VA: American Psychiatric Publishing, Inc. 2010.
34. Vadivelu N, Schreck M, Lopez J, et al. Pain after mastectomy and breast reconstruction. Am Surg. 2008;74(4):285-296.
35. Stevens PE, Dibble SL, Miaskowski C. Prevalence, characteristics and impact of post-mastectomy pain syndrome: an investigation of women’s experiences. Pain. 1995;61:61-68.
36. Tasmuth T, Hartel B, Kalso ET. Venlafaxine in neuropathic pain following treatment of breast cancer. Eur J Pain. 2002;6:17-24.
37. Reuben SS, Makari-Judson G, Lurie SD. Evaluation of efficacy of the perioperative administration of venlafaxine XR in the prevention of post-mastectomy pain syndrome. J Pain Sympt Manage. 2004;27(2):133-139.
38. 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. 2001;70(1):1-10.
39. Kalso ET, Tasmuth T, Neuvonen PJ. Amitriptyline effectively relieves neuropathic pain following treatment of breast cancer. Pain. 1996;64:293-302.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/depression-treatment-for-women-with.html#comments
Psychological distress among patients with breast cancer is common and is linked to worse clinical outcomes. Depressive and anxiety symptoms affect up to 40% of breast cancer patients,1 and depression is associated with a higher relative risk of mortality in individuals with breast cancer.2 Psychotropic medications and psychotherapy used to treat depression in patients without carcinoma also are appropriate and effective for breast cancer patients. However, some patients present distinct challenges to standard treatment. For example, growing evidence suggests that some selective serotonin reuptake inhibitors (SSRIs) may reduce the effectiveness of tamoxifen, a chemotherapeutic agent. This article discusses challenges in diagnosing and treating depression in breast cancer patients and reviews evidence supporting appropriate psychiatric care.
Increased vulnerability
In 10% to 30% of women, a breast cancer diagnosis may lead to increased vulnerability to depressive disorders, including adjustment disorders with depressed mood, major depressive disorder (MDD), and mood disorders related to general medical conditions.3,4 The risk of developing a depressive disorder is highest in the year after receiving the breast cancer diagnosis.4
A woman’s risk of developing a depressive disorder may depend on the type of cancer treatment she receives. For example, breast asymmetry is common after breast conserving surgery. Waljee et al5 found that women with breast asymmetry had increased fears of cancer recurrence and more feelings of self-consciousness. More pronounced asymmetry led to a higher incidence of depressive symptoms. However, among 90 patients undergoing bilateral prophylactic mastectomy, the rate of depression had not changed 1 year after the procedure.6 Chemotherapy, particularly at high doses, is a risk factor for depression.4,7,8
Self-blame for developing breast cancer can affect mood. In 2007, Friedman et al9 determined that higher levels of self-blame correlated with higher levels of depression and decreased quality of life. Women often blamed themselves for various reasons, including:
- poor coping skills
- anxiety about their health and treatments
- inability to express emotions
- delays in medical consultation.
Exacerbated symptoms and side effects. Women with depression often experience increased side effects from cancer treatments, and the subjective experience of these effects—including hot flashes, cognitive impairment, pain, and sexual dysfunction—likely is intensified.4 Somatic symptoms of depression may be exacerbated by cancer treatment side effects or mistaken for effects of the treatment. When somatic symptoms of depression are mistaken for treatment side effects, depression—and the opportunity to treat it—can be overlooked.10
Depression may be a risk factor for poor adherence to cancer treatment. In a quantitative review of studies correlating depression and medical treatment noncompliance, DiMatteo et al11 determined that compared with nondepressed patients, those with depression were 3 times more likely to not adhere to treatment recommendations; this review was not limited to cancer patients. Depressive symptoms—notably poor concentration and amotivation—can create the impression that a patient is poorly adherent. Women with comorbid depression and breast cancer may have difficulty understanding treatment recommendations or remembering daily treatment goals.4
Appropriate screening tools
Factors that may increase a breast cancer patient’s risk for developing a psychiatric disorder are listed in Table 1.10 Many depression screening tools are available; below we describe 3 commonly used for patients with breast cancer.
The National Comprehensive Cancer Center Distress Thermometer allows patients to rate their overall distress level over the past week on a scale from 0 to 10, using a visual analogue.12 The Distress Thermometer has been validated for several cancer populations and in different parts of the world. A score of 7 has both good sensitivity and specificity for detecting depression in breast cancer patients. Consider a complete psychiatric evaluation for patients with scores ≥7.13
The Profile of Mood States questionnaire14 is a reliable, valid 65-item questionnaire often used in studies of mood dysregulation and breast cancer. Subscales include depression-dejection, tension-anxiety, anger-hostility, confusion-bewilderment, vigor-activity, and fatigue-inertia. Using a 5-point Likert scale, patients rate their symptoms over the past week. Subscale scores are then added to a total mood disturbance score.14,15
The Hospital Anxiety and Depression Scale (HADS) is a sensitive, reliable 14-item scale that is commonly used to study depression and anxiety in patients with breast cancer.16 HADS includes two 7-item subscales—anxiety and depression—and answers are scored on a 4-point Likert scale. Patients are asked to respond quickly and avoid thinking too long about their answers.
Table 1
Risk factors for psychiatric distress related to breast cancer
Past psychiatric illness |
Family history of psychiatric illness |
Younger age (<45 years) |
Having young children |
Limited social support |
Substance use |
Single status |
Pain |
Physical disability |
Poor family coherence |
Financial strain |
Source: Adapted from reference 10 |
Psychotherapeutic options
Behavioral therapies can diminish symptoms of depression, according to a review of studies and practice guidelines on managing depression in cancer patients.17 Group interventions, in particular, can be valuable. Anderson et al18 found that group cohesion, member connectedness, and more sessions correlated with decreased psychological distress.
Psychoeducation aims to provide medical information and discuss cancer’s causes, prognosis, and treatment strategies. Group settings can help improve communication and problem-solving skills. In a randomized controlled trial (RCT) of 203 women with breast cancer, psychoeducational group treatment reduced depression, anger, and fatigue.19
Cognitive-behavioral therapy (CBT) helps patients identify and restructure negative thoughts and increase positive, adaptive behaviors. Hunter et al20 noted significant improvement in depressed mood, anxiety, and sleep in 17 women experiencing menopausal symptoms who received group CBT after completing breast cancer treatment. In 1 study, 124 patients with metastatic breast cancer who received 8 weekly sessions of group CBT reported reduced depression and mood disturbance and improved self-esteem compared with a no-therapy control group.21
Supportive-expressive therapy (SET) is a manual-based therapy that focuses on increasing social support, improving symptom control, and enhancing communication between the patient and treatment team. Affective expression helps lead the therapist to issues that should be addressed. Evidence on the effectiveness of SET for patients with breast cancer is mixed. A study of 357 women with breast cancer who were randomly assigned to 12-week group SET or an educational control condition found no evidence that SET reduced distress.22 However, a trial of 485 women with advanced breast cancer who were randomly assigned to group SET plus relaxation therapy or relaxation therapy alone showed that SET improved quality of life, including protection against depression.23
Mindfulness-based stress reduction (MBSR) is a standardized form of meditation and yoga. Clinicians teach patients visualization, breathing exercises, and meditation to help them become aware of the body’s reaction to stress and how to regulate it. In an RCT of 84 female breast cancer survivors, a 6-week MBSR program diminished depressive symptoms, improved physical functioning, and reduced fear of cancer recurrence.24
Evidence for antidepressants
SSRIs. Expert consensus guidelines on treating depression in women recommend an SSRI as a first-line agent.25 In RCTs, fluoxetine, paroxetine, and sertraline were more effective than placebo in treating depression and related symptoms specifically in women with breast cancer (Table 2).26-28
The interaction between SSRIs and chemotherapy agents is a concern. Tamoxifen decreases the rate of death from breast cancer in hormone receptor positive breast cancers.29 Endoxifen, a potent anti-estrogen, is an active metabolite of tamoxifen via cytochrome P450 (CYP) 2D6. Goetz et al30 demonstrated that women with decreased or inhibited metabolism via CYP2D6 had significantly shorter times to breast cancer recurrence, compared with women with extensive CYP2D6 metabolism.
SSRIs can varyingly inhibit CYP2D6. In a prospective trial of 158 breast cancer patients receiving tamoxifen, paroxetine and fluoxetine were found to be strong inhibitors of CYP2D6 and led to low levels of endoxifen.31 In contrast, weaker inhibitors, including sertraline and citalopram, led to intermediate levels of endoxifen. In a retrospective cohort study, Kelly et al32 demonstrated that women treated with paroxetine, in combination with tamoxifen, had an increased risk of death compared with women treated with other SSRIs or venlafaxine and tamoxifen. They estimated that paroxetine use in women treated with tamoxifen would lead to 1 additional breast cancer death per 20 women within 5 years of discontinuing tamoxifen.
According to American Psychiatric Association practice guidelines for treatment of MDD, depressed breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on CYP2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine.33
Serotonin-norepinephrine reuptake inhibitors (SNRIs) may be used to treat depressive disorders. In addition, venlafaxine may be helpful in treating post-mastectomy pain syndrome. Approximately one-half of patients who undergo mastectomy or breast reconstruction may experience a postoperative pain syndrome.34 The most common symptom is a burning, stabbing pain in the axilla, arm, and chest wall of the affected side. This pain is worsened by movement and is poorly responsive to opioids.35
In a 10-week RCT of 13 patients with neuropathic pain after breast cancer treatment, venlafaxine significantly improved pain relief compared with placebo, although the drug did not affect depression or anxiety.36 In a study of 100 patients given venlafaxine or placebo for 2 weeks starting the night before undergoing partial or radical mastectomy with axillary dissection, those receiving venlafaxine had a significantly lower incidence of pain in the chest wall, arm, and axillary region, and scores of pain with movement were decreased.37 There was no difference in opioid usage between groups.
Tricyclic antidepressants have been demonstrated to be effective in breast cancer patients. Side effects—notably anticholinergic effects—limit their use as antidepressants, especially when compared with SSRI treatment. In a study that randomly assigned 179 women with breast cancer to paroxetine, 20 to 40 mg/d, or amitriptyline, 75 to 150 mg/d, anticholinergic effects were almost twice as frequent in the amitriptyline group (19%) compared with paroxetine (11%).38 In a 4-week double-blind, placebo-controlled crossover trial of 15 breast cancer patients, amitriptyline significantly relieved neuropathic pain, but its adverse effects made most patients unwilling to use the medication regularly.39
Table 2
Evidence supporting SSRI use in patients with breast cancer*
Study | Design | Results |
---|---|---|
Navari et al, 200826 | 193 patients with newly diagnosed early-stage breast cancer were randomized to fluoxetine, 20 mg/d, or placebo for 6 months | Fluoxetine reduced depressive symptoms, improved quality of life, and led to higher completion of adjuvant chemotherapy and/or hormone therapy |
Roscoe et al, 200527 | 94 women with breast cancer receiving at least 4 cycles of chemotherapy were randomized to paroxetine, 20 mg/d, or placebo | Paroxetine significantly reduced depression during chemotherapy |
Kimick et al, 200628 | 62 women with early-stage breast cancer receiving the chemotherapy agent tamoxifen who reported hot flashes were randomized to sertraline, 50 mg/d, or placebo for 6 weeks | Sertraline was significantly more effective than placebo at reducing hot flashes |
* Breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on cytochrome P450 2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine SSRIs: selective serotonin reuptake inhibitors |
Related Resources
- American Cancer Society. www.cancer.org.
- National Comprehensive Cancer network. www.nccn.org.
Drug Brand Names
- Amitripyline • Elavil
- Citalopram • Celexa
- Desvenlafaxine • Pristiq
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Paroxetine • Paxil
- Sertraline • Zoloft
- Tamoxifen • Nolvadex
- Venlafaxine • Effexor
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/depression-treatment-for-women-with.html#comments
Psychological distress among patients with breast cancer is common and is linked to worse clinical outcomes. Depressive and anxiety symptoms affect up to 40% of breast cancer patients,1 and depression is associated with a higher relative risk of mortality in individuals with breast cancer.2 Psychotropic medications and psychotherapy used to treat depression in patients without carcinoma also are appropriate and effective for breast cancer patients. However, some patients present distinct challenges to standard treatment. For example, growing evidence suggests that some selective serotonin reuptake inhibitors (SSRIs) may reduce the effectiveness of tamoxifen, a chemotherapeutic agent. This article discusses challenges in diagnosing and treating depression in breast cancer patients and reviews evidence supporting appropriate psychiatric care.
Increased vulnerability
In 10% to 30% of women, a breast cancer diagnosis may lead to increased vulnerability to depressive disorders, including adjustment disorders with depressed mood, major depressive disorder (MDD), and mood disorders related to general medical conditions.3,4 The risk of developing a depressive disorder is highest in the year after receiving the breast cancer diagnosis.4
A woman’s risk of developing a depressive disorder may depend on the type of cancer treatment she receives. For example, breast asymmetry is common after breast conserving surgery. Waljee et al5 found that women with breast asymmetry had increased fears of cancer recurrence and more feelings of self-consciousness. More pronounced asymmetry led to a higher incidence of depressive symptoms. However, among 90 patients undergoing bilateral prophylactic mastectomy, the rate of depression had not changed 1 year after the procedure.6 Chemotherapy, particularly at high doses, is a risk factor for depression.4,7,8
Self-blame for developing breast cancer can affect mood. In 2007, Friedman et al9 determined that higher levels of self-blame correlated with higher levels of depression and decreased quality of life. Women often blamed themselves for various reasons, including:
- poor coping skills
- anxiety about their health and treatments
- inability to express emotions
- delays in medical consultation.
Exacerbated symptoms and side effects. Women with depression often experience increased side effects from cancer treatments, and the subjective experience of these effects—including hot flashes, cognitive impairment, pain, and sexual dysfunction—likely is intensified.4 Somatic symptoms of depression may be exacerbated by cancer treatment side effects or mistaken for effects of the treatment. When somatic symptoms of depression are mistaken for treatment side effects, depression—and the opportunity to treat it—can be overlooked.10
Depression may be a risk factor for poor adherence to cancer treatment. In a quantitative review of studies correlating depression and medical treatment noncompliance, DiMatteo et al11 determined that compared with nondepressed patients, those with depression were 3 times more likely to not adhere to treatment recommendations; this review was not limited to cancer patients. Depressive symptoms—notably poor concentration and amotivation—can create the impression that a patient is poorly adherent. Women with comorbid depression and breast cancer may have difficulty understanding treatment recommendations or remembering daily treatment goals.4
Appropriate screening tools
Factors that may increase a breast cancer patient’s risk for developing a psychiatric disorder are listed in Table 1.10 Many depression screening tools are available; below we describe 3 commonly used for patients with breast cancer.
The National Comprehensive Cancer Center Distress Thermometer allows patients to rate their overall distress level over the past week on a scale from 0 to 10, using a visual analogue.12 The Distress Thermometer has been validated for several cancer populations and in different parts of the world. A score of 7 has both good sensitivity and specificity for detecting depression in breast cancer patients. Consider a complete psychiatric evaluation for patients with scores ≥7.13
The Profile of Mood States questionnaire14 is a reliable, valid 65-item questionnaire often used in studies of mood dysregulation and breast cancer. Subscales include depression-dejection, tension-anxiety, anger-hostility, confusion-bewilderment, vigor-activity, and fatigue-inertia. Using a 5-point Likert scale, patients rate their symptoms over the past week. Subscale scores are then added to a total mood disturbance score.14,15
The Hospital Anxiety and Depression Scale (HADS) is a sensitive, reliable 14-item scale that is commonly used to study depression and anxiety in patients with breast cancer.16 HADS includes two 7-item subscales—anxiety and depression—and answers are scored on a 4-point Likert scale. Patients are asked to respond quickly and avoid thinking too long about their answers.
Table 1
Risk factors for psychiatric distress related to breast cancer
Past psychiatric illness |
Family history of psychiatric illness |
Younger age (<45 years) |
Having young children |
Limited social support |
Substance use |
Single status |
Pain |
Physical disability |
Poor family coherence |
Financial strain |
Source: Adapted from reference 10 |
Psychotherapeutic options
Behavioral therapies can diminish symptoms of depression, according to a review of studies and practice guidelines on managing depression in cancer patients.17 Group interventions, in particular, can be valuable. Anderson et al18 found that group cohesion, member connectedness, and more sessions correlated with decreased psychological distress.
Psychoeducation aims to provide medical information and discuss cancer’s causes, prognosis, and treatment strategies. Group settings can help improve communication and problem-solving skills. In a randomized controlled trial (RCT) of 203 women with breast cancer, psychoeducational group treatment reduced depression, anger, and fatigue.19
Cognitive-behavioral therapy (CBT) helps patients identify and restructure negative thoughts and increase positive, adaptive behaviors. Hunter et al20 noted significant improvement in depressed mood, anxiety, and sleep in 17 women experiencing menopausal symptoms who received group CBT after completing breast cancer treatment. In 1 study, 124 patients with metastatic breast cancer who received 8 weekly sessions of group CBT reported reduced depression and mood disturbance and improved self-esteem compared with a no-therapy control group.21
Supportive-expressive therapy (SET) is a manual-based therapy that focuses on increasing social support, improving symptom control, and enhancing communication between the patient and treatment team. Affective expression helps lead the therapist to issues that should be addressed. Evidence on the effectiveness of SET for patients with breast cancer is mixed. A study of 357 women with breast cancer who were randomly assigned to 12-week group SET or an educational control condition found no evidence that SET reduced distress.22 However, a trial of 485 women with advanced breast cancer who were randomly assigned to group SET plus relaxation therapy or relaxation therapy alone showed that SET improved quality of life, including protection against depression.23
Mindfulness-based stress reduction (MBSR) is a standardized form of meditation and yoga. Clinicians teach patients visualization, breathing exercises, and meditation to help them become aware of the body’s reaction to stress and how to regulate it. In an RCT of 84 female breast cancer survivors, a 6-week MBSR program diminished depressive symptoms, improved physical functioning, and reduced fear of cancer recurrence.24
Evidence for antidepressants
SSRIs. Expert consensus guidelines on treating depression in women recommend an SSRI as a first-line agent.25 In RCTs, fluoxetine, paroxetine, and sertraline were more effective than placebo in treating depression and related symptoms specifically in women with breast cancer (Table 2).26-28
The interaction between SSRIs and chemotherapy agents is a concern. Tamoxifen decreases the rate of death from breast cancer in hormone receptor positive breast cancers.29 Endoxifen, a potent anti-estrogen, is an active metabolite of tamoxifen via cytochrome P450 (CYP) 2D6. Goetz et al30 demonstrated that women with decreased or inhibited metabolism via CYP2D6 had significantly shorter times to breast cancer recurrence, compared with women with extensive CYP2D6 metabolism.
SSRIs can varyingly inhibit CYP2D6. In a prospective trial of 158 breast cancer patients receiving tamoxifen, paroxetine and fluoxetine were found to be strong inhibitors of CYP2D6 and led to low levels of endoxifen.31 In contrast, weaker inhibitors, including sertraline and citalopram, led to intermediate levels of endoxifen. In a retrospective cohort study, Kelly et al32 demonstrated that women treated with paroxetine, in combination with tamoxifen, had an increased risk of death compared with women treated with other SSRIs or venlafaxine and tamoxifen. They estimated that paroxetine use in women treated with tamoxifen would lead to 1 additional breast cancer death per 20 women within 5 years of discontinuing tamoxifen.
According to American Psychiatric Association practice guidelines for treatment of MDD, depressed breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on CYP2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine.33
Serotonin-norepinephrine reuptake inhibitors (SNRIs) may be used to treat depressive disorders. In addition, venlafaxine may be helpful in treating post-mastectomy pain syndrome. Approximately one-half of patients who undergo mastectomy or breast reconstruction may experience a postoperative pain syndrome.34 The most common symptom is a burning, stabbing pain in the axilla, arm, and chest wall of the affected side. This pain is worsened by movement and is poorly responsive to opioids.35
In a 10-week RCT of 13 patients with neuropathic pain after breast cancer treatment, venlafaxine significantly improved pain relief compared with placebo, although the drug did not affect depression or anxiety.36 In a study of 100 patients given venlafaxine or placebo for 2 weeks starting the night before undergoing partial or radical mastectomy with axillary dissection, those receiving venlafaxine had a significantly lower incidence of pain in the chest wall, arm, and axillary region, and scores of pain with movement were decreased.37 There was no difference in opioid usage between groups.
Tricyclic antidepressants have been demonstrated to be effective in breast cancer patients. Side effects—notably anticholinergic effects—limit their use as antidepressants, especially when compared with SSRI treatment. In a study that randomly assigned 179 women with breast cancer to paroxetine, 20 to 40 mg/d, or amitriptyline, 75 to 150 mg/d, anticholinergic effects were almost twice as frequent in the amitriptyline group (19%) compared with paroxetine (11%).38 In a 4-week double-blind, placebo-controlled crossover trial of 15 breast cancer patients, amitriptyline significantly relieved neuropathic pain, but its adverse effects made most patients unwilling to use the medication regularly.39
Table 2
Evidence supporting SSRI use in patients with breast cancer*
Study | Design | Results |
---|---|---|
Navari et al, 200826 | 193 patients with newly diagnosed early-stage breast cancer were randomized to fluoxetine, 20 mg/d, or placebo for 6 months | Fluoxetine reduced depressive symptoms, improved quality of life, and led to higher completion of adjuvant chemotherapy and/or hormone therapy |
Roscoe et al, 200527 | 94 women with breast cancer receiving at least 4 cycles of chemotherapy were randomized to paroxetine, 20 mg/d, or placebo | Paroxetine significantly reduced depression during chemotherapy |
Kimick et al, 200628 | 62 women with early-stage breast cancer receiving the chemotherapy agent tamoxifen who reported hot flashes were randomized to sertraline, 50 mg/d, or placebo for 6 weeks | Sertraline was significantly more effective than placebo at reducing hot flashes |
* Breast cancer patients who receive tamoxifen generally should be treated with an antidepressant that has minimal effect on cytochrome P450 2D6 metabolism, such as citalopram, escitalopram, venlafaxine, or desvenlafaxine SSRIs: selective serotonin reuptake inhibitors |
Related Resources
- American Cancer Society. www.cancer.org.
- National Comprehensive Cancer network. www.nccn.org.
Drug Brand Names
- Amitripyline • Elavil
- Citalopram • Celexa
- Desvenlafaxine • Pristiq
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Paroxetine • Paxil
- Sertraline • Zoloft
- Tamoxifen • Nolvadex
- Venlafaxine • Effexor
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Von Ah D, Kang DH. Correlates of mood disturbance in women with breast cancer: patterns over time. J Adv Nurs. 2008;61(6):676-689.
2. Hjerl K, Andersen EW, Keiding N, et al. Depression as a prognostic factor for breast cancer mortality. Psychosomatics. 2003;44:24-30.
3. Stanton AL. Psychosocial concerns and interventions for cancer survivors. J Clin Oncol. 2006;24(32):5132-5137.
4. Fann JR, Thomas-Rich AM, Katon WJ, et al. Major depression after breast cancer: a review of epidemiology and treatment. Gen Hosp Psychiatry. 2008;30:112-126.
5. Waljee JF, Hu ES, Ubel PA, et al. Effect of esthetic outcome after breast-conserving surgery on psychosocial functioning and quality of life. J Clin Oncol. 2008;26(20):3331-3337.
6. Brandberg Y, Sandelin K, Erikson S, et al. Psychological reactions, quality of life, and body image after bilateral prophylactic mastectomy in women at high risk for breast cancer: a prospective 1-year follow-up study. J Clin Oncol. 2008;26(24):3943-3949.
7. Lee KC, Ray T, Hunkeler E, et al. Tamoxifen treatment and new onset depression in breast cancer patients. Psychosomatics. 2007;48:205-210.
8. Thornton LM, Carson WE, Shapiro CL, et al. Delayed emotional recovery after taxane-based chemotherapy. Cancer. 2008;113(3):638-647.
9. Friedman LC, Romero C, Elledge R, et al. Attribution of blame, self-forgiving attitude and psychological adjustment in women with breast cancer. J Behav Med. 2007;30:351-357.
10. Spiegel D, Riba M. Psychological aspects of cancer. In: DeVita VT, Lawrence TS, Rosenberg SA, eds. Principles and practice of oncology. 8th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2008:2817–2826.
11. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment. Arch Intern Med. 2000;160:2101-2107.
12. National Comprehensive Cancer Network. Practice guidelines in oncology—distress management v.1.2010. 2010. Available at: http://www.nccn.org. Accessed October 5, 2010.
13. Hegel MT, Collins ED, Kearing S. Sensitivity and specificity of the distress thermometer for depression in newly diagnosed breast cancer patients. Psychooncology. 2008;17:556-560.
14. Lorr M, McNair DM, Droppleman LF. POMS profile of mood states. Available at: http://www.mhs.com/product.aspx?gr=cli&prod=poms&id=overview. Accessed September 29, 2010.
15. Classen C, Butler LD, Koopman C. Supportive-expressive group therapy and distress in patients with metastatic breast cancer: a randomized clinical intervention trial. Arch Gen Psychiatry. 2001;58:494-501.
16. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67:361-370.
17. Barsevick AM, Sweeney C, Haney E, et al. A systematic qualitative analysis of psychoeducational interventions for depression in patients with cancer. Oncol Nurs Forum. 2002;29(1):73-84.
18. Andersen BL, Shelby RA, Golden-Kreutz DM. RCT of a psychological intervention for patients with cancer: I. Mechanisms of change. J Consult Clin Psychol. 2007;75(6):927-938.
19. Dolbreault S, Cayrou S, Bredart A, et al. The effectiveness of a psycho-educational group after early-stage breast cancer treatment: results of a randomized French study. Psychooncology. 2009;18:647-656.
20. Hunter MS, Coventry S, Hamed H, et al. Evaluation of a group cognitive behavioural intervention for women suffering from menopausal symptoms following breast cancer treatment. Psychooncology. 2009;18(5):560-563.
21. Edelman S, Bell DR, Kidman AD. A group cognitive behaviour therapy programme with metastatic breast cancer patients. Psychooncology. 1999;8(4):295-305.
22. Classen CC, Kraemer HC, Blasey C, et al. Supportive-expressive group therapy for primary breast cancer patients: a randomized prospective multicenter trial. Psychooncology. 2008;17:438-447.
23. Kissane DW, Grabsch B, Clarke DM, et al. Supportive-expressive group therapy for women with metastatic breast cancer: survival and psychosocial outcome from a randomized control trial. Psychooncology. 2007;16(4):277-286.
24. Lengacher CA, Johnson-Mallard V, Post-White J, et al. Randomized controlled trial of mindfulness-based stress reduction (MBSR) for survivors of breast cancer. Psychooncology. 2009;18:1261-1272.
25. Altshuler LL, Cohen LS, Moline ML, et al. Treatment of depression in women: a summary of expert consensus guidelines. J Psychiatr Pract. 2001;7(3):185-208.
26. Navari RM, Brenner MC, Wilson MN. Treatment of depressive symptoms in patients with early stage breast cancer undergoing adjuvant therapy. Breast Cancer Res Treat. 2008;112(1):197-201.
27. Roscoe JA, Morrow GR, Hickok JT, et al. Effect of paroxetine hydrochloride on fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res Treat. 2005;89(3):243-249.
28. Kimmick GG, Lovato J, McQuellon R, et al. Randomized, double-blind, placebo-controlled crossover study of sertraline (Zoloft) for treatment of hot flashes in women with early stage breast cancer taking tamoxifen. Breast J. 2006;12(2):114-122.
29. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomized trials. Lancet. 2005;365(9472):1687-1717.
30. Goetz MP, Knox SK, Suman VJ, et al. The impact of cytochrome P450 2D6 metabolism in women receiving adjuvant tamoxifen. Breast Cancer Res Treat. 2007;101(1):113-121.
31. Borges S, Desta Z, Li L, et al. Quantitative effect of CYP2D6 genotype and inhibitors on tamoxifen metabolism: implication for optimization of breast cancer treatment. Clin Pharmacol Ther. 2006;80(1):61-74.
32. Kelly CM, Juurlink DN, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population based cohort study. BMJ. 2010;8:340;c693.-
33. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. 3rd ed. Arlington, VA: American Psychiatric Publishing, Inc. 2010.
34. Vadivelu N, Schreck M, Lopez J, et al. Pain after mastectomy and breast reconstruction. Am Surg. 2008;74(4):285-296.
35. Stevens PE, Dibble SL, Miaskowski C. Prevalence, characteristics and impact of post-mastectomy pain syndrome: an investigation of women’s experiences. Pain. 1995;61:61-68.
36. Tasmuth T, Hartel B, Kalso ET. Venlafaxine in neuropathic pain following treatment of breast cancer. Eur J Pain. 2002;6:17-24.
37. Reuben SS, Makari-Judson G, Lurie SD. Evaluation of efficacy of the perioperative administration of venlafaxine XR in the prevention of post-mastectomy pain syndrome. J Pain Sympt Manage. 2004;27(2):133-139.
38. 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. 2001;70(1):1-10.
39. Kalso ET, Tasmuth T, Neuvonen PJ. Amitriptyline effectively relieves neuropathic pain following treatment of breast cancer. Pain. 1996;64:293-302.
1. Von Ah D, Kang DH. Correlates of mood disturbance in women with breast cancer: patterns over time. J Adv Nurs. 2008;61(6):676-689.
2. Hjerl K, Andersen EW, Keiding N, et al. Depression as a prognostic factor for breast cancer mortality. Psychosomatics. 2003;44:24-30.
3. Stanton AL. Psychosocial concerns and interventions for cancer survivors. J Clin Oncol. 2006;24(32):5132-5137.
4. Fann JR, Thomas-Rich AM, Katon WJ, et al. Major depression after breast cancer: a review of epidemiology and treatment. Gen Hosp Psychiatry. 2008;30:112-126.
5. Waljee JF, Hu ES, Ubel PA, et al. Effect of esthetic outcome after breast-conserving surgery on psychosocial functioning and quality of life. J Clin Oncol. 2008;26(20):3331-3337.
6. Brandberg Y, Sandelin K, Erikson S, et al. Psychological reactions, quality of life, and body image after bilateral prophylactic mastectomy in women at high risk for breast cancer: a prospective 1-year follow-up study. J Clin Oncol. 2008;26(24):3943-3949.
7. Lee KC, Ray T, Hunkeler E, et al. Tamoxifen treatment and new onset depression in breast cancer patients. Psychosomatics. 2007;48:205-210.
8. Thornton LM, Carson WE, Shapiro CL, et al. Delayed emotional recovery after taxane-based chemotherapy. Cancer. 2008;113(3):638-647.
9. Friedman LC, Romero C, Elledge R, et al. Attribution of blame, self-forgiving attitude and psychological adjustment in women with breast cancer. J Behav Med. 2007;30:351-357.
10. Spiegel D, Riba M. Psychological aspects of cancer. In: DeVita VT, Lawrence TS, Rosenberg SA, eds. Principles and practice of oncology. 8th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2008:2817–2826.
11. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment. Arch Intern Med. 2000;160:2101-2107.
12. National Comprehensive Cancer Network. Practice guidelines in oncology—distress management v.1.2010. 2010. Available at: http://www.nccn.org. Accessed October 5, 2010.
13. Hegel MT, Collins ED, Kearing S. Sensitivity and specificity of the distress thermometer for depression in newly diagnosed breast cancer patients. Psychooncology. 2008;17:556-560.
14. Lorr M, McNair DM, Droppleman LF. POMS profile of mood states. Available at: http://www.mhs.com/product.aspx?gr=cli&prod=poms&id=overview. Accessed September 29, 2010.
15. Classen C, Butler LD, Koopman C. Supportive-expressive group therapy and distress in patients with metastatic breast cancer: a randomized clinical intervention trial. Arch Gen Psychiatry. 2001;58:494-501.
16. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67:361-370.
17. Barsevick AM, Sweeney C, Haney E, et al. A systematic qualitative analysis of psychoeducational interventions for depression in patients with cancer. Oncol Nurs Forum. 2002;29(1):73-84.
18. Andersen BL, Shelby RA, Golden-Kreutz DM. RCT of a psychological intervention for patients with cancer: I. Mechanisms of change. J Consult Clin Psychol. 2007;75(6):927-938.
19. Dolbreault S, Cayrou S, Bredart A, et al. The effectiveness of a psycho-educational group after early-stage breast cancer treatment: results of a randomized French study. Psychooncology. 2009;18:647-656.
20. Hunter MS, Coventry S, Hamed H, et al. Evaluation of a group cognitive behavioural intervention for women suffering from menopausal symptoms following breast cancer treatment. Psychooncology. 2009;18(5):560-563.
21. Edelman S, Bell DR, Kidman AD. A group cognitive behaviour therapy programme with metastatic breast cancer patients. Psychooncology. 1999;8(4):295-305.
22. Classen CC, Kraemer HC, Blasey C, et al. Supportive-expressive group therapy for primary breast cancer patients: a randomized prospective multicenter trial. Psychooncology. 2008;17:438-447.
23. Kissane DW, Grabsch B, Clarke DM, et al. Supportive-expressive group therapy for women with metastatic breast cancer: survival and psychosocial outcome from a randomized control trial. Psychooncology. 2007;16(4):277-286.
24. Lengacher CA, Johnson-Mallard V, Post-White J, et al. Randomized controlled trial of mindfulness-based stress reduction (MBSR) for survivors of breast cancer. Psychooncology. 2009;18:1261-1272.
25. Altshuler LL, Cohen LS, Moline ML, et al. Treatment of depression in women: a summary of expert consensus guidelines. J Psychiatr Pract. 2001;7(3):185-208.
26. Navari RM, Brenner MC, Wilson MN. Treatment of depressive symptoms in patients with early stage breast cancer undergoing adjuvant therapy. Breast Cancer Res Treat. 2008;112(1):197-201.
27. Roscoe JA, Morrow GR, Hickok JT, et al. Effect of paroxetine hydrochloride on fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res Treat. 2005;89(3):243-249.
28. Kimmick GG, Lovato J, McQuellon R, et al. Randomized, double-blind, placebo-controlled crossover study of sertraline (Zoloft) for treatment of hot flashes in women with early stage breast cancer taking tamoxifen. Breast J. 2006;12(2):114-122.
29. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomized trials. Lancet. 2005;365(9472):1687-1717.
30. Goetz MP, Knox SK, Suman VJ, et al. The impact of cytochrome P450 2D6 metabolism in women receiving adjuvant tamoxifen. Breast Cancer Res Treat. 2007;101(1):113-121.
31. Borges S, Desta Z, Li L, et al. Quantitative effect of CYP2D6 genotype and inhibitors on tamoxifen metabolism: implication for optimization of breast cancer treatment. Clin Pharmacol Ther. 2006;80(1):61-74.
32. Kelly CM, Juurlink DN, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population based cohort study. BMJ. 2010;8:340;c693.-
33. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. 3rd ed. Arlington, VA: American Psychiatric Publishing, Inc. 2010.
34. Vadivelu N, Schreck M, Lopez J, et al. Pain after mastectomy and breast reconstruction. Am Surg. 2008;74(4):285-296.
35. Stevens PE, Dibble SL, Miaskowski C. Prevalence, characteristics and impact of post-mastectomy pain syndrome: an investigation of women’s experiences. Pain. 1995;61:61-68.
36. Tasmuth T, Hartel B, Kalso ET. Venlafaxine in neuropathic pain following treatment of breast cancer. Eur J Pain. 2002;6:17-24.
37. Reuben SS, Makari-Judson G, Lurie SD. Evaluation of efficacy of the perioperative administration of venlafaxine XR in the prevention of post-mastectomy pain syndrome. J Pain Sympt Manage. 2004;27(2):133-139.
38. 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. 2001;70(1):1-10.
39. Kalso ET, Tasmuth T, Neuvonen PJ. Amitriptyline effectively relieves neuropathic pain following treatment of breast cancer. Pain. 1996;64:293-302.
Parricide: Characteristics of sons and daughters who kill their parents
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/parricide-characteristics-of-sons-and.html#comments
Mr. B, age 37, is single and lives with his elderly mother. Since being diagnosed with schizophrenia in his early 20s, he has been intermittently compliant with antipsychotic therapy. When unmedicated, Mr. B develops paranoid delusions and becomes preoccupied with the idea that his mother is plotting to kill him. He has been hospitalized twice in the last 5 years for physical aggression toward his mother. In the last 10 years, Mr. B has been placed in several group homes, but when he takes his medications, he is able to convince his mother to allow him to live with her.
During his most recent stay in his mother’s home, Mr. B again stops taking his psychotropic medications and decompensates. His mother becomes concerned about her son’s paranoid behavior—such as trying to listen in on her telephone conversations and smelling his food before he eats it—and considers having her son involuntarily committed. One day, after she prepares Mr. B a sandwich, he decides the meat is poisoned. When his mother tries to convince him to eat the sandwich, Mr. B becomes enraged and stabs her 54 times with a kitchen knife.
Mr. B is arrested without resistance. He is adjudicated incompetent to stand trial and is restored to competency within 3 months. Mr. B is found not guilty by reason of insanity (NGRI) and is civilly committed to a state psychiatric facility.
Parricide—killing one’s parents—once was referred to as “the schizophrenic crime,”1 but is now recognized as being more complex.2 In the United States, parricides accounted for 2% of all homicides from 1976 to 1998,3 which is consistent with studies from France4 and the United Kingdom.5 Parricide’s scandalous nature has long attracted the public’s fascination (see this article at CurrentPsychiatry.com).
This article primarily focuses on the interplay of the diagnostic and demographic factors seen in adults who kill their biological parents but briefly notes differences seen in juvenile perpetrators and those who kill their stepparents. Knowledge of these characteristics can help clinicians identify and more safely manage patients who may be at risk of harming their parents.
The public maintains a morbid curiosity about parricide. In ancient times, the Roman emperor Nero was responsible for the death of his mother, Agrippina. In 1892, Lizzie Borden attracted national attention—and inspired a children’s song about “40 whacks”—when she was suspected, but acquitted, of murdering her father and stepmother. Charles Whitman, infamous for his 1966 killing spree from the University of Texas at Austin tower, killed his mother before his rampage. In 1993, the trial of the Menendez brothers, who were eventually convicted of murdering their parents, was broadcast on Court TV.
Parricide also plays a role in literature and popular culture. Oedipus would have never been able to marry his mother had he not first killed his father. In the movie Psycho, Alfred Hitchcock told the story of Norman Bates, a hotel owner who killed his mother and preserved her body in the basement. In the novel Carrie, Stephen King uses matricide as a means to sever the relationship between the main character and her domineering mother. In 1989, the band Aerosmith released a song, Janie’s Got a Gun, about a girl who kills her father after he sexually abused her.
A limited evidence base
The common themes found in the literature on parricide should be interpreted cautiously because of the limitations of this research. The number of individuals assessed in these studies often is small, which limits the statistical power of the findings. Studies often are conducted in forensic hospitals, which excludes those who are imprisoned or commit suicide following the acts. Finally, most individuals studied were diagnosed with a psychiatric disorder after the crime, which makes it difficult to distinguish the primary illness from the crime’s effect on a person’s mental state. Additionally, some individuals may be tempted to exaggerate or feign psychiatric symptoms in an effort to be found NGRI or granted leniency during sentencing. Despite these limitations, several conclusions can be drawn from these investigations.
The sex of the victims and perpetrators needs to be carefully considered when reviewing characteristics of those who commit parricide. Killing a mother is matricide, and killing a father is patricide.
Sons who kill their parents
Men are more likely to kill their parents than women.6-9 In a study of 5,488 cases of parricide in the United States, 4,738 (86%) of perpetrators were male.3 Common characteristics of men who commit parricide are listed in Table 1.5,8,10-14
Matricide by sons. Although sons kill their fathers more often than their mothers,15 authors writing about parricide commonly focus on men who commit matricide. Wertham described sons who kill their mothers in terms of the “Orestes Complex,” which refers to ambivalent feelings toward the mother that ultimately manifest in homicidal rage. He noted that many matricides are committed with excessive force, occur in the bedroom, and are precipitated by trivial reasons. Wertham stated that these crimes represent the son’s unconscious hatred for his mother superimposed on sexual desire for her.16 Sigmund Freud argued that matricide served as a displacement defense against incestuous impulses.17
In 5 studies that looked at sons who killed their mothers (n=13 to 58),5,10-13 most of which examined men residing in forensic hospitals after the crime, perpetrators were noted to be immature, dependent, and passive. In a study of 16 men with schizophrenia who committed matricide, subjects perceived themselves as “weak, small, inadequate, hopeless, doubtful about sexual identity, dependent, and unable to accept a separate, adult male role.”11 Mothers generally were domineering, demanding, and possessive.
Based on our literature review, most men who committed matricide had a schizophrenia diagnosis (weighted mean 72%, range 50% to 100%); other diagnoses included depression and personality disorders. Many men were experiencing psychosis shortly before the crime, and their acts were influenced by persecutory delusions and/or auditory hallucinations. Approximately one-quarter of sons killed their mothers for altruistic reasons, such as to relieve actual or perceived suffering.
Nearly all men in these 5 studies were single and lived with their mothers before killing them, and many of the perpetrators’ fathers were absent. Mothers often were the only victims of their sons’ violent acts. In addition to delusional beliefs, sons were motivated to kill their mothers for various reasons, including threatened separation or minor arguments (eg, over food or money). Many of these homicides took place in the home. Sharp or blunt objects were the most common weapons, but guns and strangulation/asphyxiation also were used. Approximately one-half of the men used excessive violence; for example, 1 victim had 177 stab wounds. After the crimes, the perpetrators generally expressed remorse or relief.
Patricide by sons. Psychoanalysts may consider the Oedipal Complex to be the primary impetus for a son to commit patricide. By eliminating his father the son gains possession of his mother.18 Three studies looked at sons who killed their fathers; 2 examined 10 perpetrators residing in a forensic hospital after the crimes8,14 and the third was based on coroners’ reports.10 Although the sons’ personality traits were not described, the fathers were noted to be “domineering and aggressive,” and their relationships with their sons were “cruel and unusual.”8 In our review of these studies, >50% of sons were diagnosed with schizophrenia (weighted mean 60%, range 49% to 80%). Many perpetrators exhibited psychotic symptoms, including delusions and hallucinations. In 1 study, 40% of sons with psychotic symptoms perceived their fathers as posing “threats of physical or psychological annihilation.”14
In 2 of these studies all of the sons were single or separated from their spouses.8,14 Most killed only their fathers at the time of the act. Immediately before the crime, one-half of the fathers were consuming alcohol and/or arguing with their sons. Ninety percent of the fathers were killed by excessive violence. Following the acts, the sons described feeling “relief rather than remorse or guilt…leading to a feeling of freedom from the abnormal relationship.”14 One study noted that, in the course of legal proceedings, one-fifth were deemed competent to stand trial and the others were found to be incompetent and hospitalized.14
Table 1
Sons who kill their parents: Schizophrenia is common
Sons who kill their mothers | Sons who kill their fathers |
---|---|
Sons:
| Sons:
|
Mothers:
| Fathers:
|
Crime:
| Crime:
|
Source: References 5,8,10-14 |
Daughters who commit parricide
d’Orban and O’Connor conducted the only major study examining women who commit parricide,9 a retrospective evaluation of 17 women who killed a parent and were housed in a prison or hospital. The authors highlight the importance of delusional beliefs as a motive for parricide (Table 2).9
In a 1970 Japanese study of 21 women who killed parents or in-laws, half of the victims were mothers-in-law, but none were biological mothers.19 According to the authors, this finding suggests that relationships between Japanese women and their mothers-in-law often are particularly contentious; however, no research has examined this theory in the United States.
Matricide by daughters. In the d’Orban and O’Connor study,9 >80% of women who committed parricide killed their mothers. In general, the daughters were described as being “in mid-life, living alone with an elderly, domineering mother in marked social isolation.” The parent-child relationship was “characterized by mutual hostility and dependence.” Seventy-five percent of the daughters suffered from psychotic illness. Extreme violence often was used.
Patricide by daughters. Of the 3 women who killed their fathers in d’Orban and O’Connor’s study,9 none were psychotic. Furthermore, 2 women had no psychiatric diagnosis—the third had antisocial personality disorder—and “killed tyrannical fathers in response to prolonged parental violence.” One woman reported that she was forced into a long-term incestuous relationship before killing her father. The women who killed their fathers were younger (mean age 21.3) than those who killed their mothers (mean age 39.5).
Table 2
Daughters who kill their parents: Strained relationships
Daughters who kill their mothers | Daughters who kill their fathers |
---|---|
Daughters:
| Daughters:
|
Mothers:
| Fathers:
|
Crime:
| |
Source: Reference 9 |
Other perpetrators and victims
Patricide is most often committed by adults20; however, some important conclusions can be drawn regarding juveniles who kill their parents (Table 3).21-27 The most common scenario is of adolescent boys who have no history of psychosis and kill their fathers in a burst of rage brought on by ongoing abuse from parents. These murders typically are followed by feelings of relief rather than remorse.21-27
Stepparents often have a more challenging relationship with children than biological parents.28 Research indicates that stepparents are more likely than biological parents to be killed by juvenile offenders.29 Also, stepparent victims tended to be younger than biological parent victims.29
Table 3
Characteristics of juveniles who kill their parents or stepparents
Often teenage boys |
Generally lack history of psychosis |
Actions often are spontaneous |
Motivated by long-term parental abuse |
Often feel relief rather than remorse after the crime |
More likely to kill stepparents than biological parents |
Source: References 21-27 |
Clinical applications
Ask adult schizophrenia patients living with a parent about the quality of the relationship. If the relationship is characterized by conflict or abuse or if psychotic symptoms are present, assess for violent thoughts toward the parent. For patients with uncontrolled psychosis coupled with a contentious parental relationship, in addition to aggressively treating psychotic symptoms, consider initiating family therapy, anger management classes, group home placement, or involuntary hospitalization to lower the risk of parricide.
Related resources
- Heide KM, Boots DP. A comparative analysis of media reports of U.S. parricide cases with officially reported national crime data and the psychiatric and psychological literature. Int J Offender Ther Comp Criminol. 2007;51(6):646-675.
- Jacobs A. On matricide: myth, psychoanalysis, and the law of the mother. New York, NY: Columbia University Press; 2007.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
1. Gillies H. Murder in the west of Scotland. Br J Psychiatry. 1965;111:1087-1094.
2. Clark SA. Matricide: the schizophrenic crime? Med Sci Law. 1993;33(4):325-328.
3. Federal Bureau of Investigation. Crime in the United States. Washington, DC: Department of Justice; 1998.
4. Devaux C, Petit G, Perol Y, et al. Enquête sure le parricide en France. Ann Med Psychol (Paris). 1974;1:161-168.
5. Green C. Matricide by sons. Med Sci Law. 1981;21:207-214.
6. Marleau JD, Millaud F, Auclair N. A comparison of parricide and attempted parricide: a study of 39 psychotic adults. Int J Law Psychiatry. 2003;26(3):269-279.
7. Weisman AM, Ehrenclou MG, Sharma KK. Double parricide: forensic analysis and psycholegal implications. J Forensic Sci. 2002;47(2):313-317.
8. Singhal S, Dutta A. Who commits patricide? Acta Psychiatr Scand. 1990;82:40-43.
9. d’Orban PT, O’Connor A. Women who kill their parents. Br J Psychiatry. 1989;154:27-33.
10. Bourget D, Gagné P, Labelle ME. Parricide: a comparative study of matricide versus patricide. J Am Acad Psychiatry Law. 2007;35(3):306-312.
11. Singhal S, Dutta A. Who commits matricide? Med Sci Law. 1992;32:213-217.
12. Campion J, Cravens JM, Rotholc A, et al. A study of 15 matricidal men. Am J Psychiatry. 1985;142:312-317.
13. O’Connell B. Matricide (report of a meeting of the Royal Medico-Psychological Association). Lancet. 1963;1:1083-1084.
14. Cravens JM, Campion J, Rotholc A, et al. A study of 10 men charged with patricide. Am J Psychiatry. 1985;142(9):1089-1092.
15. Shon PC, Targonski JR. Declining trends in U.S. parricides, 1976-1978: testing the Freudian assumptions. Int J Law Psychiatry. 2003;26:387-402.
16. Wertham F. Dark legend: a study in murder. New York, NY: Duell, Sloan and Pearce; 1941.
17. Freud S. The interpretation of dreams. Strachey J, trans. New York, NY: Discus; 1925.
18. Freud S. Sigmund Freud: collected papers. Vol 5. New York, NY: Basic Books; 1959.
19. Hirose K. A psychiatric study of female homicide: on the cases of parricide. Acta Criminologiae et Medicinae Legalis Japonica. 1970;36:29.-
20. Heide KM. Parents who get killed and the children who kill them. J Interpers Violence. 1993;8(4):531-544.
21. Hellsten P, Katila O. Murder and homicide by children under 15 in Finland. Psychiatr Q Suppl. 1965;39:54-74.
22. Scherl DJ, Mack JE. A study of adolescent matricide. J Am Acad Child Psychiatry. 1966;5:569-593.
23. Sadoff RL. Clinical observations on parricide. Psychiatr Q. 1971;45:65-69.
24. Tanay E. Proceedings: adolescents who kill parents—reactive parricide. Aust N Z J Psychiatry. 1973;7:263-277.
25. Tuovinen M. On parricide. Psychiatrica Fennica. 1973;141-146.
26. Corder BF, Ball BC, Haizlip TM, et al. Adolescent parricide: a comparison with other adolescent murder. Am J Psychiatry. 1976;133:957-961.
27. Post S. Adolescent parricide in abusive families. Child Welfare. 1982;61:445-455.
28. Daly M, Wilson M. Evolutionary social psychology and family homicide. Science. 1988;242:520-524.
29. Heide KM. Why kids kill parents: child abuse and adolescent homicide. Columbus, OH: Ohio State University Press; 1992.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/parricide-characteristics-of-sons-and.html#comments
Mr. B, age 37, is single and lives with his elderly mother. Since being diagnosed with schizophrenia in his early 20s, he has been intermittently compliant with antipsychotic therapy. When unmedicated, Mr. B develops paranoid delusions and becomes preoccupied with the idea that his mother is plotting to kill him. He has been hospitalized twice in the last 5 years for physical aggression toward his mother. In the last 10 years, Mr. B has been placed in several group homes, but when he takes his medications, he is able to convince his mother to allow him to live with her.
During his most recent stay in his mother’s home, Mr. B again stops taking his psychotropic medications and decompensates. His mother becomes concerned about her son’s paranoid behavior—such as trying to listen in on her telephone conversations and smelling his food before he eats it—and considers having her son involuntarily committed. One day, after she prepares Mr. B a sandwich, he decides the meat is poisoned. When his mother tries to convince him to eat the sandwich, Mr. B becomes enraged and stabs her 54 times with a kitchen knife.
Mr. B is arrested without resistance. He is adjudicated incompetent to stand trial and is restored to competency within 3 months. Mr. B is found not guilty by reason of insanity (NGRI) and is civilly committed to a state psychiatric facility.
Parricide—killing one’s parents—once was referred to as “the schizophrenic crime,”1 but is now recognized as being more complex.2 In the United States, parricides accounted for 2% of all homicides from 1976 to 1998,3 which is consistent with studies from France4 and the United Kingdom.5 Parricide’s scandalous nature has long attracted the public’s fascination (see this article at CurrentPsychiatry.com).
This article primarily focuses on the interplay of the diagnostic and demographic factors seen in adults who kill their biological parents but briefly notes differences seen in juvenile perpetrators and those who kill their stepparents. Knowledge of these characteristics can help clinicians identify and more safely manage patients who may be at risk of harming their parents.
The public maintains a morbid curiosity about parricide. In ancient times, the Roman emperor Nero was responsible for the death of his mother, Agrippina. In 1892, Lizzie Borden attracted national attention—and inspired a children’s song about “40 whacks”—when she was suspected, but acquitted, of murdering her father and stepmother. Charles Whitman, infamous for his 1966 killing spree from the University of Texas at Austin tower, killed his mother before his rampage. In 1993, the trial of the Menendez brothers, who were eventually convicted of murdering their parents, was broadcast on Court TV.
Parricide also plays a role in literature and popular culture. Oedipus would have never been able to marry his mother had he not first killed his father. In the movie Psycho, Alfred Hitchcock told the story of Norman Bates, a hotel owner who killed his mother and preserved her body in the basement. In the novel Carrie, Stephen King uses matricide as a means to sever the relationship between the main character and her domineering mother. In 1989, the band Aerosmith released a song, Janie’s Got a Gun, about a girl who kills her father after he sexually abused her.
A limited evidence base
The common themes found in the literature on parricide should be interpreted cautiously because of the limitations of this research. The number of individuals assessed in these studies often is small, which limits the statistical power of the findings. Studies often are conducted in forensic hospitals, which excludes those who are imprisoned or commit suicide following the acts. Finally, most individuals studied were diagnosed with a psychiatric disorder after the crime, which makes it difficult to distinguish the primary illness from the crime’s effect on a person’s mental state. Additionally, some individuals may be tempted to exaggerate or feign psychiatric symptoms in an effort to be found NGRI or granted leniency during sentencing. Despite these limitations, several conclusions can be drawn from these investigations.
The sex of the victims and perpetrators needs to be carefully considered when reviewing characteristics of those who commit parricide. Killing a mother is matricide, and killing a father is patricide.
Sons who kill their parents
Men are more likely to kill their parents than women.6-9 In a study of 5,488 cases of parricide in the United States, 4,738 (86%) of perpetrators were male.3 Common characteristics of men who commit parricide are listed in Table 1.5,8,10-14
Matricide by sons. Although sons kill their fathers more often than their mothers,15 authors writing about parricide commonly focus on men who commit matricide. Wertham described sons who kill their mothers in terms of the “Orestes Complex,” which refers to ambivalent feelings toward the mother that ultimately manifest in homicidal rage. He noted that many matricides are committed with excessive force, occur in the bedroom, and are precipitated by trivial reasons. Wertham stated that these crimes represent the son’s unconscious hatred for his mother superimposed on sexual desire for her.16 Sigmund Freud argued that matricide served as a displacement defense against incestuous impulses.17
In 5 studies that looked at sons who killed their mothers (n=13 to 58),5,10-13 most of which examined men residing in forensic hospitals after the crime, perpetrators were noted to be immature, dependent, and passive. In a study of 16 men with schizophrenia who committed matricide, subjects perceived themselves as “weak, small, inadequate, hopeless, doubtful about sexual identity, dependent, and unable to accept a separate, adult male role.”11 Mothers generally were domineering, demanding, and possessive.
Based on our literature review, most men who committed matricide had a schizophrenia diagnosis (weighted mean 72%, range 50% to 100%); other diagnoses included depression and personality disorders. Many men were experiencing psychosis shortly before the crime, and their acts were influenced by persecutory delusions and/or auditory hallucinations. Approximately one-quarter of sons killed their mothers for altruistic reasons, such as to relieve actual or perceived suffering.
Nearly all men in these 5 studies were single and lived with their mothers before killing them, and many of the perpetrators’ fathers were absent. Mothers often were the only victims of their sons’ violent acts. In addition to delusional beliefs, sons were motivated to kill their mothers for various reasons, including threatened separation or minor arguments (eg, over food or money). Many of these homicides took place in the home. Sharp or blunt objects were the most common weapons, but guns and strangulation/asphyxiation also were used. Approximately one-half of the men used excessive violence; for example, 1 victim had 177 stab wounds. After the crimes, the perpetrators generally expressed remorse or relief.
Patricide by sons. Psychoanalysts may consider the Oedipal Complex to be the primary impetus for a son to commit patricide. By eliminating his father the son gains possession of his mother.18 Three studies looked at sons who killed their fathers; 2 examined 10 perpetrators residing in a forensic hospital after the crimes8,14 and the third was based on coroners’ reports.10 Although the sons’ personality traits were not described, the fathers were noted to be “domineering and aggressive,” and their relationships with their sons were “cruel and unusual.”8 In our review of these studies, >50% of sons were diagnosed with schizophrenia (weighted mean 60%, range 49% to 80%). Many perpetrators exhibited psychotic symptoms, including delusions and hallucinations. In 1 study, 40% of sons with psychotic symptoms perceived their fathers as posing “threats of physical or psychological annihilation.”14
In 2 of these studies all of the sons were single or separated from their spouses.8,14 Most killed only their fathers at the time of the act. Immediately before the crime, one-half of the fathers were consuming alcohol and/or arguing with their sons. Ninety percent of the fathers were killed by excessive violence. Following the acts, the sons described feeling “relief rather than remorse or guilt…leading to a feeling of freedom from the abnormal relationship.”14 One study noted that, in the course of legal proceedings, one-fifth were deemed competent to stand trial and the others were found to be incompetent and hospitalized.14
Table 1
Sons who kill their parents: Schizophrenia is common
Sons who kill their mothers | Sons who kill their fathers |
---|---|
Sons:
| Sons:
|
Mothers:
| Fathers:
|
Crime:
| Crime:
|
Source: References 5,8,10-14 |
Daughters who commit parricide
d’Orban and O’Connor conducted the only major study examining women who commit parricide,9 a retrospective evaluation of 17 women who killed a parent and were housed in a prison or hospital. The authors highlight the importance of delusional beliefs as a motive for parricide (Table 2).9
In a 1970 Japanese study of 21 women who killed parents or in-laws, half of the victims were mothers-in-law, but none were biological mothers.19 According to the authors, this finding suggests that relationships between Japanese women and their mothers-in-law often are particularly contentious; however, no research has examined this theory in the United States.
Matricide by daughters. In the d’Orban and O’Connor study,9 >80% of women who committed parricide killed their mothers. In general, the daughters were described as being “in mid-life, living alone with an elderly, domineering mother in marked social isolation.” The parent-child relationship was “characterized by mutual hostility and dependence.” Seventy-five percent of the daughters suffered from psychotic illness. Extreme violence often was used.
Patricide by daughters. Of the 3 women who killed their fathers in d’Orban and O’Connor’s study,9 none were psychotic. Furthermore, 2 women had no psychiatric diagnosis—the third had antisocial personality disorder—and “killed tyrannical fathers in response to prolonged parental violence.” One woman reported that she was forced into a long-term incestuous relationship before killing her father. The women who killed their fathers were younger (mean age 21.3) than those who killed their mothers (mean age 39.5).
Table 2
Daughters who kill their parents: Strained relationships
Daughters who kill their mothers | Daughters who kill their fathers |
---|---|
Daughters:
| Daughters:
|
Mothers:
| Fathers:
|
Crime:
| |
Source: Reference 9 |
Other perpetrators and victims
Patricide is most often committed by adults20; however, some important conclusions can be drawn regarding juveniles who kill their parents (Table 3).21-27 The most common scenario is of adolescent boys who have no history of psychosis and kill their fathers in a burst of rage brought on by ongoing abuse from parents. These murders typically are followed by feelings of relief rather than remorse.21-27
Stepparents often have a more challenging relationship with children than biological parents.28 Research indicates that stepparents are more likely than biological parents to be killed by juvenile offenders.29 Also, stepparent victims tended to be younger than biological parent victims.29
Table 3
Characteristics of juveniles who kill their parents or stepparents
Often teenage boys |
Generally lack history of psychosis |
Actions often are spontaneous |
Motivated by long-term parental abuse |
Often feel relief rather than remorse after the crime |
More likely to kill stepparents than biological parents |
Source: References 21-27 |
Clinical applications
Ask adult schizophrenia patients living with a parent about the quality of the relationship. If the relationship is characterized by conflict or abuse or if psychotic symptoms are present, assess for violent thoughts toward the parent. For patients with uncontrolled psychosis coupled with a contentious parental relationship, in addition to aggressively treating psychotic symptoms, consider initiating family therapy, anger management classes, group home placement, or involuntary hospitalization to lower the risk of parricide.
Related resources
- Heide KM, Boots DP. A comparative analysis of media reports of U.S. parricide cases with officially reported national crime data and the psychiatric and psychological literature. Int J Offender Ther Comp Criminol. 2007;51(6):646-675.
- Jacobs A. On matricide: myth, psychoanalysis, and the law of the mother. New York, NY: Columbia University Press; 2007.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/parricide-characteristics-of-sons-and.html#comments
Mr. B, age 37, is single and lives with his elderly mother. Since being diagnosed with schizophrenia in his early 20s, he has been intermittently compliant with antipsychotic therapy. When unmedicated, Mr. B develops paranoid delusions and becomes preoccupied with the idea that his mother is plotting to kill him. He has been hospitalized twice in the last 5 years for physical aggression toward his mother. In the last 10 years, Mr. B has been placed in several group homes, but when he takes his medications, he is able to convince his mother to allow him to live with her.
During his most recent stay in his mother’s home, Mr. B again stops taking his psychotropic medications and decompensates. His mother becomes concerned about her son’s paranoid behavior—such as trying to listen in on her telephone conversations and smelling his food before he eats it—and considers having her son involuntarily committed. One day, after she prepares Mr. B a sandwich, he decides the meat is poisoned. When his mother tries to convince him to eat the sandwich, Mr. B becomes enraged and stabs her 54 times with a kitchen knife.
Mr. B is arrested without resistance. He is adjudicated incompetent to stand trial and is restored to competency within 3 months. Mr. B is found not guilty by reason of insanity (NGRI) and is civilly committed to a state psychiatric facility.
Parricide—killing one’s parents—once was referred to as “the schizophrenic crime,”1 but is now recognized as being more complex.2 In the United States, parricides accounted for 2% of all homicides from 1976 to 1998,3 which is consistent with studies from France4 and the United Kingdom.5 Parricide’s scandalous nature has long attracted the public’s fascination (see this article at CurrentPsychiatry.com).
This article primarily focuses on the interplay of the diagnostic and demographic factors seen in adults who kill their biological parents but briefly notes differences seen in juvenile perpetrators and those who kill their stepparents. Knowledge of these characteristics can help clinicians identify and more safely manage patients who may be at risk of harming their parents.
The public maintains a morbid curiosity about parricide. In ancient times, the Roman emperor Nero was responsible for the death of his mother, Agrippina. In 1892, Lizzie Borden attracted national attention—and inspired a children’s song about “40 whacks”—when she was suspected, but acquitted, of murdering her father and stepmother. Charles Whitman, infamous for his 1966 killing spree from the University of Texas at Austin tower, killed his mother before his rampage. In 1993, the trial of the Menendez brothers, who were eventually convicted of murdering their parents, was broadcast on Court TV.
Parricide also plays a role in literature and popular culture. Oedipus would have never been able to marry his mother had he not first killed his father. In the movie Psycho, Alfred Hitchcock told the story of Norman Bates, a hotel owner who killed his mother and preserved her body in the basement. In the novel Carrie, Stephen King uses matricide as a means to sever the relationship between the main character and her domineering mother. In 1989, the band Aerosmith released a song, Janie’s Got a Gun, about a girl who kills her father after he sexually abused her.
A limited evidence base
The common themes found in the literature on parricide should be interpreted cautiously because of the limitations of this research. The number of individuals assessed in these studies often is small, which limits the statistical power of the findings. Studies often are conducted in forensic hospitals, which excludes those who are imprisoned or commit suicide following the acts. Finally, most individuals studied were diagnosed with a psychiatric disorder after the crime, which makes it difficult to distinguish the primary illness from the crime’s effect on a person’s mental state. Additionally, some individuals may be tempted to exaggerate or feign psychiatric symptoms in an effort to be found NGRI or granted leniency during sentencing. Despite these limitations, several conclusions can be drawn from these investigations.
The sex of the victims and perpetrators needs to be carefully considered when reviewing characteristics of those who commit parricide. Killing a mother is matricide, and killing a father is patricide.
Sons who kill their parents
Men are more likely to kill their parents than women.6-9 In a study of 5,488 cases of parricide in the United States, 4,738 (86%) of perpetrators were male.3 Common characteristics of men who commit parricide are listed in Table 1.5,8,10-14
Matricide by sons. Although sons kill their fathers more often than their mothers,15 authors writing about parricide commonly focus on men who commit matricide. Wertham described sons who kill their mothers in terms of the “Orestes Complex,” which refers to ambivalent feelings toward the mother that ultimately manifest in homicidal rage. He noted that many matricides are committed with excessive force, occur in the bedroom, and are precipitated by trivial reasons. Wertham stated that these crimes represent the son’s unconscious hatred for his mother superimposed on sexual desire for her.16 Sigmund Freud argued that matricide served as a displacement defense against incestuous impulses.17
In 5 studies that looked at sons who killed their mothers (n=13 to 58),5,10-13 most of which examined men residing in forensic hospitals after the crime, perpetrators were noted to be immature, dependent, and passive. In a study of 16 men with schizophrenia who committed matricide, subjects perceived themselves as “weak, small, inadequate, hopeless, doubtful about sexual identity, dependent, and unable to accept a separate, adult male role.”11 Mothers generally were domineering, demanding, and possessive.
Based on our literature review, most men who committed matricide had a schizophrenia diagnosis (weighted mean 72%, range 50% to 100%); other diagnoses included depression and personality disorders. Many men were experiencing psychosis shortly before the crime, and their acts were influenced by persecutory delusions and/or auditory hallucinations. Approximately one-quarter of sons killed their mothers for altruistic reasons, such as to relieve actual or perceived suffering.
Nearly all men in these 5 studies were single and lived with their mothers before killing them, and many of the perpetrators’ fathers were absent. Mothers often were the only victims of their sons’ violent acts. In addition to delusional beliefs, sons were motivated to kill their mothers for various reasons, including threatened separation or minor arguments (eg, over food or money). Many of these homicides took place in the home. Sharp or blunt objects were the most common weapons, but guns and strangulation/asphyxiation also were used. Approximately one-half of the men used excessive violence; for example, 1 victim had 177 stab wounds. After the crimes, the perpetrators generally expressed remorse or relief.
Patricide by sons. Psychoanalysts may consider the Oedipal Complex to be the primary impetus for a son to commit patricide. By eliminating his father the son gains possession of his mother.18 Three studies looked at sons who killed their fathers; 2 examined 10 perpetrators residing in a forensic hospital after the crimes8,14 and the third was based on coroners’ reports.10 Although the sons’ personality traits were not described, the fathers were noted to be “domineering and aggressive,” and their relationships with their sons were “cruel and unusual.”8 In our review of these studies, >50% of sons were diagnosed with schizophrenia (weighted mean 60%, range 49% to 80%). Many perpetrators exhibited psychotic symptoms, including delusions and hallucinations. In 1 study, 40% of sons with psychotic symptoms perceived their fathers as posing “threats of physical or psychological annihilation.”14
In 2 of these studies all of the sons were single or separated from their spouses.8,14 Most killed only their fathers at the time of the act. Immediately before the crime, one-half of the fathers were consuming alcohol and/or arguing with their sons. Ninety percent of the fathers were killed by excessive violence. Following the acts, the sons described feeling “relief rather than remorse or guilt…leading to a feeling of freedom from the abnormal relationship.”14 One study noted that, in the course of legal proceedings, one-fifth were deemed competent to stand trial and the others were found to be incompetent and hospitalized.14
Table 1
Sons who kill their parents: Schizophrenia is common
Sons who kill their mothers | Sons who kill their fathers |
---|---|
Sons:
| Sons:
|
Mothers:
| Fathers:
|
Crime:
| Crime:
|
Source: References 5,8,10-14 |
Daughters who commit parricide
d’Orban and O’Connor conducted the only major study examining women who commit parricide,9 a retrospective evaluation of 17 women who killed a parent and were housed in a prison or hospital. The authors highlight the importance of delusional beliefs as a motive for parricide (Table 2).9
In a 1970 Japanese study of 21 women who killed parents or in-laws, half of the victims were mothers-in-law, but none were biological mothers.19 According to the authors, this finding suggests that relationships between Japanese women and their mothers-in-law often are particularly contentious; however, no research has examined this theory in the United States.
Matricide by daughters. In the d’Orban and O’Connor study,9 >80% of women who committed parricide killed their mothers. In general, the daughters were described as being “in mid-life, living alone with an elderly, domineering mother in marked social isolation.” The parent-child relationship was “characterized by mutual hostility and dependence.” Seventy-five percent of the daughters suffered from psychotic illness. Extreme violence often was used.
Patricide by daughters. Of the 3 women who killed their fathers in d’Orban and O’Connor’s study,9 none were psychotic. Furthermore, 2 women had no psychiatric diagnosis—the third had antisocial personality disorder—and “killed tyrannical fathers in response to prolonged parental violence.” One woman reported that she was forced into a long-term incestuous relationship before killing her father. The women who killed their fathers were younger (mean age 21.3) than those who killed their mothers (mean age 39.5).
Table 2
Daughters who kill their parents: Strained relationships
Daughters who kill their mothers | Daughters who kill their fathers |
---|---|
Daughters:
| Daughters:
|
Mothers:
| Fathers:
|
Crime:
| |
Source: Reference 9 |
Other perpetrators and victims
Patricide is most often committed by adults20; however, some important conclusions can be drawn regarding juveniles who kill their parents (Table 3).21-27 The most common scenario is of adolescent boys who have no history of psychosis and kill their fathers in a burst of rage brought on by ongoing abuse from parents. These murders typically are followed by feelings of relief rather than remorse.21-27
Stepparents often have a more challenging relationship with children than biological parents.28 Research indicates that stepparents are more likely than biological parents to be killed by juvenile offenders.29 Also, stepparent victims tended to be younger than biological parent victims.29
Table 3
Characteristics of juveniles who kill their parents or stepparents
Often teenage boys |
Generally lack history of psychosis |
Actions often are spontaneous |
Motivated by long-term parental abuse |
Often feel relief rather than remorse after the crime |
More likely to kill stepparents than biological parents |
Source: References 21-27 |
Clinical applications
Ask adult schizophrenia patients living with a parent about the quality of the relationship. If the relationship is characterized by conflict or abuse or if psychotic symptoms are present, assess for violent thoughts toward the parent. For patients with uncontrolled psychosis coupled with a contentious parental relationship, in addition to aggressively treating psychotic symptoms, consider initiating family therapy, anger management classes, group home placement, or involuntary hospitalization to lower the risk of parricide.
Related resources
- Heide KM, Boots DP. A comparative analysis of media reports of U.S. parricide cases with officially reported national crime data and the psychiatric and psychological literature. Int J Offender Ther Comp Criminol. 2007;51(6):646-675.
- Jacobs A. On matricide: myth, psychoanalysis, and the law of the mother. New York, NY: Columbia University Press; 2007.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
1. Gillies H. Murder in the west of Scotland. Br J Psychiatry. 1965;111:1087-1094.
2. Clark SA. Matricide: the schizophrenic crime? Med Sci Law. 1993;33(4):325-328.
3. Federal Bureau of Investigation. Crime in the United States. Washington, DC: Department of Justice; 1998.
4. Devaux C, Petit G, Perol Y, et al. Enquête sure le parricide en France. Ann Med Psychol (Paris). 1974;1:161-168.
5. Green C. Matricide by sons. Med Sci Law. 1981;21:207-214.
6. Marleau JD, Millaud F, Auclair N. A comparison of parricide and attempted parricide: a study of 39 psychotic adults. Int J Law Psychiatry. 2003;26(3):269-279.
7. Weisman AM, Ehrenclou MG, Sharma KK. Double parricide: forensic analysis and psycholegal implications. J Forensic Sci. 2002;47(2):313-317.
8. Singhal S, Dutta A. Who commits patricide? Acta Psychiatr Scand. 1990;82:40-43.
9. d’Orban PT, O’Connor A. Women who kill their parents. Br J Psychiatry. 1989;154:27-33.
10. Bourget D, Gagné P, Labelle ME. Parricide: a comparative study of matricide versus patricide. J Am Acad Psychiatry Law. 2007;35(3):306-312.
11. Singhal S, Dutta A. Who commits matricide? Med Sci Law. 1992;32:213-217.
12. Campion J, Cravens JM, Rotholc A, et al. A study of 15 matricidal men. Am J Psychiatry. 1985;142:312-317.
13. O’Connell B. Matricide (report of a meeting of the Royal Medico-Psychological Association). Lancet. 1963;1:1083-1084.
14. Cravens JM, Campion J, Rotholc A, et al. A study of 10 men charged with patricide. Am J Psychiatry. 1985;142(9):1089-1092.
15. Shon PC, Targonski JR. Declining trends in U.S. parricides, 1976-1978: testing the Freudian assumptions. Int J Law Psychiatry. 2003;26:387-402.
16. Wertham F. Dark legend: a study in murder. New York, NY: Duell, Sloan and Pearce; 1941.
17. Freud S. The interpretation of dreams. Strachey J, trans. New York, NY: Discus; 1925.
18. Freud S. Sigmund Freud: collected papers. Vol 5. New York, NY: Basic Books; 1959.
19. Hirose K. A psychiatric study of female homicide: on the cases of parricide. Acta Criminologiae et Medicinae Legalis Japonica. 1970;36:29.-
20. Heide KM. Parents who get killed and the children who kill them. J Interpers Violence. 1993;8(4):531-544.
21. Hellsten P, Katila O. Murder and homicide by children under 15 in Finland. Psychiatr Q Suppl. 1965;39:54-74.
22. Scherl DJ, Mack JE. A study of adolescent matricide. J Am Acad Child Psychiatry. 1966;5:569-593.
23. Sadoff RL. Clinical observations on parricide. Psychiatr Q. 1971;45:65-69.
24. Tanay E. Proceedings: adolescents who kill parents—reactive parricide. Aust N Z J Psychiatry. 1973;7:263-277.
25. Tuovinen M. On parricide. Psychiatrica Fennica. 1973;141-146.
26. Corder BF, Ball BC, Haizlip TM, et al. Adolescent parricide: a comparison with other adolescent murder. Am J Psychiatry. 1976;133:957-961.
27. Post S. Adolescent parricide in abusive families. Child Welfare. 1982;61:445-455.
28. Daly M, Wilson M. Evolutionary social psychology and family homicide. Science. 1988;242:520-524.
29. Heide KM. Why kids kill parents: child abuse and adolescent homicide. Columbus, OH: Ohio State University Press; 1992.
1. Gillies H. Murder in the west of Scotland. Br J Psychiatry. 1965;111:1087-1094.
2. Clark SA. Matricide: the schizophrenic crime? Med Sci Law. 1993;33(4):325-328.
3. Federal Bureau of Investigation. Crime in the United States. Washington, DC: Department of Justice; 1998.
4. Devaux C, Petit G, Perol Y, et al. Enquête sure le parricide en France. Ann Med Psychol (Paris). 1974;1:161-168.
5. Green C. Matricide by sons. Med Sci Law. 1981;21:207-214.
6. Marleau JD, Millaud F, Auclair N. A comparison of parricide and attempted parricide: a study of 39 psychotic adults. Int J Law Psychiatry. 2003;26(3):269-279.
7. Weisman AM, Ehrenclou MG, Sharma KK. Double parricide: forensic analysis and psycholegal implications. J Forensic Sci. 2002;47(2):313-317.
8. Singhal S, Dutta A. Who commits patricide? Acta Psychiatr Scand. 1990;82:40-43.
9. d’Orban PT, O’Connor A. Women who kill their parents. Br J Psychiatry. 1989;154:27-33.
10. Bourget D, Gagné P, Labelle ME. Parricide: a comparative study of matricide versus patricide. J Am Acad Psychiatry Law. 2007;35(3):306-312.
11. Singhal S, Dutta A. Who commits matricide? Med Sci Law. 1992;32:213-217.
12. Campion J, Cravens JM, Rotholc A, et al. A study of 15 matricidal men. Am J Psychiatry. 1985;142:312-317.
13. O’Connell B. Matricide (report of a meeting of the Royal Medico-Psychological Association). Lancet. 1963;1:1083-1084.
14. Cravens JM, Campion J, Rotholc A, et al. A study of 10 men charged with patricide. Am J Psychiatry. 1985;142(9):1089-1092.
15. Shon PC, Targonski JR. Declining trends in U.S. parricides, 1976-1978: testing the Freudian assumptions. Int J Law Psychiatry. 2003;26:387-402.
16. Wertham F. Dark legend: a study in murder. New York, NY: Duell, Sloan and Pearce; 1941.
17. Freud S. The interpretation of dreams. Strachey J, trans. New York, NY: Discus; 1925.
18. Freud S. Sigmund Freud: collected papers. Vol 5. New York, NY: Basic Books; 1959.
19. Hirose K. A psychiatric study of female homicide: on the cases of parricide. Acta Criminologiae et Medicinae Legalis Japonica. 1970;36:29.-
20. Heide KM. Parents who get killed and the children who kill them. J Interpers Violence. 1993;8(4):531-544.
21. Hellsten P, Katila O. Murder and homicide by children under 15 in Finland. Psychiatr Q Suppl. 1965;39:54-74.
22. Scherl DJ, Mack JE. A study of adolescent matricide. J Am Acad Child Psychiatry. 1966;5:569-593.
23. Sadoff RL. Clinical observations on parricide. Psychiatr Q. 1971;45:65-69.
24. Tanay E. Proceedings: adolescents who kill parents—reactive parricide. Aust N Z J Psychiatry. 1973;7:263-277.
25. Tuovinen M. On parricide. Psychiatrica Fennica. 1973;141-146.
26. Corder BF, Ball BC, Haizlip TM, et al. Adolescent parricide: a comparison with other adolescent murder. Am J Psychiatry. 1976;133:957-961.
27. Post S. Adolescent parricide in abusive families. Child Welfare. 1982;61:445-455.
28. Daly M, Wilson M. Evolutionary social psychology and family homicide. Science. 1988;242:520-524.
29. Heide KM. Why kids kill parents: child abuse and adolescent homicide. Columbus, OH: Ohio State University Press; 1992.
Hallucinations in children: Diagnostic and treatment strategies
Discuss this article at http://currentpsychiatry.blogspot.com/2010/10/hallucinations-in-children-diagnostic.html#comments
Hallucinations in children are of grave concern to parents and clinicians, but aren’t necessarily a symptom of mental illness. In adults, hallucinations usually are linked to serious psychopathology; however, in children they are not uncommon and may be part of normal development (Box).
A hallucination is a false auditory, visual, gustatory, tactile, or olfactory perception not associated with real external stimuli.1 It must be differentiated from similar phenomenon such as illusions (misperception of actual stimuli), elaborate fantasies, imaginary companions, and eidetic images (visual images stored in memory).
Although hallucinations frequently are considered synonymous with psychotic disorders, in children this rare. Neurobiologic studies (fMRI) of adults show activation of Broca’s area (left inferior frontal gyrus) seconds before patients perceive auditory verbal hallucinations, which suggests that auditory hallucinations may be misidentified self-talk.a,b According to Piaget,c children age <7 may have difficulty distinguishing between events occurring while dreaming and awake. He further theorized that nonpathologic hallucinations could become pathologic when combined with trauma such as abuse. Straussd suggested that psychosis might lie on a continuum with normal phenomenon. In a case series, Wilking and Paulie described how developmental difficulties, deprivation, sociocultural conditions, and family relationships could contribute to impaired reality testing.
Imaginary friends or companions are common among all children. Children who have imaginary friends are more likely to report hearing “voices.”f Imaginary friends:
- appear, function, and disappear at the wish of the child
- pose no threat and often are a source of comfort
- often can be described in detail
- are not ego-dystonic.g
Also, children with imaginary friends will not show evidence of a thought disorder.
Source:
a. Shergill SS, Brammer MJ, Amaro E, et al. Temporal course of auditory hallucinations. Br J Psychiatry. 2004;185:516-517.
b. Shergill SS, Brammer JJ, Williams SC, et al. Mapping auditory hallucinations in schizophrenia; using functional magnetic resonance imaging. Arch Gen Psychiatry. 2000;57;1033-1038.
c. Piaget J. The child’s conception of the world. London, United Kingdom: Routledge and Kegan Paul; 1929.
d. Strauss JS. Hallucinations and delusions as points on continua function. Rating scale evidence. Arch Gen Psychiatry. 1969;21:581-586.
e. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
f. Pearson D, Burrow A, FitzGerald C, et al. Auditory hallucinations in normal child populations. Pers Individ Dif. 2001;31:401-407.
g. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002.
Common, yet a cause for concern
Epidemiologic studies show 2.8% of adults report hallucinations before age 21.2 Nonpsychotic children as young as age 5 have reported hallucinations.3 Hallucinatory phenomenon may be present in 8% to 21% of all 11-year-old children; two-thirds of these patients have no DSM-IV-TR diagnosis.4,5 However, 1 evaluation of 62 nonpsychotic hallucinating children treated in a psychiatric emergency department (ED):
- 34% had depression
- 22% had attention-deficit/hyperactivity disorder (ADHD)
- 21% had disruptive behavior disorders
- 23% had other diagnoses.6
Studies suggest that children who have hallucinations but no other psychotic symptoms have a better long-term prognosis than those with additional psychotic symptoms.7 A 17-year longitudinal study of children with hallucinations and concurrent emotional and conduct problems found:
- up to 50% of patients still experience hallucinations at age 30
- hallucinations did not significantly predict clinical outcome at age 30
- childhood hallucinations did not increase the risk for psychosis, depression, organic brain disorder, or other psychiatric illnesses.7
In a study of children with psychosis and disruptive disorders, at 2- to 8-years follow-up 50% met criteria for major depressive disorder, bipolar disorder, or schizophreniform disorders.8 In a 15-year longitudinal study of 11-year-olds, self-reported psychotic symptoms—such as delusional beliefs and hallucinatory experiences—predicted a high risk of schizophreniform disorder at age 26.9 These studies suggest that experiencing significant disruptions in thoughts and perceptions during childhood may be related to later development of prominent mood and thought disorders.
Differential diagnosis
Table 1 lists possible causes of hallucination in children.6,10-13 Hallucinations during childhood can occur in the context of several psychiatric disorders, including:
They can also manifest as comorbid or associated symptoms of disorders not commonly associated with hallucinations, such as ADHD, disruptive disorders, anxiety disorders, and prodromal clinical states. Medications, substance use, and organic and metabolic disorders also must be considered in the differential diagnosis (Table 3).
Hallucinations may occur in low-functioning or anxious children, in the context of psychosocial adversity or abuse, and during bereavement of a deceased parent when the surviving parent is emotionally unavailable.11-13,15-17 Rule out hypnagogic and hypnopompic hallucinations, which are predominantly visual hallucinations that occur immediately before falling asleep and during the transition from sleep to wakefulness, respectively.18 Rarely, a child who has had hallucinations for some time may learn to complain of them when he or she is not hallucinating in order to obtain a primary or secondary gain, such as getting attention from caregivers.
Little is known about psychosis and hallucinations in preschoolers (age ≤5); therefore, their language use may help assessment. Because of cognitive immaturity, children often use illogical thinking and loose association and may describe their thoughts as “voices.” This is common in children with language disorders—and sometimes in healthy patients—who may talk about voices because they cannot describe their own thoughts.
Children with ADHD and/or oppositional defiant disorder often are impulsive and show poor judgment and may blame voices for telling them to do bad things. These “hallucinations” may represent internal thoughts battling with the child’s conscience.6 Auditory and visual hallucinations have been reported in children with Tourette syndrome, especially when associated with ADHD or obsessive-compulsive disorder.19
Medical causes. Electrolyte disturbances, metabolic disorders, fever, and serious infections are common nonpsychiatric causes of hallucinations.20 Brain neoplasm—particularly in visual association areas, the temporal lobe, or portions of the optic nerve or retina—also may produce hallucinations, which can be complex with full images.21
Medications such as steroids and anticholinergics may cause hallucinations. Case studies report visual and tactile hallucinations with methylphenidate therapy that resolve after discontinuing the medication.22 Illicit substances, including cannabis, lysergic acid diethylamide (LSD), cocaine, amphetamines, 3,4-methylenedioxymeth-amphetamine (ecstasy), opiates, and barbiturates, can induce hallucinations.
Suspect substance-induced hallucinations if your patient shows:
- acute onset of hallucinations
- dilated pupils
- extreme agitation or drowsiness
- other signs of intoxication.
Hallucinations caused by seizure disorders are rare but can be somatosensory, visual (occipital lobe focus), auditory, olfactory (uncinate, complex partial), or gustatory. The hallucinations may be unformed (flashing lights or rushing noises) or formed (images, spoken words, or music) and could be part of the aura arising from the temporal lobe (dreamlike, flashbacks). Command hallucinations are rare and adult and pediatric patients usually sense they are not real.23
Migraines occur in approximately 5% of prepubertal children and often are comorbid with affective and anxiety disorders.24 Hallucinations associated with migraine commonly are visual, but gustatory, olfactory, and auditory hallucinations also can occur, with or without headaches.3 Any hallucination associated with headaches should be investigated neurologically. Other diagnostic aspects of hallucinations to consider while interviewing children are listed in Table 4.25-28
Table 1
Possible causes of hallucinations in children and adolescents
Normal development |
Nonpsychotic psychopathology |
Psychosocial adversity |
Psychotic illness |
Stress |
Family dysfunction |
Deprivation |
Developmental difficulties |
Sociocultural interaction (immigration) |
Poorly differentiated male and female family roles |
Presence or absence of different mother figures |
Cultural factors (witches, ghosts, spiritualism) |
Hallucination of deceased parent, when unresolved mourning persists in the surviving parent |
Source: References 6,10-13 |
Table 2
Content of hallucinations may point to their cause
Schizophrenia or other psychotic disorders | May hear several voices making a critical commentary Command hallucinations telling patients to harm themselves or others Bizarre voices like ‘a computer in my head’ or aliens Voices of someone familiar or a ‘relative’ Visual hallucinations of devils, scary faces, space creatures, and skeletons |
Depressive disorders | Usually a single voice speaking from outside the patient’s head with derogatory or suicidal content |
Bipolar disorder | Usually involves grandiose ideas about power, worth, knowledge, family, or relationship |
Bereavement | Usually a transient (visual or auditory) perception of the deceased person |
Posttraumatic stress disorder | Transient visual hallucinations, usually with phobic content |
Source: Reference 11 |
Table 3
Hallucinations in young patients: Differential diagnoses
Psychiatric disorders that are usually defined by psychotic features, including:
|
Psychiatric disorders that commonly do not include hallucinations but in which hallucinations can occur as comorbid or associated symptoms, such as:
|
Prodromal and at-risk clinical states of psychiatric disorders (psychotic and mood disorders) |
Medications (steroids, anticholinergics, stimulants) |
Drug intoxication and abuse
|
Organic or nonpsychiatric disorders
|
Metabolic disorders
|
Source: a. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625. b. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380. c. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581. d. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002. e. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34. f. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785. g. Sosland MD, Edelsohn GA. Hallucinations in children and adolescents. Curr Psychiatry Rep. 2005;7:180-188. |
Table 4
Diagnostic considerations when assessing a hallucinating child
General: |
Hallucinations are a symptom, not a diagnosis |
They can have a developmental, neurologic, metabolic, or psychiatric basis |
Visual, gustatory, and olfactory hallucinations suggest a medical- or substance-related origin |
Schizophrenia is rare before age 11 |
In pervasive developmental disorders, schizophrenia is diagnosed only if prominent delusions and hallucinations are present for at least 1 month |
Hallucinations are not uncommon in major depressive disorder but may be associated with higher risk of bipolar disorder |
During interviewing, remember that children: |
are highly suggestible |
may answer questions in the affirmative to get attention or to please the interviewer |
may not fully or partially understand what is being asked |
may blame their misbehavior on voices to escape punishment |
may not distinguish between night terrors and illusions |
Source:References 25-28 |
Psychotic disorder not otherwise specified
(NOS) in children often is overused and misused. One reason could be that DSM-IV-TR does not have a category for hallucinations in nonpsychotic children or for children who are at risk for psychosis. However, recommendations regarding the diagnosis of psychotic disorder NOS note it:
- may be used if uncertainty persists after ruling out all other diagnoses
- should be avoided when hallucinations occur in nonpsychotic children
- should not be used longitudinally if a clinician later determines a specific disorder accounts for the hallucinations.29
Treatment
Addressing underlying medical or psychiatric illness, including substance use, is primary. Some adolescents or children may think that cannabis use is relatively benign. Discuss the risks of cannabis use in an age-appropriate manner.
In the ED. Children or adolescents who present with hallucinations in the ED should undergo a thorough evaluation that explores the differential diagnoses. Suggestions for evaluating patients in this setting appear in Table 5.21
Prodromal or at-risk children. There is no consensus on how early to initiate treatment for children in the prodromal stages of psychosis. Early identification and treatment is imperative because duration of untreated psychosis (DUP) is a primary predictor of treatment response in first-admission patients, and longer DUP corresponds to poorer prognosis in children.30
Assessment scales for early identification of psychosis have limitations because most are not standardized for use in children age <14. To assess symptoms and predict future psychosis in children consider using:
- Scale of Prodromal Symptoms
- Structured Interview for Prodromal Symptoms
- Comprehensive Assessment of At-Risk Mental States
- Bonn Scale for the Assessment of Basic Symptoms.
A hallucinating child may be considered prodromal if he or she has:
- 30% drop in Global Assessment Functioning score in the past month
- a first-degree relative with affective or nonaffective psychotic disorder or schizotypal personality disorder.31
Antipsychotics. When treating children, use antipsychotics with caution and consider short- and long-term risks and benefits. Early treatment is indicated when hallucinations are among a patient’s psychotic symptoms; however, antipsychotic use for children in the prodromal phase lacks consensus. McGlashan et al32 showed that in 60 high-risk patients (mean age 16), olanzapine, 5 to 15 mg/d, reduced conversion to psychosis by 50% over 6 months. McGorry et al33 observed that in 59 ultra-high risk patients (mean age 20), adding low-dose risperidone (1 to 2 mg/d) and cognitive-behavioral therapy (CBT) was superior to case management and supportive psychotherapy in preventing psychosis after 6 months of treatment, but this difference was not maintained at 6 months of follow-up.
CBT slows progression to psychosis in ultra-high risk patients and reduces positive symptoms more specifically than it improves emotional dysfunction.34 CBT for psychosis is based on the concept that auditory hallucinations have an underlying personalized meaning or cognitive schema.35 The initial goal of treatment is to engage the child and understand:
- What do the hallucinations mean to the patient?
- How did they start?
- Can the child start or stop them?
- What does the patient think they are?
The clinician then strives to help the child identify alternative explanations for these hallucinations and develop coping strategies.36 “Normalizing” the voices helps the child attribute these voices to a less anxiety-provoking cause.37 Consider suggesting common reasons for the hallucinations, such as:
- lack of sleep
- isolation
- dehydration
- extreme stress
- strong thoughts (obsessions)
- fever and illness
- lack of food
- drugs and alcohol.
If your patient learns that any of these reasons could explain the hallucinations, he or she may start to have a less delusional understanding of them. Suggest that the voices are “real” only if your patient believes it.
Help children develop coping strategies to control auditory hallucinations such as:
- humming
- listening to music
- reading (forwards and backwards)
- talking to others
- exercising
- singing
- medication
- ignoring the voices.
With normalization and other coping strategies, children with visual hallucinations can learn to transform in their mind the frightful image to a funnier one, which is less anxiety-provoking and gives them a sense of control.
Table 5
Suggestions for evaluating hallucinating children in the ED
Evaluate risk factors for suicidality |
Rule out medical and neurologic causes, including substance abuse/intoxication |
Identify underlying psychopathological, psychosocial, and cultural factors |
Contact key adult informants for collateral information |
Decide if hallucinations are psychotic or nonpsychotic |
Reassure your patients that hearing voices does not mean that they are ‘crazy’ |
Evaluate hallucinations in the context of other features of psychoses (onset, frequency, severity, and chronicity) |
Initiate psychotherapy and antidepressants if needed for patients with underlying depression, anxiety, or PTSD |
ED: emergency department; PTSD: posttraumatic stress disorder Source: Reference 21 |
Related Resources
- Bartels-Velthuis AA, Jenner JA, van de Willige G, et al. Prevalence and correlates of auditory vocal hallucinations in middle childhood. Br J Psychiatry. 2010;196(1):41-46.
- Cepeda C. Psychotic symptoms in children and adolescents: assessment, differential diagnosis, and treatment. New York, NY: Routledge; 2007.
Drug Brand Names
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Risperidone • Risperdal
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Sadock BJ, Sadock VA. Kaplan and Sadock’s concise textbook of clinical psychiatry. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2008:26.
2. Fenning S, Susser ES, Pilowsky DJ, et al. Childhood hallucinations preceding the first psychotic episode. J Nerv Ment Dis. 1997;185:115-117.
3. Schreier HA. Auditory hallucinations in nonpsychotic children with affective syndromes and migraines: report of 13 cases. J Child Neurol. 1998;13:377-382.
4. McGee R, Williams S, Poulton R. Hallucinations in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 2000;39:12-13.
5. Yoshizumi T, Murase S, Honjo S, et al. Hallucinatory experiences in a community sample of Japanese children. J Am Acad Child Adolesc Psychiatry. 2004;43:1030-1036.
6. Edelsohn GA, Rabinovich H, Portnoy R. Hallucinations in nonpsychotic children: findings from a psychiatric emergency service. Ann N Y Acad Sciences. 2003;1008:261-264.
7. Rothstein A. Hallucinatory phenomenon in childhood: a critique of the literature. J Am Acad Child Psychiatry. 1981;20:623-635.
8. Nicholson R, Lenane M, Brookner F, et al. Children and adolescents with psychotic disorder not otherwise specified: a 2- to 8-year follow-up study. Compr Psychiatry. 2001;42:319-325.
9. Poulton R, Caspi A, Moffitt TE, et al. Children’s self reported psychotic symptoms and adult schizophreniform disorder: a 15-year longitudinal study. Arch Gen Psychiatry. 2000;57:1053-1058.
10. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
11. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625.
12. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380.
13. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581.
14. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry. 9th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2003:1276-1277,1283.
15. Famularo R, Kinscherff R, Fenton T. Psychiatric diagnosis of maltreated children: preliminary findings. J Am Acad Child Adolesc Psychiatry. 1992;31:863-867.
16. Putnam FW, Peterson G. Further validation of the child dissociative checklist. Dissociation. 1994;7:204-211.
17. Kaufman J, Birmaher B, Clayton S, et al. Case study: trauma-related hallucinations. J Am Acad Child Adolesc Psychiatry. 1997;36(11):1602-1605.
18. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 8th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2005.
19. Kerbeshian J, Burd L. Are schizophreniform symptoms present in attenuated form in children with Tourette Disorder and other developmental disorders? Can J Psychiatry. 1987;32(2):123-135.
20. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34.
21. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785.
22. Gross-Tsur V, Joseph A, Shalev RS. Hallucinations during methylphenidate therapy. Neurology. 2004;63:753-754.
23. Wyllie E. The treatment of epilepsy: principles and practices, Philadelphia, PA: Lippincott Williams and Wilkins; 1993.
24. Saeed MA, Pumariega AJ, Cinciripini PM. Psychopharmacological management of migraine in children in children and adolescents. J Child Adolesc Psychopharmacol. 1992;2:199-211.
25. Chambers WJ, Puig-Antich J, Tabrizi MA, et al. Psychotic symptoms in prepubertal major depressive disorder. Arch Gen Psychiatry. 1982;39:921-927.
26. Volkmar FR. Childhood and adolescent psychosis: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 1996;35:843-851.
27. First MB, Frances A, Pincus HA. DSM-IV handbook of differential diagnosis. Washington, DC: American Psychiatric Press; 1995.
28. Birmaher B, Ryan ND, Williamson DE, et al. Childhood and adolescent depression: a review of the past 10 years. Part I. J Am Acad Child Adolesc Psychiatry. 1996;35:1427-1439.
29. Meyer SE, Bearden CE, Lux CR, et al. The psychosis prodrome in adolescent patients viewed through the lens of DSM-IV. J Child Adolesc Psychopharmacol. 2005;15(3):434-451.
30. Drake RJ, Haley CJ, Akhtar S, et al. Causes and consequences of duration of untreated psychosis in schizophrenia. Br J Psychiatry. 2000;177:511-515.
31. McGorry PD, Yung AR, Phillips LJ. The “close-in” or ultra high-risk model: a safe and effective strategy for research and clinical intervention in prepsychotic mental disorder. Schizophr Bull. 2003;29(4):771-790.
32. McGlashan TH, Zipursky RB, Perkins DO, et al. Olanzapine versus placebo treatment of schizophrenia prodrome: one year results. Poster presented at: International Congress of Schizophrenia Research, Colorado Springs, CO, April-May 2003.
33. McGorry PD, Yung AR, Phillips LJ, et al. Randomized controlled trial of interventions designed to reduce the risk of progression to first episode psychosis in a clinical sample with subthreshold symptoms. Arch Gen Psychiatry. 2002;59:921-928.
34. French P, Shryane N, Bentall RP, et al. Effects of cognitive therapy on the longitudinal development of psychotic experiences in people at high risk of developing psychosis. Br J Psychiatry Suppl. 2007;51:s82-87.
35. Turkington D, Siddle R. Cognitive therapy for the treatment of delusions. Advances in Psychiatric Treatment. 1998;4:235-242.
36. Sosland MD, Pinninti N. Five ways to quiet auditory hallucinations. Current Psychiatry. 2005;4:40.-
37. Kingdon DG, Turkington D. Cognitive behavioral therapy of schizophrenia, New York, NY: Guilford Press; 1994.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/10/hallucinations-in-children-diagnostic.html#comments
Hallucinations in children are of grave concern to parents and clinicians, but aren’t necessarily a symptom of mental illness. In adults, hallucinations usually are linked to serious psychopathology; however, in children they are not uncommon and may be part of normal development (Box).
A hallucination is a false auditory, visual, gustatory, tactile, or olfactory perception not associated with real external stimuli.1 It must be differentiated from similar phenomenon such as illusions (misperception of actual stimuli), elaborate fantasies, imaginary companions, and eidetic images (visual images stored in memory).
Although hallucinations frequently are considered synonymous with psychotic disorders, in children this rare. Neurobiologic studies (fMRI) of adults show activation of Broca’s area (left inferior frontal gyrus) seconds before patients perceive auditory verbal hallucinations, which suggests that auditory hallucinations may be misidentified self-talk.a,b According to Piaget,c children age <7 may have difficulty distinguishing between events occurring while dreaming and awake. He further theorized that nonpathologic hallucinations could become pathologic when combined with trauma such as abuse. Straussd suggested that psychosis might lie on a continuum with normal phenomenon. In a case series, Wilking and Paulie described how developmental difficulties, deprivation, sociocultural conditions, and family relationships could contribute to impaired reality testing.
Imaginary friends or companions are common among all children. Children who have imaginary friends are more likely to report hearing “voices.”f Imaginary friends:
- appear, function, and disappear at the wish of the child
- pose no threat and often are a source of comfort
- often can be described in detail
- are not ego-dystonic.g
Also, children with imaginary friends will not show evidence of a thought disorder.
Source:
a. Shergill SS, Brammer MJ, Amaro E, et al. Temporal course of auditory hallucinations. Br J Psychiatry. 2004;185:516-517.
b. Shergill SS, Brammer JJ, Williams SC, et al. Mapping auditory hallucinations in schizophrenia; using functional magnetic resonance imaging. Arch Gen Psychiatry. 2000;57;1033-1038.
c. Piaget J. The child’s conception of the world. London, United Kingdom: Routledge and Kegan Paul; 1929.
d. Strauss JS. Hallucinations and delusions as points on continua function. Rating scale evidence. Arch Gen Psychiatry. 1969;21:581-586.
e. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
f. Pearson D, Burrow A, FitzGerald C, et al. Auditory hallucinations in normal child populations. Pers Individ Dif. 2001;31:401-407.
g. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002.
Common, yet a cause for concern
Epidemiologic studies show 2.8% of adults report hallucinations before age 21.2 Nonpsychotic children as young as age 5 have reported hallucinations.3 Hallucinatory phenomenon may be present in 8% to 21% of all 11-year-old children; two-thirds of these patients have no DSM-IV-TR diagnosis.4,5 However, 1 evaluation of 62 nonpsychotic hallucinating children treated in a psychiatric emergency department (ED):
- 34% had depression
- 22% had attention-deficit/hyperactivity disorder (ADHD)
- 21% had disruptive behavior disorders
- 23% had other diagnoses.6
Studies suggest that children who have hallucinations but no other psychotic symptoms have a better long-term prognosis than those with additional psychotic symptoms.7 A 17-year longitudinal study of children with hallucinations and concurrent emotional and conduct problems found:
- up to 50% of patients still experience hallucinations at age 30
- hallucinations did not significantly predict clinical outcome at age 30
- childhood hallucinations did not increase the risk for psychosis, depression, organic brain disorder, or other psychiatric illnesses.7
In a study of children with psychosis and disruptive disorders, at 2- to 8-years follow-up 50% met criteria for major depressive disorder, bipolar disorder, or schizophreniform disorders.8 In a 15-year longitudinal study of 11-year-olds, self-reported psychotic symptoms—such as delusional beliefs and hallucinatory experiences—predicted a high risk of schizophreniform disorder at age 26.9 These studies suggest that experiencing significant disruptions in thoughts and perceptions during childhood may be related to later development of prominent mood and thought disorders.
Differential diagnosis
Table 1 lists possible causes of hallucination in children.6,10-13 Hallucinations during childhood can occur in the context of several psychiatric disorders, including:
They can also manifest as comorbid or associated symptoms of disorders not commonly associated with hallucinations, such as ADHD, disruptive disorders, anxiety disorders, and prodromal clinical states. Medications, substance use, and organic and metabolic disorders also must be considered in the differential diagnosis (Table 3).
Hallucinations may occur in low-functioning or anxious children, in the context of psychosocial adversity or abuse, and during bereavement of a deceased parent when the surviving parent is emotionally unavailable.11-13,15-17 Rule out hypnagogic and hypnopompic hallucinations, which are predominantly visual hallucinations that occur immediately before falling asleep and during the transition from sleep to wakefulness, respectively.18 Rarely, a child who has had hallucinations for some time may learn to complain of them when he or she is not hallucinating in order to obtain a primary or secondary gain, such as getting attention from caregivers.
Little is known about psychosis and hallucinations in preschoolers (age ≤5); therefore, their language use may help assessment. Because of cognitive immaturity, children often use illogical thinking and loose association and may describe their thoughts as “voices.” This is common in children with language disorders—and sometimes in healthy patients—who may talk about voices because they cannot describe their own thoughts.
Children with ADHD and/or oppositional defiant disorder often are impulsive and show poor judgment and may blame voices for telling them to do bad things. These “hallucinations” may represent internal thoughts battling with the child’s conscience.6 Auditory and visual hallucinations have been reported in children with Tourette syndrome, especially when associated with ADHD or obsessive-compulsive disorder.19
Medical causes. Electrolyte disturbances, metabolic disorders, fever, and serious infections are common nonpsychiatric causes of hallucinations.20 Brain neoplasm—particularly in visual association areas, the temporal lobe, or portions of the optic nerve or retina—also may produce hallucinations, which can be complex with full images.21
Medications such as steroids and anticholinergics may cause hallucinations. Case studies report visual and tactile hallucinations with methylphenidate therapy that resolve after discontinuing the medication.22 Illicit substances, including cannabis, lysergic acid diethylamide (LSD), cocaine, amphetamines, 3,4-methylenedioxymeth-amphetamine (ecstasy), opiates, and barbiturates, can induce hallucinations.
Suspect substance-induced hallucinations if your patient shows:
- acute onset of hallucinations
- dilated pupils
- extreme agitation or drowsiness
- other signs of intoxication.
Hallucinations caused by seizure disorders are rare but can be somatosensory, visual (occipital lobe focus), auditory, olfactory (uncinate, complex partial), or gustatory. The hallucinations may be unformed (flashing lights or rushing noises) or formed (images, spoken words, or music) and could be part of the aura arising from the temporal lobe (dreamlike, flashbacks). Command hallucinations are rare and adult and pediatric patients usually sense they are not real.23
Migraines occur in approximately 5% of prepubertal children and often are comorbid with affective and anxiety disorders.24 Hallucinations associated with migraine commonly are visual, but gustatory, olfactory, and auditory hallucinations also can occur, with or without headaches.3 Any hallucination associated with headaches should be investigated neurologically. Other diagnostic aspects of hallucinations to consider while interviewing children are listed in Table 4.25-28
Table 1
Possible causes of hallucinations in children and adolescents
Normal development |
Nonpsychotic psychopathology |
Psychosocial adversity |
Psychotic illness |
Stress |
Family dysfunction |
Deprivation |
Developmental difficulties |
Sociocultural interaction (immigration) |
Poorly differentiated male and female family roles |
Presence or absence of different mother figures |
Cultural factors (witches, ghosts, spiritualism) |
Hallucination of deceased parent, when unresolved mourning persists in the surviving parent |
Source: References 6,10-13 |
Table 2
Content of hallucinations may point to their cause
Schizophrenia or other psychotic disorders | May hear several voices making a critical commentary Command hallucinations telling patients to harm themselves or others Bizarre voices like ‘a computer in my head’ or aliens Voices of someone familiar or a ‘relative’ Visual hallucinations of devils, scary faces, space creatures, and skeletons |
Depressive disorders | Usually a single voice speaking from outside the patient’s head with derogatory or suicidal content |
Bipolar disorder | Usually involves grandiose ideas about power, worth, knowledge, family, or relationship |
Bereavement | Usually a transient (visual or auditory) perception of the deceased person |
Posttraumatic stress disorder | Transient visual hallucinations, usually with phobic content |
Source: Reference 11 |
Table 3
Hallucinations in young patients: Differential diagnoses
Psychiatric disorders that are usually defined by psychotic features, including:
|
Psychiatric disorders that commonly do not include hallucinations but in which hallucinations can occur as comorbid or associated symptoms, such as:
|
Prodromal and at-risk clinical states of psychiatric disorders (psychotic and mood disorders) |
Medications (steroids, anticholinergics, stimulants) |
Drug intoxication and abuse
|
Organic or nonpsychiatric disorders
|
Metabolic disorders
|
Source: a. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625. b. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380. c. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581. d. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002. e. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34. f. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785. g. Sosland MD, Edelsohn GA. Hallucinations in children and adolescents. Curr Psychiatry Rep. 2005;7:180-188. |
Table 4
Diagnostic considerations when assessing a hallucinating child
General: |
Hallucinations are a symptom, not a diagnosis |
They can have a developmental, neurologic, metabolic, or psychiatric basis |
Visual, gustatory, and olfactory hallucinations suggest a medical- or substance-related origin |
Schizophrenia is rare before age 11 |
In pervasive developmental disorders, schizophrenia is diagnosed only if prominent delusions and hallucinations are present for at least 1 month |
Hallucinations are not uncommon in major depressive disorder but may be associated with higher risk of bipolar disorder |
During interviewing, remember that children: |
are highly suggestible |
may answer questions in the affirmative to get attention or to please the interviewer |
may not fully or partially understand what is being asked |
may blame their misbehavior on voices to escape punishment |
may not distinguish between night terrors and illusions |
Source:References 25-28 |
Psychotic disorder not otherwise specified
(NOS) in children often is overused and misused. One reason could be that DSM-IV-TR does not have a category for hallucinations in nonpsychotic children or for children who are at risk for psychosis. However, recommendations regarding the diagnosis of psychotic disorder NOS note it:
- may be used if uncertainty persists after ruling out all other diagnoses
- should be avoided when hallucinations occur in nonpsychotic children
- should not be used longitudinally if a clinician later determines a specific disorder accounts for the hallucinations.29
Treatment
Addressing underlying medical or psychiatric illness, including substance use, is primary. Some adolescents or children may think that cannabis use is relatively benign. Discuss the risks of cannabis use in an age-appropriate manner.
In the ED. Children or adolescents who present with hallucinations in the ED should undergo a thorough evaluation that explores the differential diagnoses. Suggestions for evaluating patients in this setting appear in Table 5.21
Prodromal or at-risk children. There is no consensus on how early to initiate treatment for children in the prodromal stages of psychosis. Early identification and treatment is imperative because duration of untreated psychosis (DUP) is a primary predictor of treatment response in first-admission patients, and longer DUP corresponds to poorer prognosis in children.30
Assessment scales for early identification of psychosis have limitations because most are not standardized for use in children age <14. To assess symptoms and predict future psychosis in children consider using:
- Scale of Prodromal Symptoms
- Structured Interview for Prodromal Symptoms
- Comprehensive Assessment of At-Risk Mental States
- Bonn Scale for the Assessment of Basic Symptoms.
A hallucinating child may be considered prodromal if he or she has:
- 30% drop in Global Assessment Functioning score in the past month
- a first-degree relative with affective or nonaffective psychotic disorder or schizotypal personality disorder.31
Antipsychotics. When treating children, use antipsychotics with caution and consider short- and long-term risks and benefits. Early treatment is indicated when hallucinations are among a patient’s psychotic symptoms; however, antipsychotic use for children in the prodromal phase lacks consensus. McGlashan et al32 showed that in 60 high-risk patients (mean age 16), olanzapine, 5 to 15 mg/d, reduced conversion to psychosis by 50% over 6 months. McGorry et al33 observed that in 59 ultra-high risk patients (mean age 20), adding low-dose risperidone (1 to 2 mg/d) and cognitive-behavioral therapy (CBT) was superior to case management and supportive psychotherapy in preventing psychosis after 6 months of treatment, but this difference was not maintained at 6 months of follow-up.
CBT slows progression to psychosis in ultra-high risk patients and reduces positive symptoms more specifically than it improves emotional dysfunction.34 CBT for psychosis is based on the concept that auditory hallucinations have an underlying personalized meaning or cognitive schema.35 The initial goal of treatment is to engage the child and understand:
- What do the hallucinations mean to the patient?
- How did they start?
- Can the child start or stop them?
- What does the patient think they are?
The clinician then strives to help the child identify alternative explanations for these hallucinations and develop coping strategies.36 “Normalizing” the voices helps the child attribute these voices to a less anxiety-provoking cause.37 Consider suggesting common reasons for the hallucinations, such as:
- lack of sleep
- isolation
- dehydration
- extreme stress
- strong thoughts (obsessions)
- fever and illness
- lack of food
- drugs and alcohol.
If your patient learns that any of these reasons could explain the hallucinations, he or she may start to have a less delusional understanding of them. Suggest that the voices are “real” only if your patient believes it.
Help children develop coping strategies to control auditory hallucinations such as:
- humming
- listening to music
- reading (forwards and backwards)
- talking to others
- exercising
- singing
- medication
- ignoring the voices.
With normalization and other coping strategies, children with visual hallucinations can learn to transform in their mind the frightful image to a funnier one, which is less anxiety-provoking and gives them a sense of control.
Table 5
Suggestions for evaluating hallucinating children in the ED
Evaluate risk factors for suicidality |
Rule out medical and neurologic causes, including substance abuse/intoxication |
Identify underlying psychopathological, psychosocial, and cultural factors |
Contact key adult informants for collateral information |
Decide if hallucinations are psychotic or nonpsychotic |
Reassure your patients that hearing voices does not mean that they are ‘crazy’ |
Evaluate hallucinations in the context of other features of psychoses (onset, frequency, severity, and chronicity) |
Initiate psychotherapy and antidepressants if needed for patients with underlying depression, anxiety, or PTSD |
ED: emergency department; PTSD: posttraumatic stress disorder Source: Reference 21 |
Related Resources
- Bartels-Velthuis AA, Jenner JA, van de Willige G, et al. Prevalence and correlates of auditory vocal hallucinations in middle childhood. Br J Psychiatry. 2010;196(1):41-46.
- Cepeda C. Psychotic symptoms in children and adolescents: assessment, differential diagnosis, and treatment. New York, NY: Routledge; 2007.
Drug Brand Names
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Risperidone • Risperdal
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/10/hallucinations-in-children-diagnostic.html#comments
Hallucinations in children are of grave concern to parents and clinicians, but aren’t necessarily a symptom of mental illness. In adults, hallucinations usually are linked to serious psychopathology; however, in children they are not uncommon and may be part of normal development (Box).
A hallucination is a false auditory, visual, gustatory, tactile, or olfactory perception not associated with real external stimuli.1 It must be differentiated from similar phenomenon such as illusions (misperception of actual stimuli), elaborate fantasies, imaginary companions, and eidetic images (visual images stored in memory).
Although hallucinations frequently are considered synonymous with psychotic disorders, in children this rare. Neurobiologic studies (fMRI) of adults show activation of Broca’s area (left inferior frontal gyrus) seconds before patients perceive auditory verbal hallucinations, which suggests that auditory hallucinations may be misidentified self-talk.a,b According to Piaget,c children age <7 may have difficulty distinguishing between events occurring while dreaming and awake. He further theorized that nonpathologic hallucinations could become pathologic when combined with trauma such as abuse. Straussd suggested that psychosis might lie on a continuum with normal phenomenon. In a case series, Wilking and Paulie described how developmental difficulties, deprivation, sociocultural conditions, and family relationships could contribute to impaired reality testing.
Imaginary friends or companions are common among all children. Children who have imaginary friends are more likely to report hearing “voices.”f Imaginary friends:
- appear, function, and disappear at the wish of the child
- pose no threat and often are a source of comfort
- often can be described in detail
- are not ego-dystonic.g
Also, children with imaginary friends will not show evidence of a thought disorder.
Source:
a. Shergill SS, Brammer MJ, Amaro E, et al. Temporal course of auditory hallucinations. Br J Psychiatry. 2004;185:516-517.
b. Shergill SS, Brammer JJ, Williams SC, et al. Mapping auditory hallucinations in schizophrenia; using functional magnetic resonance imaging. Arch Gen Psychiatry. 2000;57;1033-1038.
c. Piaget J. The child’s conception of the world. London, United Kingdom: Routledge and Kegan Paul; 1929.
d. Strauss JS. Hallucinations and delusions as points on continua function. Rating scale evidence. Arch Gen Psychiatry. 1969;21:581-586.
e. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
f. Pearson D, Burrow A, FitzGerald C, et al. Auditory hallucinations in normal child populations. Pers Individ Dif. 2001;31:401-407.
g. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002.
Common, yet a cause for concern
Epidemiologic studies show 2.8% of adults report hallucinations before age 21.2 Nonpsychotic children as young as age 5 have reported hallucinations.3 Hallucinatory phenomenon may be present in 8% to 21% of all 11-year-old children; two-thirds of these patients have no DSM-IV-TR diagnosis.4,5 However, 1 evaluation of 62 nonpsychotic hallucinating children treated in a psychiatric emergency department (ED):
- 34% had depression
- 22% had attention-deficit/hyperactivity disorder (ADHD)
- 21% had disruptive behavior disorders
- 23% had other diagnoses.6
Studies suggest that children who have hallucinations but no other psychotic symptoms have a better long-term prognosis than those with additional psychotic symptoms.7 A 17-year longitudinal study of children with hallucinations and concurrent emotional and conduct problems found:
- up to 50% of patients still experience hallucinations at age 30
- hallucinations did not significantly predict clinical outcome at age 30
- childhood hallucinations did not increase the risk for psychosis, depression, organic brain disorder, or other psychiatric illnesses.7
In a study of children with psychosis and disruptive disorders, at 2- to 8-years follow-up 50% met criteria for major depressive disorder, bipolar disorder, or schizophreniform disorders.8 In a 15-year longitudinal study of 11-year-olds, self-reported psychotic symptoms—such as delusional beliefs and hallucinatory experiences—predicted a high risk of schizophreniform disorder at age 26.9 These studies suggest that experiencing significant disruptions in thoughts and perceptions during childhood may be related to later development of prominent mood and thought disorders.
Differential diagnosis
Table 1 lists possible causes of hallucination in children.6,10-13 Hallucinations during childhood can occur in the context of several psychiatric disorders, including:
They can also manifest as comorbid or associated symptoms of disorders not commonly associated with hallucinations, such as ADHD, disruptive disorders, anxiety disorders, and prodromal clinical states. Medications, substance use, and organic and metabolic disorders also must be considered in the differential diagnosis (Table 3).
Hallucinations may occur in low-functioning or anxious children, in the context of psychosocial adversity or abuse, and during bereavement of a deceased parent when the surviving parent is emotionally unavailable.11-13,15-17 Rule out hypnagogic and hypnopompic hallucinations, which are predominantly visual hallucinations that occur immediately before falling asleep and during the transition from sleep to wakefulness, respectively.18 Rarely, a child who has had hallucinations for some time may learn to complain of them when he or she is not hallucinating in order to obtain a primary or secondary gain, such as getting attention from caregivers.
Little is known about psychosis and hallucinations in preschoolers (age ≤5); therefore, their language use may help assessment. Because of cognitive immaturity, children often use illogical thinking and loose association and may describe their thoughts as “voices.” This is common in children with language disorders—and sometimes in healthy patients—who may talk about voices because they cannot describe their own thoughts.
Children with ADHD and/or oppositional defiant disorder often are impulsive and show poor judgment and may blame voices for telling them to do bad things. These “hallucinations” may represent internal thoughts battling with the child’s conscience.6 Auditory and visual hallucinations have been reported in children with Tourette syndrome, especially when associated with ADHD or obsessive-compulsive disorder.19
Medical causes. Electrolyte disturbances, metabolic disorders, fever, and serious infections are common nonpsychiatric causes of hallucinations.20 Brain neoplasm—particularly in visual association areas, the temporal lobe, or portions of the optic nerve or retina—also may produce hallucinations, which can be complex with full images.21
Medications such as steroids and anticholinergics may cause hallucinations. Case studies report visual and tactile hallucinations with methylphenidate therapy that resolve after discontinuing the medication.22 Illicit substances, including cannabis, lysergic acid diethylamide (LSD), cocaine, amphetamines, 3,4-methylenedioxymeth-amphetamine (ecstasy), opiates, and barbiturates, can induce hallucinations.
Suspect substance-induced hallucinations if your patient shows:
- acute onset of hallucinations
- dilated pupils
- extreme agitation or drowsiness
- other signs of intoxication.
Hallucinations caused by seizure disorders are rare but can be somatosensory, visual (occipital lobe focus), auditory, olfactory (uncinate, complex partial), or gustatory. The hallucinations may be unformed (flashing lights or rushing noises) or formed (images, spoken words, or music) and could be part of the aura arising from the temporal lobe (dreamlike, flashbacks). Command hallucinations are rare and adult and pediatric patients usually sense they are not real.23
Migraines occur in approximately 5% of prepubertal children and often are comorbid with affective and anxiety disorders.24 Hallucinations associated with migraine commonly are visual, but gustatory, olfactory, and auditory hallucinations also can occur, with or without headaches.3 Any hallucination associated with headaches should be investigated neurologically. Other diagnostic aspects of hallucinations to consider while interviewing children are listed in Table 4.25-28
Table 1
Possible causes of hallucinations in children and adolescents
Normal development |
Nonpsychotic psychopathology |
Psychosocial adversity |
Psychotic illness |
Stress |
Family dysfunction |
Deprivation |
Developmental difficulties |
Sociocultural interaction (immigration) |
Poorly differentiated male and female family roles |
Presence or absence of different mother figures |
Cultural factors (witches, ghosts, spiritualism) |
Hallucination of deceased parent, when unresolved mourning persists in the surviving parent |
Source: References 6,10-13 |
Table 2
Content of hallucinations may point to their cause
Schizophrenia or other psychotic disorders | May hear several voices making a critical commentary Command hallucinations telling patients to harm themselves or others Bizarre voices like ‘a computer in my head’ or aliens Voices of someone familiar or a ‘relative’ Visual hallucinations of devils, scary faces, space creatures, and skeletons |
Depressive disorders | Usually a single voice speaking from outside the patient’s head with derogatory or suicidal content |
Bipolar disorder | Usually involves grandiose ideas about power, worth, knowledge, family, or relationship |
Bereavement | Usually a transient (visual or auditory) perception of the deceased person |
Posttraumatic stress disorder | Transient visual hallucinations, usually with phobic content |
Source: Reference 11 |
Table 3
Hallucinations in young patients: Differential diagnoses
Psychiatric disorders that are usually defined by psychotic features, including:
|
Psychiatric disorders that commonly do not include hallucinations but in which hallucinations can occur as comorbid or associated symptoms, such as:
|
Prodromal and at-risk clinical states of psychiatric disorders (psychotic and mood disorders) |
Medications (steroids, anticholinergics, stimulants) |
Drug intoxication and abuse
|
Organic or nonpsychiatric disorders
|
Metabolic disorders
|
Source: a. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625. b. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380. c. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581. d. Lewis M. Child and adolescent psychiatry: a comprehensive textbook. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2002. e. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34. f. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785. g. Sosland MD, Edelsohn GA. Hallucinations in children and adolescents. Curr Psychiatry Rep. 2005;7:180-188. |
Table 4
Diagnostic considerations when assessing a hallucinating child
General: |
Hallucinations are a symptom, not a diagnosis |
They can have a developmental, neurologic, metabolic, or psychiatric basis |
Visual, gustatory, and olfactory hallucinations suggest a medical- or substance-related origin |
Schizophrenia is rare before age 11 |
In pervasive developmental disorders, schizophrenia is diagnosed only if prominent delusions and hallucinations are present for at least 1 month |
Hallucinations are not uncommon in major depressive disorder but may be associated with higher risk of bipolar disorder |
During interviewing, remember that children: |
are highly suggestible |
may answer questions in the affirmative to get attention or to please the interviewer |
may not fully or partially understand what is being asked |
may blame their misbehavior on voices to escape punishment |
may not distinguish between night terrors and illusions |
Source:References 25-28 |
Psychotic disorder not otherwise specified
(NOS) in children often is overused and misused. One reason could be that DSM-IV-TR does not have a category for hallucinations in nonpsychotic children or for children who are at risk for psychosis. However, recommendations regarding the diagnosis of psychotic disorder NOS note it:
- may be used if uncertainty persists after ruling out all other diagnoses
- should be avoided when hallucinations occur in nonpsychotic children
- should not be used longitudinally if a clinician later determines a specific disorder accounts for the hallucinations.29
Treatment
Addressing underlying medical or psychiatric illness, including substance use, is primary. Some adolescents or children may think that cannabis use is relatively benign. Discuss the risks of cannabis use in an age-appropriate manner.
In the ED. Children or adolescents who present with hallucinations in the ED should undergo a thorough evaluation that explores the differential diagnoses. Suggestions for evaluating patients in this setting appear in Table 5.21
Prodromal or at-risk children. There is no consensus on how early to initiate treatment for children in the prodromal stages of psychosis. Early identification and treatment is imperative because duration of untreated psychosis (DUP) is a primary predictor of treatment response in first-admission patients, and longer DUP corresponds to poorer prognosis in children.30
Assessment scales for early identification of psychosis have limitations because most are not standardized for use in children age <14. To assess symptoms and predict future psychosis in children consider using:
- Scale of Prodromal Symptoms
- Structured Interview for Prodromal Symptoms
- Comprehensive Assessment of At-Risk Mental States
- Bonn Scale for the Assessment of Basic Symptoms.
A hallucinating child may be considered prodromal if he or she has:
- 30% drop in Global Assessment Functioning score in the past month
- a first-degree relative with affective or nonaffective psychotic disorder or schizotypal personality disorder.31
Antipsychotics. When treating children, use antipsychotics with caution and consider short- and long-term risks and benefits. Early treatment is indicated when hallucinations are among a patient’s psychotic symptoms; however, antipsychotic use for children in the prodromal phase lacks consensus. McGlashan et al32 showed that in 60 high-risk patients (mean age 16), olanzapine, 5 to 15 mg/d, reduced conversion to psychosis by 50% over 6 months. McGorry et al33 observed that in 59 ultra-high risk patients (mean age 20), adding low-dose risperidone (1 to 2 mg/d) and cognitive-behavioral therapy (CBT) was superior to case management and supportive psychotherapy in preventing psychosis after 6 months of treatment, but this difference was not maintained at 6 months of follow-up.
CBT slows progression to psychosis in ultra-high risk patients and reduces positive symptoms more specifically than it improves emotional dysfunction.34 CBT for psychosis is based on the concept that auditory hallucinations have an underlying personalized meaning or cognitive schema.35 The initial goal of treatment is to engage the child and understand:
- What do the hallucinations mean to the patient?
- How did they start?
- Can the child start or stop them?
- What does the patient think they are?
The clinician then strives to help the child identify alternative explanations for these hallucinations and develop coping strategies.36 “Normalizing” the voices helps the child attribute these voices to a less anxiety-provoking cause.37 Consider suggesting common reasons for the hallucinations, such as:
- lack of sleep
- isolation
- dehydration
- extreme stress
- strong thoughts (obsessions)
- fever and illness
- lack of food
- drugs and alcohol.
If your patient learns that any of these reasons could explain the hallucinations, he or she may start to have a less delusional understanding of them. Suggest that the voices are “real” only if your patient believes it.
Help children develop coping strategies to control auditory hallucinations such as:
- humming
- listening to music
- reading (forwards and backwards)
- talking to others
- exercising
- singing
- medication
- ignoring the voices.
With normalization and other coping strategies, children with visual hallucinations can learn to transform in their mind the frightful image to a funnier one, which is less anxiety-provoking and gives them a sense of control.
Table 5
Suggestions for evaluating hallucinating children in the ED
Evaluate risk factors for suicidality |
Rule out medical and neurologic causes, including substance abuse/intoxication |
Identify underlying psychopathological, psychosocial, and cultural factors |
Contact key adult informants for collateral information |
Decide if hallucinations are psychotic or nonpsychotic |
Reassure your patients that hearing voices does not mean that they are ‘crazy’ |
Evaluate hallucinations in the context of other features of psychoses (onset, frequency, severity, and chronicity) |
Initiate psychotherapy and antidepressants if needed for patients with underlying depression, anxiety, or PTSD |
ED: emergency department; PTSD: posttraumatic stress disorder Source: Reference 21 |
Related Resources
- Bartels-Velthuis AA, Jenner JA, van de Willige G, et al. Prevalence and correlates of auditory vocal hallucinations in middle childhood. Br J Psychiatry. 2010;196(1):41-46.
- Cepeda C. Psychotic symptoms in children and adolescents: assessment, differential diagnosis, and treatment. New York, NY: Routledge; 2007.
Drug Brand Names
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Risperidone • Risperdal
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Sadock BJ, Sadock VA. Kaplan and Sadock’s concise textbook of clinical psychiatry. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2008:26.
2. Fenning S, Susser ES, Pilowsky DJ, et al. Childhood hallucinations preceding the first psychotic episode. J Nerv Ment Dis. 1997;185:115-117.
3. Schreier HA. Auditory hallucinations in nonpsychotic children with affective syndromes and migraines: report of 13 cases. J Child Neurol. 1998;13:377-382.
4. McGee R, Williams S, Poulton R. Hallucinations in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 2000;39:12-13.
5. Yoshizumi T, Murase S, Honjo S, et al. Hallucinatory experiences in a community sample of Japanese children. J Am Acad Child Adolesc Psychiatry. 2004;43:1030-1036.
6. Edelsohn GA, Rabinovich H, Portnoy R. Hallucinations in nonpsychotic children: findings from a psychiatric emergency service. Ann N Y Acad Sciences. 2003;1008:261-264.
7. Rothstein A. Hallucinatory phenomenon in childhood: a critique of the literature. J Am Acad Child Psychiatry. 1981;20:623-635.
8. Nicholson R, Lenane M, Brookner F, et al. Children and adolescents with psychotic disorder not otherwise specified: a 2- to 8-year follow-up study. Compr Psychiatry. 2001;42:319-325.
9. Poulton R, Caspi A, Moffitt TE, et al. Children’s self reported psychotic symptoms and adult schizophreniform disorder: a 15-year longitudinal study. Arch Gen Psychiatry. 2000;57:1053-1058.
10. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
11. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625.
12. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380.
13. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581.
14. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry. 9th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2003:1276-1277,1283.
15. Famularo R, Kinscherff R, Fenton T. Psychiatric diagnosis of maltreated children: preliminary findings. J Am Acad Child Adolesc Psychiatry. 1992;31:863-867.
16. Putnam FW, Peterson G. Further validation of the child dissociative checklist. Dissociation. 1994;7:204-211.
17. Kaufman J, Birmaher B, Clayton S, et al. Case study: trauma-related hallucinations. J Am Acad Child Adolesc Psychiatry. 1997;36(11):1602-1605.
18. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 8th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2005.
19. Kerbeshian J, Burd L. Are schizophreniform symptoms present in attenuated form in children with Tourette Disorder and other developmental disorders? Can J Psychiatry. 1987;32(2):123-135.
20. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34.
21. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785.
22. Gross-Tsur V, Joseph A, Shalev RS. Hallucinations during methylphenidate therapy. Neurology. 2004;63:753-754.
23. Wyllie E. The treatment of epilepsy: principles and practices, Philadelphia, PA: Lippincott Williams and Wilkins; 1993.
24. Saeed MA, Pumariega AJ, Cinciripini PM. Psychopharmacological management of migraine in children in children and adolescents. J Child Adolesc Psychopharmacol. 1992;2:199-211.
25. Chambers WJ, Puig-Antich J, Tabrizi MA, et al. Psychotic symptoms in prepubertal major depressive disorder. Arch Gen Psychiatry. 1982;39:921-927.
26. Volkmar FR. Childhood and adolescent psychosis: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 1996;35:843-851.
27. First MB, Frances A, Pincus HA. DSM-IV handbook of differential diagnosis. Washington, DC: American Psychiatric Press; 1995.
28. Birmaher B, Ryan ND, Williamson DE, et al. Childhood and adolescent depression: a review of the past 10 years. Part I. J Am Acad Child Adolesc Psychiatry. 1996;35:1427-1439.
29. Meyer SE, Bearden CE, Lux CR, et al. The psychosis prodrome in adolescent patients viewed through the lens of DSM-IV. J Child Adolesc Psychopharmacol. 2005;15(3):434-451.
30. Drake RJ, Haley CJ, Akhtar S, et al. Causes and consequences of duration of untreated psychosis in schizophrenia. Br J Psychiatry. 2000;177:511-515.
31. McGorry PD, Yung AR, Phillips LJ. The “close-in” or ultra high-risk model: a safe and effective strategy for research and clinical intervention in prepsychotic mental disorder. Schizophr Bull. 2003;29(4):771-790.
32. McGlashan TH, Zipursky RB, Perkins DO, et al. Olanzapine versus placebo treatment of schizophrenia prodrome: one year results. Poster presented at: International Congress of Schizophrenia Research, Colorado Springs, CO, April-May 2003.
33. McGorry PD, Yung AR, Phillips LJ, et al. Randomized controlled trial of interventions designed to reduce the risk of progression to first episode psychosis in a clinical sample with subthreshold symptoms. Arch Gen Psychiatry. 2002;59:921-928.
34. French P, Shryane N, Bentall RP, et al. Effects of cognitive therapy on the longitudinal development of psychotic experiences in people at high risk of developing psychosis. Br J Psychiatry Suppl. 2007;51:s82-87.
35. Turkington D, Siddle R. Cognitive therapy for the treatment of delusions. Advances in Psychiatric Treatment. 1998;4:235-242.
36. Sosland MD, Pinninti N. Five ways to quiet auditory hallucinations. Current Psychiatry. 2005;4:40.-
37. Kingdon DG, Turkington D. Cognitive behavioral therapy of schizophrenia, New York, NY: Guilford Press; 1994.
1. Sadock BJ, Sadock VA. Kaplan and Sadock’s concise textbook of clinical psychiatry. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2008:26.
2. Fenning S, Susser ES, Pilowsky DJ, et al. Childhood hallucinations preceding the first psychotic episode. J Nerv Ment Dis. 1997;185:115-117.
3. Schreier HA. Auditory hallucinations in nonpsychotic children with affective syndromes and migraines: report of 13 cases. J Child Neurol. 1998;13:377-382.
4. McGee R, Williams S, Poulton R. Hallucinations in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 2000;39:12-13.
5. Yoshizumi T, Murase S, Honjo S, et al. Hallucinatory experiences in a community sample of Japanese children. J Am Acad Child Adolesc Psychiatry. 2004;43:1030-1036.
6. Edelsohn GA, Rabinovich H, Portnoy R. Hallucinations in nonpsychotic children: findings from a psychiatric emergency service. Ann N Y Acad Sciences. 2003;1008:261-264.
7. Rothstein A. Hallucinatory phenomenon in childhood: a critique of the literature. J Am Acad Child Psychiatry. 1981;20:623-635.
8. Nicholson R, Lenane M, Brookner F, et al. Children and adolescents with psychotic disorder not otherwise specified: a 2- to 8-year follow-up study. Compr Psychiatry. 2001;42:319-325.
9. Poulton R, Caspi A, Moffitt TE, et al. Children’s self reported psychotic symptoms and adult schizophreniform disorder: a 15-year longitudinal study. Arch Gen Psychiatry. 2000;57:1053-1058.
10. Wilking VN, Paoli C. The hallucinatory experience: an attempt at a psychodynamic classification and reconsideration of its diagnostic significance. J Am Acad Child Psychiatry. 1966;5:431-440.
11. Schreier HA. Hallucinations in nonpsychotic children: more common than we think? J Am Acad Child Adolesc Psychiatry. 1999;38(5):623-625.
12. Kotsopoulos S, Konigsberg J, Cote A, et al. Hallucinatory experiences in nonpsychotic children. J Am Acad Child Adolesc Psychiatry. 1987;26:375-380.
13. Yates TT, Bannard JR. The “haunted” child: grief, hallucinations and family dynamics. J Am Acad Child Adolesc Psychiatry. 1988;27:573-581.
14. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry. 9th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2003:1276-1277,1283.
15. Famularo R, Kinscherff R, Fenton T. Psychiatric diagnosis of maltreated children: preliminary findings. J Am Acad Child Adolesc Psychiatry. 1992;31:863-867.
16. Putnam FW, Peterson G. Further validation of the child dissociative checklist. Dissociation. 1994;7:204-211.
17. Kaufman J, Birmaher B, Clayton S, et al. Case study: trauma-related hallucinations. J Am Acad Child Adolesc Psychiatry. 1997;36(11):1602-1605.
18. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 8th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2005.
19. Kerbeshian J, Burd L. Are schizophreniform symptoms present in attenuated form in children with Tourette Disorder and other developmental disorders? Can J Psychiatry. 1987;32(2):123-135.
20. Pao M, Lohman C, Gracey D, et al. Visual, tactile, and phobic hallucinations: recognition and management in the emergency department. Pediatr Emerg Care. 2004;20:30-34.
21. Edelsohn GA. Hallucinations in children and adolescents: considerations in the emergency setting. Am J Psychiatry. 2006;163(5):781-785.
22. Gross-Tsur V, Joseph A, Shalev RS. Hallucinations during methylphenidate therapy. Neurology. 2004;63:753-754.
23. Wyllie E. The treatment of epilepsy: principles and practices, Philadelphia, PA: Lippincott Williams and Wilkins; 1993.
24. Saeed MA, Pumariega AJ, Cinciripini PM. Psychopharmacological management of migraine in children in children and adolescents. J Child Adolesc Psychopharmacol. 1992;2:199-211.
25. Chambers WJ, Puig-Antich J, Tabrizi MA, et al. Psychotic symptoms in prepubertal major depressive disorder. Arch Gen Psychiatry. 1982;39:921-927.
26. Volkmar FR. Childhood and adolescent psychosis: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 1996;35:843-851.
27. First MB, Frances A, Pincus HA. DSM-IV handbook of differential diagnosis. Washington, DC: American Psychiatric Press; 1995.
28. Birmaher B, Ryan ND, Williamson DE, et al. Childhood and adolescent depression: a review of the past 10 years. Part I. J Am Acad Child Adolesc Psychiatry. 1996;35:1427-1439.
29. Meyer SE, Bearden CE, Lux CR, et al. The psychosis prodrome in adolescent patients viewed through the lens of DSM-IV. J Child Adolesc Psychopharmacol. 2005;15(3):434-451.
30. Drake RJ, Haley CJ, Akhtar S, et al. Causes and consequences of duration of untreated psychosis in schizophrenia. Br J Psychiatry. 2000;177:511-515.
31. McGorry PD, Yung AR, Phillips LJ. The “close-in” or ultra high-risk model: a safe and effective strategy for research and clinical intervention in prepsychotic mental disorder. Schizophr Bull. 2003;29(4):771-790.
32. McGlashan TH, Zipursky RB, Perkins DO, et al. Olanzapine versus placebo treatment of schizophrenia prodrome: one year results. Poster presented at: International Congress of Schizophrenia Research, Colorado Springs, CO, April-May 2003.
33. McGorry PD, Yung AR, Phillips LJ, et al. Randomized controlled trial of interventions designed to reduce the risk of progression to first episode psychosis in a clinical sample with subthreshold symptoms. Arch Gen Psychiatry. 2002;59:921-928.
34. French P, Shryane N, Bentall RP, et al. Effects of cognitive therapy on the longitudinal development of psychotic experiences in people at high risk of developing psychosis. Br J Psychiatry Suppl. 2007;51:s82-87.
35. Turkington D, Siddle R. Cognitive therapy for the treatment of delusions. Advances in Psychiatric Treatment. 1998;4:235-242.
36. Sosland MD, Pinninti N. Five ways to quiet auditory hallucinations. Current Psychiatry. 2005;4:40.-
37. Kingdon DG, Turkington D. Cognitive behavioral therapy of schizophrenia, New York, NY: Guilford Press; 1994.
CAM for your anxious patient: What the evidence says
The number of people with psychiatric disorders who use complementary and alternative medicine (CAM) is on the rise. In surveys of patients seeking psychiatric care, estimates of CAM use range from 8% to 57%; the most frequent uses are for depression and anxiety disorders. A population-based study in the United States found that 9% of respondents had anxiety attacks and 57% of these individuals had used CAM.1 Similarly, in a Finnish population-based study (N=5,987) 35% of subjects reported some form of CAM use in the previous year; those with comorbid anxiety and depressive disorders used CAM most frequently.2
Unfortunately, a MEDLINE search shows that the number of studies examining psychotropic medications dwarfs the number of studies on even the most common CAM treatments used for psychiatric disorders. Far more patients with diagnosed mental disorders are studied in trials of standard treatments than CAM treatments. Because very few studies evaluate the cost-effectiveness of CAM treatments for psychiatric disorders, the risk-to-benefit ratio is difficult to calculate. Although several CAM treatments for depressive disorders have enough support to be considered options,3 CAM options for anxiety disorders are fewer and have less evidence of efficacy.
For these reasons, it is hard to recommend any CAM treatment as first line. Despite the relative lack of high quality research on CAM treatment outcomes, high rates of CAM use make it critical for clinicians to understand what treatments are available—or at least which treatments should be favored if patients are intent on trying them. We review the current research for yoga, exercise, bibliotherapy, and the dietary supplements kava and inositol for treating anxiety disorders and suggest those that warrant consideration for patients who do not respond, respond partially, or suffer from side effects from selective serotonin reuptake inhibitors (SSRIs) or benzodiazepines.
Limitations of CAM research
There are several limitations to the research literature on CAM approaches for anxiety disorders.4 First, there is a wide diversity of practices considered alternative or complementary and various ways in which these methods are applied across cultures. Some authors consider complementary medicines to be only herbal remedies, whereas others include individual therapies such as acupuncture, aromatherapy, herbal therapy, homeopathy, iridology, naturopathy, and reflexology.5 This article defines “alternative” treatments as those other than a form of psychotherapy or an FDA-approved medication that substitute for standard psychiatric treatment, and “complementary” approaches as those used to augment standard psychiatric treatments.
Anxiety and stress are ubiquitous, perhaps motivating interest in CAM options and prompting research on heterogeneous groups of individuals with poorly defined clinical syndromes or with isolated symptoms of anxiety or subjective distress. Few studies examine well-defined patient groups with diagnosed anxiety disorders. There are also multiple research design problems, including poorly specified treatments, poorly chosen placebos, and interpreting nonsignificant differences from established treatments as equivalence in underpowered studies.
The CAM treatments reviewed in this article have ≥2 randomized controlled trials (RCTs) that support their use for patients with diagnosed anxiety disorders, and ≥1 study that shows that the treatment can induce remission.
Yoga
In 2005 Kirkwood et al carried out the first systematic review of research evidence for the effectiveness of yoga in anxiety treatment.6 Of 19 studies identified, 4 RCTs and 1 nonrandomized trial met their inclusion criteria, which were an anxiety disorder diagnosis, use of yoga or yoga-based exercises alone, and anxiety rating scales used as outcome measures. Most found significant improvement in anxiety symptoms with yoga compared with placebo. Details of the 5 trials evaluated in Kirkwood’s review are summarized in Table 1.7-11
Since the 2005 review, 3 additional studies of yoga and anxiety have been published, but none would meet Kirkwood’s inclusion criteria. One that evaluated a heterogeneous group of patients using an intervention with multiple components—only 1 of which was yoga—found the intervention significantly reduced anxiety scores.12 A second study comparing yoga with relaxation in 131 patients with mild-to-moderate stress but no anxiety disorder diagnosis showed yoga was as effective as relaxation in improving anxiety symptoms as measured by the anxiety subscale of the State Trait Personality Inventory.13 In a study of 183 nonrandomized survivors of the 2004 southeast Asia tsunami with posttraumatic stress disorder (PTSD) symptoms, yoga-based breathing either alone or paired with trauma reduction exposure techniques significantly reduced PTSD symptoms compared with wait-list controls.14
Conclusion. Few controlled studies evaluated yoga for anxiety disorders, and all have significant methodologic limitations and/or poor methodology reporting. The diagnostic conditions treated and both yoga interventions and control conditions varied. However, these limited results are encouraging, particularly for treatment of obsessive-compulsive disorder (OCD). There is little information regarding safety or contraindications of yoga. Reported attrition rates were high in most studies, which may raise concerns about patient motivation and compliance.
Table 1
Evidence on the effectiveness of yoga for anxiety disorders
Study | Design | Results |
---|---|---|
Vahia et al, 19737 | 36 patients with psychoneurosis randomly assigned to yoga (N=15) or a control intervention of relaxation, postures, breathing, and writing (N=12) | Significant difference between groups in TAS scores after but not before treatment. Reduction in mean TAS score for yoga group but not control group |
Vahia et al,19738 | 39 patients received 6 weeks of yoga (N=21) or medication (amitriptyline and chlordiazepoxide on a variable dosage schedule) (N=18) | Yoga showed significantly greater reductions in TAS in this non-randomized sample |
Sahasi et al, 19899 | 91 patients randomly assigned to yoga practiced daily for 40 minutes (N=38) or diazepam at unspecified frequency or doses (N=53) for 3 months | Mean reduction in IPAT with yoga (3.39, P < .05) vs control group (0.36, P > .05). Attrition rate was 21.1% in yoga group and 66% in controls |
Sharma et al, 199110 | 71 patients with anxiety neurosis randomly assigned to 1-week yoga training, then daily practice (N=41) or control (N=30, placebo capsule) | HAM-A measured at 3 weekly intervals for 12 weeks. Significant between group mean difference at 3 weeks (greater improvement in yoga group compared with controls). Significant improvement in yoga group between 3 and 6 weeks but not for controls |
Shannahoff-Khalsa et al, 199911 | 21 OCD patients randomly assigned to kundalini yoga (N=11) or relaxation and mindfulness meditation (N=10). Multiple outcome measures; Y-BOCS was primary | Seven in each group completed 3 months; patients who practiced yoga demonstrated greater improvements on Y-BOCS. Intent-to-treat analysis (Y-BOCS) for the baseline and 3-month tests showed that only the yoga group improved. Groups were merged for an additional year of yoga; at 15 months, the final group (N=11) improved 71% on the Y-BOCS |
HAM-A: Hamilton Anxiety Rating scale; IPAT: Institute for Personality and Ability Testing, Anxiety Scale; OCD: obsessive-compulsive disorder; TAS: Taylor’s Anxiety Scale; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Exercise
The literature examining the relationship between exercise and depression is extensive, but much less has been published about exercise in patients with anxiety disorders (Table 2).15-17 In a 10-week trial, Broocks and colleagues compared clomipramine, exercise (running), and placebo in 46 outpatients with panic disorder.15 Both exercise and clomipramine, 112.5 mg/d, significantly reduced panic symptoms compared with placebo, but clomipramine was more effective and faster-acting.
A more recent RCT compared group cognitive-behavioral therapy (GCBT) plus a home-based walking program vs GCBT and in 21 patients with panic disorder, generalized anxiety disorder (GAD), or social phobia.16 Compared with GCBT plus educational sessions, GCBT plus walking had a significant effect on self-reported depression, anxiety, and stress. Results differed by diagnosis; the most marked effects occurred in individuals with social phobia, whereas benefits for those with panic disorder or GAD were questionable.
Fifteen patients with OCD were recruited to participate in a 12-week, moderate-intensity aerobic exercise program added to their standard behavioral and/or pharmacologic treatment.17 Subjects demonstrated improvement in negative mood, anxiety, obsessions, and compulsions after each exercise session. Changes after each session persisted over the 12-week intervention, although the magnitude attenuated over the duration of the intervention.
Conclusion. Although initial results from small trials suggest exercise may help improve anxiety symptoms, further studies are needed to determine how to best use exercise training to treat anxious patients, specifically regarding dose-response relationship, differences in effectiveness between aerobic and resistance training, and the mechanisms by which exercise improves psychiatric symptoms.
Table 2
Exercise for anxiety: More research is needed
Study | Design | Results |
---|---|---|
Broocks et al, 199915 | 46 patients with panic disorder randomly assigned to 10 weeks of running, clomipramine, or wplacebo pills | Both exercise and clomipramine resulted in significant decreases in symptoms but clomipramine improved symptoms earlier and more effectively |
Merom et al, 200816 | 21 patients with panic disorder, GAD, or social phobia randomly assigned to GCBT and either a home-based walking program or educational sessions | GCBT plus walking had a significant effect on depression, anxiety, and stress compared with GCBT plus educational sessions |
Abrantes et al, 200917 | 15 patients with OCD assigned to a 12-week exercise intervention that was added to their standard behavioral and/or pharmacologic treatment | Subjects reported improved mood, anxiety, obsessions, and compulsions after each exercise session |
GAD: generalized anxiety disorder; GCBT: group cognitive-behavioral therapy |
Bibliotherapy
Investigation of bibliotherapy for treatment of anxiety disorders has been limited (Table 3).18-20 A 2009 RCT demonstrated that for 21 patients with mild-to-moderate social phobia, bibliotherapy—in the form of an 8-week self-directed CBT program with minimal therapist involvement—was superior to a wait-list control and induced clinically significant change in approximately one-third of patients.20
Rapee et al randomly assigned 267 children age 6 to 12 with anxiety disorders to bibliotherapy that consisted of parents treating their children in the home with written materials, 9 sessions of GCBT, or a wait-list control condition.19 Bibliotherapy provided by parents demonstrated benefit compared with wait-listing but was not as efficacious as GCBT at post-treatment and 3-month follow-up.
Lidren and colleagues randomly assigned 36 adult patients with panic disorder to bibliotherapy, group therapy combined with bibliotherapy, or a waitlist.18 Both treatments were more effective than wait-listing in reducing the frequency of panic attacks, severity of physical panic symptoms, catastrophic cognitions, agoraphobic avoidance, and depression. Both interventions maintained their effects at 3-and 6-month follow-up and produced clinically significant change in most patients.
Conclusion. Some preliminary evidence supports the effectiveness of bibliotherapy for social anxiety disorder, childhood anxiety disorders, and panic disorder.
Table 3
Preliminary evidence supports bibliotherapy for select anxiety disorders
Study | Design | Results |
---|---|---|
Lidren et al, 199418 | 36 adults with panic disorder randomly assigned to bibliotherapy, bibliotherapy plus group therapy, or wait-list control | Both bibliotherapy and bibliotherapy plus group therapy were more effective than wait-listing in reducing the frequency of panic attacks and severity of physical panic symptoms |
Rapee et al, 200619 | 267 children with anxiety disorders randomly assigned to bibliotherapy (parents treating their children in the home with written materials with no therapist contact), 9 sessions of group CBT, or wait-list control | Parent-delivered bibliotherapy was beneficial compared with wait-listing but was not as efficacious as group CBT |
Abramowitz et al, 200920 | 21 patients with mild-to-moderate social phobia underwent an 8-week self-directed CBT program with minimal therapist involvement | Bibliotherapy was superior to wait-listing. One-third of patients experienced clinically significant change |
CBT: cognitive-behavioral therapy |
Dietary supplements
Many dietary and herbal supplements are purported to have therapeutic efficacy for anxiety symptoms. Because of inadequate FDA regulation of manufacturing and marketing of these agents, most of these supplements have not been tested on patients with anxiety disorders.21 Limited evidence supports the use of kava for GAD and inositol for panic disorder (Table 4).22-28
Kava. Multiple double-blind RCTs found kava (Piper methysticum)—a plant indigenous to South Pacific islands—has effects greater than placebo and comparable to standard treatments for mild to moderately severe GAD. A Cochrane meta-analysis22 of 11 trials with 645 participants concluded that kava is effective for reducing GAD symptoms, with risks comparable to standard treatments for up to 6 months of use.
Case reports of kava-associated liver toxicity led to a marketing ban in Canada in 2000, followed shortly by Germany, Australia, and the United Kingdom. In 2002 the FDA issued a Consumer Advisory29 discouraging kava use. Since then a flurry of research has looked for sources of possible toxicity, including individual sensitivities,29 excessive dosing, use of toxic parts of the kava plant instead of the roots,30,31 interactions with other hepatoactive substances, and non-water based extraction methods. RCTs demonstrating kava’s efficacy and safety were characterized by careful dosing supervision, use of standardized kava extracts, and avoidance of interactions with other hepatoactive medications or CAM treatments. Doses ≤300 mg/d are recommended.22
RCTs that used the standardized acetone extract WS149023 found that women and younger adults show more positive effects from kava, and showed no liver toxicity when used for 1 to 24 weeks. A recent RCT that used kava extracts obtained via water-based methods showed kava had significant anxiolytic effects.24 However, a study of liver toxicity reports found that water-based extractions, acetonic extractions, and ethanol extractions all have been associated with toxic hepatic reactions.32 Aqueous extraction does not guarantee safety, and the extraction solvent does not cause toxicity. A recent report of a severe liver reaction to the native drink by a tourist in Samoa33 suggests that aqueous extractions from the root stock— the type of kava used by South Pacific islanders—also can be unsafe.
Conclusion. Multiple RCTs have found kava relatively safe and effective for treating anxiety symptoms. Caution is necessary, however, because of reports of liver toxicity associated with its use. Physician oversight and monitoring of kava use are appropriate.
Inositol. Evidence from RCTs suggests inositol, a natural isomer of glucose and a precursor in the phosphatidylinositol cycle, can significantly improve panic disorder symptoms.25-28 In 1 trial, efficacy and side effects were comparable to fluvoxamine.28 Effective doses ranged from 12 g/d to 18 g/d. Researchers tested inositol as monotherapy or augmentation to SSRIs for patients with mild-to-moderate OCD. In small double-blind crossover RCTs, inositol monotherapy significantly reduced Yale-Brown Obsessive Compulsive Scale scores compared with placebo26 but inositol augmentation added nothing to the effects of SSRIs.27
Conclusion. Inositol appears to be effective in improving symptoms of panic disorder. Its use for other anxiety disorders is unproven.
Table 4
Dietary supplements for anxiety disorders
Study | Design | Results |
---|---|---|
Kava | ||
Pittler et al, 200322 | Meta-analysis of 11 RCTs with a total of 645 GAD patients | Compared with placebo, kava significantly reduced anxiety as measured by total HAM-A score |
Witte et al, 200523 | Meta-analysis of 6 RCTs using kava extract WS1490 in patients with nonpsychotic anxiety disorders | Kava reduced HAM-A score more than placebo and seemed to be more effective in women and younger adults |
Sarris et al, 200924 | 60 adults with ≥1 month of elevated generalized anxiety randomly assigned to an aqueous extract of kava | Aqueous-extract kava was significantly more effective than placebo in reducing HAM-A score |
Inositol | ||
Benjamin et al, 199525 | 21 patients with panic disorder with or without agoraphobia randomly assigned to inositol, 12 g/d, or placebo | Inositol significantly reduced frequency and severity of panic attacks and severity of agoraphobia compared with placebo |
Fux et al, 199626 | 13 OCD patients randomly assigned to inositol,18 g/d, or placebo for 6 weeks | Patients taking inositol had significantly lower Y-BOCS scores compared with those receiving placebo |
Fux et al, 199927 | 10 OCD patients receiving an SSRI randomly assigned to augmentation with inositol, 18 g/d, or placebo for 6 weeks | No significant differences between treatments |
Palatnik et al, 200128 | In a crossover trial, 20 panic disorder patients completed 1 month of inositol, up to 18 g/d, and 1 month of fluvoxamine, up to 150 mg/d | Improvements in HAM-A, CGI, and agoraphobia scores were similar for both treatments |
CGI: Clinical Global Impression scale; GAD: generalized anxiety disorder; HAM-A: Hamilton Anxiety Rating scale; OCD: obsessive-compulsive disorder; RCTs: randomized controlled trials; SSRI: selective serotonin reuptake inhibitor; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Supervision is recommended
The evidence base for most CAM interventions commonly used for anxiety is relatively poor and recent systematic reviews found few methodologically rigorous studies. This has not, however, diminished CAM treatments’ popularity. Despite a paucity of high-quality studies regarding CAM for anxiety disorders, there is enough data supporting yoga, exercise, bibliotherapy, kava, and inositol to allow psychiatrists to collaborate with patients who wish to try these treatments. Advise patients that they may need physician supervision similar to that used with standard psychiatric treatments.
Related Resources
- National Center for Complementary and Alternative Medicine. http://nccam.nih.gov.
- The Journal of Alternative and Complementary Medicine. www.liebertonline.com/loi/acm.
- MedlinePlus: Complementary and Alternative Medicine. www.nlm.nih.gov/medlineplus/complementaryandalternativemedicine.html.
Drug Brand Names
- Amitriptyline • Elavil
- Chlordiazepoxide • Librium
- Clomipramine • Anafranil
- Diazepam • Valium
- Fluvoxamine • Luvox
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Kessler RC, Soukup J, Davis RB, et al. The use of complementary and alternative therapies to treat anxiety and depression in the United States. Am J Psychiatry. 2001;158(2):289-294.
2. Wahlström M, Sihvo S, Haukkala A, et al. Use of mental health services and complementary and alternative medicine in persons with common mental disorders. Acta Psychiatr Scand. 2008;118(1):73-80.
3. Saeed SA, Bloch RM, Antonacci DJ, et al. CAM for your depressed patient: 6 recommended options. Current Psychiatry. 2009;8(10):39-47.
4. Pilkington K. Searching for CAM evidence: an evaluation of therapy-specific search strategies. J Altern Complement Med. 2007;13(4):451-459.
5. van der Watt G, Laugharne J, Janca A. Complementary and alternative medicine in the treatment of anxiety and depression. Curr Opin Psychiatry. 2008;21(1):37-42.
6. Kirkwood G, Rampes H, Tuffrey V, et al. Yoga for anxiety: a systematic review of the research evidence. Br J Sports Med. 2005;39(12):884-891.
7. Vahia NS, Doongaji DR, Jeste DV, et al. Psychophysiologic therapy based on the concepts of Patanjali. A new approach to the treatment of neurotic and psychosomatic disorders. Am J Psychother. 1973;27(4):557-565.
8. Vahia NS, Doongaji DR, Jeste DV, et al. Further experience with the therapy based upon concepts of Patanjali in the treatment of psychiatric disorders. Indian J Psychiatry. 1973;15(1):32-37.
9. Sahasi G, Mohan D, Kacker C. Effectiveness of yogic techniques in the management of anxiety. J Pers Clin Stud. 1989;5(1):51-55.
10. Sharma I, Azmi SA, Settiwar RM. Evaluation of the effect of pranayama in anxiety state. Alternative Medicine. 1991;3:227-235.
11. Shannahoff-Khalsa DS, Ray LE, Levine S, et al. Randomized controlled trial of yogic meditation techniques for patients with obsessive-compulsive disorder. CNS Spectr. 1999;4(12):34-47.
12. Gupta N, Khera S, Vempati RP, et al. Effect of yoga based lifestyle intervention on state and trait anxiety. Indian J Physiol Pharmacol. 2006;50(1):41-47.
13. Smith C, Hancock H, Blake-Mortimer J, et al. A randomised comparative trial of yoga and relaxation to reduce stress and anxiety. Complement Ther Med. 2007;15(2):77-83.
14. Descilo T, Vedamurtachar A, Gerbarg PL, et al. Effects of a yoga breath intervention alone and in combination with an exposure therapy for post-traumatic stress disorder and depression in survivors of the 2004 South-East Asia tsunami. Acta Psychiatr Scand. 2010;121(4):289-300.
15. Broocks A, Bandelow B, Pekrun G, et al. Comparison of aerobic exercise, clomipramine, and placebo in the treatment of panic disorder. Am J Psychiatry. 1998;155(5):603-609.
16. Merom D, Phongsavan P, Wagner R, et al. Promoting walking as an adjunct intervention to group cognitive behavioral therapy for anxiety disorders—a pilot group randomized trial. J Anxiety Disord. 2008;22(6):959-968.
17. Abrantes AM, Strong DR, Cohn A, et al. Acute changes in obsessions and compulsions following moderate-intensity aerobic exercise among patients with obsessive-compulsive disorder. J Anxiety Disord. 2009;23(7):923-927.
18. Lidren DM, Watkins PL, Gould RA, et al. A comparison of bibliotherapy and group therapy in the treatment of panic disorder. J Consult Clin Psychol. 1994;62(4):865-869.
19. Rapee RM, Abbott MJ, Lyneham HJ. Bibliotherapy for children with anxiety disorders using written materials for parents: a randomized controlled trial. J Consult Clin Psychol. 2006;74(3):436-444.
20. Abramowitz JS, Moore EL, Braddock AE, et al. Self-help cognitive-behavioral therapy with minimal therapist contact for social phobia: a controlled trial. J Behav Ther Exp Psychiatry. 2009;40(1):98-105.
21. Saeed SA, Bloch RM, Antonacci DJ. Herbal and dietary supplements for treatment of anxiety disorders. Am Fam Physician. 2007;76(4):549-556.
22. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-
23. Witte S, Loew D, Gaus W. Meta-analysis of the efficacy of the acetonic kava-kava extract WS1490 in patients with non-psychotic anxiety disorders. Phytother Res. 2005;19(3):183-188.
24. Sarris J, Kavanagh DJ, Byrne G, et al. The Kava Anxiety Depression Spectrum Study (KADSS): a randomized, placebo-controlled crossover trial using an aqueous extract of piper methysticum. Psychopharmacology (Berl). 2009;205:399-407.
25. Benjamin J, Levine J, Fux M, et al. Double-blind, placebo-controlled, crossover trial of inositol treatment for panic disorder. Am J Psychiatry. 1995;152(7):1084-1086.
26. Fux M, Levine J, Aviv A, et al. Inositol treatment of obsessive-compulsive disorder. Am J Psychiatry. 1996;153(9):1219-1221.
27. Fux M, Benjamin J, Belmaker RH. Inositol versus placebo augmentation of serotonin reuptake inhibitors in the treatment of obsessive-compulsive disorder: a double-blind cross-over study. Int J Neuropsychopharmcol. 1999;2(3):193-195.
28. Palatnik A, Frolov K, Fux M, et al. Double-blind, controlled, crossover trial of inositol versus fluvoxamine for the treatment of panic disorder. J Clin Psychopharmacol. 2001;21(3):335-339.
29. US Food and Drug Administration. Consumer advisory: kava-containing dietary supplements may be associated with severe injury. Available at: http://www.fda.gov/Food/ResourcesForYou/Consumers/ucm085482.htm. Accessed August 25, 2010.
30. Dragull K, Yoshida WY, Tang CS. Piperidine alkaloids from piper methysticum. Phytochemistry. 2003;63(2):193-198.
31. Simkins A, Thurston D, Colyar M, et al. Nature’s wrath? A closer look at complications with five popular herbs. Adv Nurse Pract. 2005;13(6):55-56.
32. Teschke R, Genthner A, Wolff A. Kava hepatotoxicity: comparison of aqueous, ethanolic, acetonic kava extracts and kava-herbs mixtures. J Ethnopharmacol. 2009;123(3):378-384.
33. Christl SU, Seifert A, Seeler D. Toxic hepatitis after consumption of traditional kava preparation. J Travel Med. 2009;16(1):55-56.
The number of people with psychiatric disorders who use complementary and alternative medicine (CAM) is on the rise. In surveys of patients seeking psychiatric care, estimates of CAM use range from 8% to 57%; the most frequent uses are for depression and anxiety disorders. A population-based study in the United States found that 9% of respondents had anxiety attacks and 57% of these individuals had used CAM.1 Similarly, in a Finnish population-based study (N=5,987) 35% of subjects reported some form of CAM use in the previous year; those with comorbid anxiety and depressive disorders used CAM most frequently.2
Unfortunately, a MEDLINE search shows that the number of studies examining psychotropic medications dwarfs the number of studies on even the most common CAM treatments used for psychiatric disorders. Far more patients with diagnosed mental disorders are studied in trials of standard treatments than CAM treatments. Because very few studies evaluate the cost-effectiveness of CAM treatments for psychiatric disorders, the risk-to-benefit ratio is difficult to calculate. Although several CAM treatments for depressive disorders have enough support to be considered options,3 CAM options for anxiety disorders are fewer and have less evidence of efficacy.
For these reasons, it is hard to recommend any CAM treatment as first line. Despite the relative lack of high quality research on CAM treatment outcomes, high rates of CAM use make it critical for clinicians to understand what treatments are available—or at least which treatments should be favored if patients are intent on trying them. We review the current research for yoga, exercise, bibliotherapy, and the dietary supplements kava and inositol for treating anxiety disorders and suggest those that warrant consideration for patients who do not respond, respond partially, or suffer from side effects from selective serotonin reuptake inhibitors (SSRIs) or benzodiazepines.
Limitations of CAM research
There are several limitations to the research literature on CAM approaches for anxiety disorders.4 First, there is a wide diversity of practices considered alternative or complementary and various ways in which these methods are applied across cultures. Some authors consider complementary medicines to be only herbal remedies, whereas others include individual therapies such as acupuncture, aromatherapy, herbal therapy, homeopathy, iridology, naturopathy, and reflexology.5 This article defines “alternative” treatments as those other than a form of psychotherapy or an FDA-approved medication that substitute for standard psychiatric treatment, and “complementary” approaches as those used to augment standard psychiatric treatments.
Anxiety and stress are ubiquitous, perhaps motivating interest in CAM options and prompting research on heterogeneous groups of individuals with poorly defined clinical syndromes or with isolated symptoms of anxiety or subjective distress. Few studies examine well-defined patient groups with diagnosed anxiety disorders. There are also multiple research design problems, including poorly specified treatments, poorly chosen placebos, and interpreting nonsignificant differences from established treatments as equivalence in underpowered studies.
The CAM treatments reviewed in this article have ≥2 randomized controlled trials (RCTs) that support their use for patients with diagnosed anxiety disorders, and ≥1 study that shows that the treatment can induce remission.
Yoga
In 2005 Kirkwood et al carried out the first systematic review of research evidence for the effectiveness of yoga in anxiety treatment.6 Of 19 studies identified, 4 RCTs and 1 nonrandomized trial met their inclusion criteria, which were an anxiety disorder diagnosis, use of yoga or yoga-based exercises alone, and anxiety rating scales used as outcome measures. Most found significant improvement in anxiety symptoms with yoga compared with placebo. Details of the 5 trials evaluated in Kirkwood’s review are summarized in Table 1.7-11
Since the 2005 review, 3 additional studies of yoga and anxiety have been published, but none would meet Kirkwood’s inclusion criteria. One that evaluated a heterogeneous group of patients using an intervention with multiple components—only 1 of which was yoga—found the intervention significantly reduced anxiety scores.12 A second study comparing yoga with relaxation in 131 patients with mild-to-moderate stress but no anxiety disorder diagnosis showed yoga was as effective as relaxation in improving anxiety symptoms as measured by the anxiety subscale of the State Trait Personality Inventory.13 In a study of 183 nonrandomized survivors of the 2004 southeast Asia tsunami with posttraumatic stress disorder (PTSD) symptoms, yoga-based breathing either alone or paired with trauma reduction exposure techniques significantly reduced PTSD symptoms compared with wait-list controls.14
Conclusion. Few controlled studies evaluated yoga for anxiety disorders, and all have significant methodologic limitations and/or poor methodology reporting. The diagnostic conditions treated and both yoga interventions and control conditions varied. However, these limited results are encouraging, particularly for treatment of obsessive-compulsive disorder (OCD). There is little information regarding safety or contraindications of yoga. Reported attrition rates were high in most studies, which may raise concerns about patient motivation and compliance.
Table 1
Evidence on the effectiveness of yoga for anxiety disorders
Study | Design | Results |
---|---|---|
Vahia et al, 19737 | 36 patients with psychoneurosis randomly assigned to yoga (N=15) or a control intervention of relaxation, postures, breathing, and writing (N=12) | Significant difference between groups in TAS scores after but not before treatment. Reduction in mean TAS score for yoga group but not control group |
Vahia et al,19738 | 39 patients received 6 weeks of yoga (N=21) or medication (amitriptyline and chlordiazepoxide on a variable dosage schedule) (N=18) | Yoga showed significantly greater reductions in TAS in this non-randomized sample |
Sahasi et al, 19899 | 91 patients randomly assigned to yoga practiced daily for 40 minutes (N=38) or diazepam at unspecified frequency or doses (N=53) for 3 months | Mean reduction in IPAT with yoga (3.39, P < .05) vs control group (0.36, P > .05). Attrition rate was 21.1% in yoga group and 66% in controls |
Sharma et al, 199110 | 71 patients with anxiety neurosis randomly assigned to 1-week yoga training, then daily practice (N=41) or control (N=30, placebo capsule) | HAM-A measured at 3 weekly intervals for 12 weeks. Significant between group mean difference at 3 weeks (greater improvement in yoga group compared with controls). Significant improvement in yoga group between 3 and 6 weeks but not for controls |
Shannahoff-Khalsa et al, 199911 | 21 OCD patients randomly assigned to kundalini yoga (N=11) or relaxation and mindfulness meditation (N=10). Multiple outcome measures; Y-BOCS was primary | Seven in each group completed 3 months; patients who practiced yoga demonstrated greater improvements on Y-BOCS. Intent-to-treat analysis (Y-BOCS) for the baseline and 3-month tests showed that only the yoga group improved. Groups were merged for an additional year of yoga; at 15 months, the final group (N=11) improved 71% on the Y-BOCS |
HAM-A: Hamilton Anxiety Rating scale; IPAT: Institute for Personality and Ability Testing, Anxiety Scale; OCD: obsessive-compulsive disorder; TAS: Taylor’s Anxiety Scale; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Exercise
The literature examining the relationship between exercise and depression is extensive, but much less has been published about exercise in patients with anxiety disorders (Table 2).15-17 In a 10-week trial, Broocks and colleagues compared clomipramine, exercise (running), and placebo in 46 outpatients with panic disorder.15 Both exercise and clomipramine, 112.5 mg/d, significantly reduced panic symptoms compared with placebo, but clomipramine was more effective and faster-acting.
A more recent RCT compared group cognitive-behavioral therapy (GCBT) plus a home-based walking program vs GCBT and in 21 patients with panic disorder, generalized anxiety disorder (GAD), or social phobia.16 Compared with GCBT plus educational sessions, GCBT plus walking had a significant effect on self-reported depression, anxiety, and stress. Results differed by diagnosis; the most marked effects occurred in individuals with social phobia, whereas benefits for those with panic disorder or GAD were questionable.
Fifteen patients with OCD were recruited to participate in a 12-week, moderate-intensity aerobic exercise program added to their standard behavioral and/or pharmacologic treatment.17 Subjects demonstrated improvement in negative mood, anxiety, obsessions, and compulsions after each exercise session. Changes after each session persisted over the 12-week intervention, although the magnitude attenuated over the duration of the intervention.
Conclusion. Although initial results from small trials suggest exercise may help improve anxiety symptoms, further studies are needed to determine how to best use exercise training to treat anxious patients, specifically regarding dose-response relationship, differences in effectiveness between aerobic and resistance training, and the mechanisms by which exercise improves psychiatric symptoms.
Table 2
Exercise for anxiety: More research is needed
Study | Design | Results |
---|---|---|
Broocks et al, 199915 | 46 patients with panic disorder randomly assigned to 10 weeks of running, clomipramine, or wplacebo pills | Both exercise and clomipramine resulted in significant decreases in symptoms but clomipramine improved symptoms earlier and more effectively |
Merom et al, 200816 | 21 patients with panic disorder, GAD, or social phobia randomly assigned to GCBT and either a home-based walking program or educational sessions | GCBT plus walking had a significant effect on depression, anxiety, and stress compared with GCBT plus educational sessions |
Abrantes et al, 200917 | 15 patients with OCD assigned to a 12-week exercise intervention that was added to their standard behavioral and/or pharmacologic treatment | Subjects reported improved mood, anxiety, obsessions, and compulsions after each exercise session |
GAD: generalized anxiety disorder; GCBT: group cognitive-behavioral therapy |
Bibliotherapy
Investigation of bibliotherapy for treatment of anxiety disorders has been limited (Table 3).18-20 A 2009 RCT demonstrated that for 21 patients with mild-to-moderate social phobia, bibliotherapy—in the form of an 8-week self-directed CBT program with minimal therapist involvement—was superior to a wait-list control and induced clinically significant change in approximately one-third of patients.20
Rapee et al randomly assigned 267 children age 6 to 12 with anxiety disorders to bibliotherapy that consisted of parents treating their children in the home with written materials, 9 sessions of GCBT, or a wait-list control condition.19 Bibliotherapy provided by parents demonstrated benefit compared with wait-listing but was not as efficacious as GCBT at post-treatment and 3-month follow-up.
Lidren and colleagues randomly assigned 36 adult patients with panic disorder to bibliotherapy, group therapy combined with bibliotherapy, or a waitlist.18 Both treatments were more effective than wait-listing in reducing the frequency of panic attacks, severity of physical panic symptoms, catastrophic cognitions, agoraphobic avoidance, and depression. Both interventions maintained their effects at 3-and 6-month follow-up and produced clinically significant change in most patients.
Conclusion. Some preliminary evidence supports the effectiveness of bibliotherapy for social anxiety disorder, childhood anxiety disorders, and panic disorder.
Table 3
Preliminary evidence supports bibliotherapy for select anxiety disorders
Study | Design | Results |
---|---|---|
Lidren et al, 199418 | 36 adults with panic disorder randomly assigned to bibliotherapy, bibliotherapy plus group therapy, or wait-list control | Both bibliotherapy and bibliotherapy plus group therapy were more effective than wait-listing in reducing the frequency of panic attacks and severity of physical panic symptoms |
Rapee et al, 200619 | 267 children with anxiety disorders randomly assigned to bibliotherapy (parents treating their children in the home with written materials with no therapist contact), 9 sessions of group CBT, or wait-list control | Parent-delivered bibliotherapy was beneficial compared with wait-listing but was not as efficacious as group CBT |
Abramowitz et al, 200920 | 21 patients with mild-to-moderate social phobia underwent an 8-week self-directed CBT program with minimal therapist involvement | Bibliotherapy was superior to wait-listing. One-third of patients experienced clinically significant change |
CBT: cognitive-behavioral therapy |
Dietary supplements
Many dietary and herbal supplements are purported to have therapeutic efficacy for anxiety symptoms. Because of inadequate FDA regulation of manufacturing and marketing of these agents, most of these supplements have not been tested on patients with anxiety disorders.21 Limited evidence supports the use of kava for GAD and inositol for panic disorder (Table 4).22-28
Kava. Multiple double-blind RCTs found kava (Piper methysticum)—a plant indigenous to South Pacific islands—has effects greater than placebo and comparable to standard treatments for mild to moderately severe GAD. A Cochrane meta-analysis22 of 11 trials with 645 participants concluded that kava is effective for reducing GAD symptoms, with risks comparable to standard treatments for up to 6 months of use.
Case reports of kava-associated liver toxicity led to a marketing ban in Canada in 2000, followed shortly by Germany, Australia, and the United Kingdom. In 2002 the FDA issued a Consumer Advisory29 discouraging kava use. Since then a flurry of research has looked for sources of possible toxicity, including individual sensitivities,29 excessive dosing, use of toxic parts of the kava plant instead of the roots,30,31 interactions with other hepatoactive substances, and non-water based extraction methods. RCTs demonstrating kava’s efficacy and safety were characterized by careful dosing supervision, use of standardized kava extracts, and avoidance of interactions with other hepatoactive medications or CAM treatments. Doses ≤300 mg/d are recommended.22
RCTs that used the standardized acetone extract WS149023 found that women and younger adults show more positive effects from kava, and showed no liver toxicity when used for 1 to 24 weeks. A recent RCT that used kava extracts obtained via water-based methods showed kava had significant anxiolytic effects.24 However, a study of liver toxicity reports found that water-based extractions, acetonic extractions, and ethanol extractions all have been associated with toxic hepatic reactions.32 Aqueous extraction does not guarantee safety, and the extraction solvent does not cause toxicity. A recent report of a severe liver reaction to the native drink by a tourist in Samoa33 suggests that aqueous extractions from the root stock— the type of kava used by South Pacific islanders—also can be unsafe.
Conclusion. Multiple RCTs have found kava relatively safe and effective for treating anxiety symptoms. Caution is necessary, however, because of reports of liver toxicity associated with its use. Physician oversight and monitoring of kava use are appropriate.
Inositol. Evidence from RCTs suggests inositol, a natural isomer of glucose and a precursor in the phosphatidylinositol cycle, can significantly improve panic disorder symptoms.25-28 In 1 trial, efficacy and side effects were comparable to fluvoxamine.28 Effective doses ranged from 12 g/d to 18 g/d. Researchers tested inositol as monotherapy or augmentation to SSRIs for patients with mild-to-moderate OCD. In small double-blind crossover RCTs, inositol monotherapy significantly reduced Yale-Brown Obsessive Compulsive Scale scores compared with placebo26 but inositol augmentation added nothing to the effects of SSRIs.27
Conclusion. Inositol appears to be effective in improving symptoms of panic disorder. Its use for other anxiety disorders is unproven.
Table 4
Dietary supplements for anxiety disorders
Study | Design | Results |
---|---|---|
Kava | ||
Pittler et al, 200322 | Meta-analysis of 11 RCTs with a total of 645 GAD patients | Compared with placebo, kava significantly reduced anxiety as measured by total HAM-A score |
Witte et al, 200523 | Meta-analysis of 6 RCTs using kava extract WS1490 in patients with nonpsychotic anxiety disorders | Kava reduced HAM-A score more than placebo and seemed to be more effective in women and younger adults |
Sarris et al, 200924 | 60 adults with ≥1 month of elevated generalized anxiety randomly assigned to an aqueous extract of kava | Aqueous-extract kava was significantly more effective than placebo in reducing HAM-A score |
Inositol | ||
Benjamin et al, 199525 | 21 patients with panic disorder with or without agoraphobia randomly assigned to inositol, 12 g/d, or placebo | Inositol significantly reduced frequency and severity of panic attacks and severity of agoraphobia compared with placebo |
Fux et al, 199626 | 13 OCD patients randomly assigned to inositol,18 g/d, or placebo for 6 weeks | Patients taking inositol had significantly lower Y-BOCS scores compared with those receiving placebo |
Fux et al, 199927 | 10 OCD patients receiving an SSRI randomly assigned to augmentation with inositol, 18 g/d, or placebo for 6 weeks | No significant differences between treatments |
Palatnik et al, 200128 | In a crossover trial, 20 panic disorder patients completed 1 month of inositol, up to 18 g/d, and 1 month of fluvoxamine, up to 150 mg/d | Improvements in HAM-A, CGI, and agoraphobia scores were similar for both treatments |
CGI: Clinical Global Impression scale; GAD: generalized anxiety disorder; HAM-A: Hamilton Anxiety Rating scale; OCD: obsessive-compulsive disorder; RCTs: randomized controlled trials; SSRI: selective serotonin reuptake inhibitor; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Supervision is recommended
The evidence base for most CAM interventions commonly used for anxiety is relatively poor and recent systematic reviews found few methodologically rigorous studies. This has not, however, diminished CAM treatments’ popularity. Despite a paucity of high-quality studies regarding CAM for anxiety disorders, there is enough data supporting yoga, exercise, bibliotherapy, kava, and inositol to allow psychiatrists to collaborate with patients who wish to try these treatments. Advise patients that they may need physician supervision similar to that used with standard psychiatric treatments.
Related Resources
- National Center for Complementary and Alternative Medicine. http://nccam.nih.gov.
- The Journal of Alternative and Complementary Medicine. www.liebertonline.com/loi/acm.
- MedlinePlus: Complementary and Alternative Medicine. www.nlm.nih.gov/medlineplus/complementaryandalternativemedicine.html.
Drug Brand Names
- Amitriptyline • Elavil
- Chlordiazepoxide • Librium
- Clomipramine • Anafranil
- Diazepam • Valium
- Fluvoxamine • Luvox
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
The number of people with psychiatric disorders who use complementary and alternative medicine (CAM) is on the rise. In surveys of patients seeking psychiatric care, estimates of CAM use range from 8% to 57%; the most frequent uses are for depression and anxiety disorders. A population-based study in the United States found that 9% of respondents had anxiety attacks and 57% of these individuals had used CAM.1 Similarly, in a Finnish population-based study (N=5,987) 35% of subjects reported some form of CAM use in the previous year; those with comorbid anxiety and depressive disorders used CAM most frequently.2
Unfortunately, a MEDLINE search shows that the number of studies examining psychotropic medications dwarfs the number of studies on even the most common CAM treatments used for psychiatric disorders. Far more patients with diagnosed mental disorders are studied in trials of standard treatments than CAM treatments. Because very few studies evaluate the cost-effectiveness of CAM treatments for psychiatric disorders, the risk-to-benefit ratio is difficult to calculate. Although several CAM treatments for depressive disorders have enough support to be considered options,3 CAM options for anxiety disorders are fewer and have less evidence of efficacy.
For these reasons, it is hard to recommend any CAM treatment as first line. Despite the relative lack of high quality research on CAM treatment outcomes, high rates of CAM use make it critical for clinicians to understand what treatments are available—or at least which treatments should be favored if patients are intent on trying them. We review the current research for yoga, exercise, bibliotherapy, and the dietary supplements kava and inositol for treating anxiety disorders and suggest those that warrant consideration for patients who do not respond, respond partially, or suffer from side effects from selective serotonin reuptake inhibitors (SSRIs) or benzodiazepines.
Limitations of CAM research
There are several limitations to the research literature on CAM approaches for anxiety disorders.4 First, there is a wide diversity of practices considered alternative or complementary and various ways in which these methods are applied across cultures. Some authors consider complementary medicines to be only herbal remedies, whereas others include individual therapies such as acupuncture, aromatherapy, herbal therapy, homeopathy, iridology, naturopathy, and reflexology.5 This article defines “alternative” treatments as those other than a form of psychotherapy or an FDA-approved medication that substitute for standard psychiatric treatment, and “complementary” approaches as those used to augment standard psychiatric treatments.
Anxiety and stress are ubiquitous, perhaps motivating interest in CAM options and prompting research on heterogeneous groups of individuals with poorly defined clinical syndromes or with isolated symptoms of anxiety or subjective distress. Few studies examine well-defined patient groups with diagnosed anxiety disorders. There are also multiple research design problems, including poorly specified treatments, poorly chosen placebos, and interpreting nonsignificant differences from established treatments as equivalence in underpowered studies.
The CAM treatments reviewed in this article have ≥2 randomized controlled trials (RCTs) that support their use for patients with diagnosed anxiety disorders, and ≥1 study that shows that the treatment can induce remission.
Yoga
In 2005 Kirkwood et al carried out the first systematic review of research evidence for the effectiveness of yoga in anxiety treatment.6 Of 19 studies identified, 4 RCTs and 1 nonrandomized trial met their inclusion criteria, which were an anxiety disorder diagnosis, use of yoga or yoga-based exercises alone, and anxiety rating scales used as outcome measures. Most found significant improvement in anxiety symptoms with yoga compared with placebo. Details of the 5 trials evaluated in Kirkwood’s review are summarized in Table 1.7-11
Since the 2005 review, 3 additional studies of yoga and anxiety have been published, but none would meet Kirkwood’s inclusion criteria. One that evaluated a heterogeneous group of patients using an intervention with multiple components—only 1 of which was yoga—found the intervention significantly reduced anxiety scores.12 A second study comparing yoga with relaxation in 131 patients with mild-to-moderate stress but no anxiety disorder diagnosis showed yoga was as effective as relaxation in improving anxiety symptoms as measured by the anxiety subscale of the State Trait Personality Inventory.13 In a study of 183 nonrandomized survivors of the 2004 southeast Asia tsunami with posttraumatic stress disorder (PTSD) symptoms, yoga-based breathing either alone or paired with trauma reduction exposure techniques significantly reduced PTSD symptoms compared with wait-list controls.14
Conclusion. Few controlled studies evaluated yoga for anxiety disorders, and all have significant methodologic limitations and/or poor methodology reporting. The diagnostic conditions treated and both yoga interventions and control conditions varied. However, these limited results are encouraging, particularly for treatment of obsessive-compulsive disorder (OCD). There is little information regarding safety or contraindications of yoga. Reported attrition rates were high in most studies, which may raise concerns about patient motivation and compliance.
Table 1
Evidence on the effectiveness of yoga for anxiety disorders
Study | Design | Results |
---|---|---|
Vahia et al, 19737 | 36 patients with psychoneurosis randomly assigned to yoga (N=15) or a control intervention of relaxation, postures, breathing, and writing (N=12) | Significant difference between groups in TAS scores after but not before treatment. Reduction in mean TAS score for yoga group but not control group |
Vahia et al,19738 | 39 patients received 6 weeks of yoga (N=21) or medication (amitriptyline and chlordiazepoxide on a variable dosage schedule) (N=18) | Yoga showed significantly greater reductions in TAS in this non-randomized sample |
Sahasi et al, 19899 | 91 patients randomly assigned to yoga practiced daily for 40 minutes (N=38) or diazepam at unspecified frequency or doses (N=53) for 3 months | Mean reduction in IPAT with yoga (3.39, P < .05) vs control group (0.36, P > .05). Attrition rate was 21.1% in yoga group and 66% in controls |
Sharma et al, 199110 | 71 patients with anxiety neurosis randomly assigned to 1-week yoga training, then daily practice (N=41) or control (N=30, placebo capsule) | HAM-A measured at 3 weekly intervals for 12 weeks. Significant between group mean difference at 3 weeks (greater improvement in yoga group compared with controls). Significant improvement in yoga group between 3 and 6 weeks but not for controls |
Shannahoff-Khalsa et al, 199911 | 21 OCD patients randomly assigned to kundalini yoga (N=11) or relaxation and mindfulness meditation (N=10). Multiple outcome measures; Y-BOCS was primary | Seven in each group completed 3 months; patients who practiced yoga demonstrated greater improvements on Y-BOCS. Intent-to-treat analysis (Y-BOCS) for the baseline and 3-month tests showed that only the yoga group improved. Groups were merged for an additional year of yoga; at 15 months, the final group (N=11) improved 71% on the Y-BOCS |
HAM-A: Hamilton Anxiety Rating scale; IPAT: Institute for Personality and Ability Testing, Anxiety Scale; OCD: obsessive-compulsive disorder; TAS: Taylor’s Anxiety Scale; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Exercise
The literature examining the relationship between exercise and depression is extensive, but much less has been published about exercise in patients with anxiety disorders (Table 2).15-17 In a 10-week trial, Broocks and colleagues compared clomipramine, exercise (running), and placebo in 46 outpatients with panic disorder.15 Both exercise and clomipramine, 112.5 mg/d, significantly reduced panic symptoms compared with placebo, but clomipramine was more effective and faster-acting.
A more recent RCT compared group cognitive-behavioral therapy (GCBT) plus a home-based walking program vs GCBT and in 21 patients with panic disorder, generalized anxiety disorder (GAD), or social phobia.16 Compared with GCBT plus educational sessions, GCBT plus walking had a significant effect on self-reported depression, anxiety, and stress. Results differed by diagnosis; the most marked effects occurred in individuals with social phobia, whereas benefits for those with panic disorder or GAD were questionable.
Fifteen patients with OCD were recruited to participate in a 12-week, moderate-intensity aerobic exercise program added to their standard behavioral and/or pharmacologic treatment.17 Subjects demonstrated improvement in negative mood, anxiety, obsessions, and compulsions after each exercise session. Changes after each session persisted over the 12-week intervention, although the magnitude attenuated over the duration of the intervention.
Conclusion. Although initial results from small trials suggest exercise may help improve anxiety symptoms, further studies are needed to determine how to best use exercise training to treat anxious patients, specifically regarding dose-response relationship, differences in effectiveness between aerobic and resistance training, and the mechanisms by which exercise improves psychiatric symptoms.
Table 2
Exercise for anxiety: More research is needed
Study | Design | Results |
---|---|---|
Broocks et al, 199915 | 46 patients with panic disorder randomly assigned to 10 weeks of running, clomipramine, or wplacebo pills | Both exercise and clomipramine resulted in significant decreases in symptoms but clomipramine improved symptoms earlier and more effectively |
Merom et al, 200816 | 21 patients with panic disorder, GAD, or social phobia randomly assigned to GCBT and either a home-based walking program or educational sessions | GCBT plus walking had a significant effect on depression, anxiety, and stress compared with GCBT plus educational sessions |
Abrantes et al, 200917 | 15 patients with OCD assigned to a 12-week exercise intervention that was added to their standard behavioral and/or pharmacologic treatment | Subjects reported improved mood, anxiety, obsessions, and compulsions after each exercise session |
GAD: generalized anxiety disorder; GCBT: group cognitive-behavioral therapy |
Bibliotherapy
Investigation of bibliotherapy for treatment of anxiety disorders has been limited (Table 3).18-20 A 2009 RCT demonstrated that for 21 patients with mild-to-moderate social phobia, bibliotherapy—in the form of an 8-week self-directed CBT program with minimal therapist involvement—was superior to a wait-list control and induced clinically significant change in approximately one-third of patients.20
Rapee et al randomly assigned 267 children age 6 to 12 with anxiety disorders to bibliotherapy that consisted of parents treating their children in the home with written materials, 9 sessions of GCBT, or a wait-list control condition.19 Bibliotherapy provided by parents demonstrated benefit compared with wait-listing but was not as efficacious as GCBT at post-treatment and 3-month follow-up.
Lidren and colleagues randomly assigned 36 adult patients with panic disorder to bibliotherapy, group therapy combined with bibliotherapy, or a waitlist.18 Both treatments were more effective than wait-listing in reducing the frequency of panic attacks, severity of physical panic symptoms, catastrophic cognitions, agoraphobic avoidance, and depression. Both interventions maintained their effects at 3-and 6-month follow-up and produced clinically significant change in most patients.
Conclusion. Some preliminary evidence supports the effectiveness of bibliotherapy for social anxiety disorder, childhood anxiety disorders, and panic disorder.
Table 3
Preliminary evidence supports bibliotherapy for select anxiety disorders
Study | Design | Results |
---|---|---|
Lidren et al, 199418 | 36 adults with panic disorder randomly assigned to bibliotherapy, bibliotherapy plus group therapy, or wait-list control | Both bibliotherapy and bibliotherapy plus group therapy were more effective than wait-listing in reducing the frequency of panic attacks and severity of physical panic symptoms |
Rapee et al, 200619 | 267 children with anxiety disorders randomly assigned to bibliotherapy (parents treating their children in the home with written materials with no therapist contact), 9 sessions of group CBT, or wait-list control | Parent-delivered bibliotherapy was beneficial compared with wait-listing but was not as efficacious as group CBT |
Abramowitz et al, 200920 | 21 patients with mild-to-moderate social phobia underwent an 8-week self-directed CBT program with minimal therapist involvement | Bibliotherapy was superior to wait-listing. One-third of patients experienced clinically significant change |
CBT: cognitive-behavioral therapy |
Dietary supplements
Many dietary and herbal supplements are purported to have therapeutic efficacy for anxiety symptoms. Because of inadequate FDA regulation of manufacturing and marketing of these agents, most of these supplements have not been tested on patients with anxiety disorders.21 Limited evidence supports the use of kava for GAD and inositol for panic disorder (Table 4).22-28
Kava. Multiple double-blind RCTs found kava (Piper methysticum)—a plant indigenous to South Pacific islands—has effects greater than placebo and comparable to standard treatments for mild to moderately severe GAD. A Cochrane meta-analysis22 of 11 trials with 645 participants concluded that kava is effective for reducing GAD symptoms, with risks comparable to standard treatments for up to 6 months of use.
Case reports of kava-associated liver toxicity led to a marketing ban in Canada in 2000, followed shortly by Germany, Australia, and the United Kingdom. In 2002 the FDA issued a Consumer Advisory29 discouraging kava use. Since then a flurry of research has looked for sources of possible toxicity, including individual sensitivities,29 excessive dosing, use of toxic parts of the kava plant instead of the roots,30,31 interactions with other hepatoactive substances, and non-water based extraction methods. RCTs demonstrating kava’s efficacy and safety were characterized by careful dosing supervision, use of standardized kava extracts, and avoidance of interactions with other hepatoactive medications or CAM treatments. Doses ≤300 mg/d are recommended.22
RCTs that used the standardized acetone extract WS149023 found that women and younger adults show more positive effects from kava, and showed no liver toxicity when used for 1 to 24 weeks. A recent RCT that used kava extracts obtained via water-based methods showed kava had significant anxiolytic effects.24 However, a study of liver toxicity reports found that water-based extractions, acetonic extractions, and ethanol extractions all have been associated with toxic hepatic reactions.32 Aqueous extraction does not guarantee safety, and the extraction solvent does not cause toxicity. A recent report of a severe liver reaction to the native drink by a tourist in Samoa33 suggests that aqueous extractions from the root stock— the type of kava used by South Pacific islanders—also can be unsafe.
Conclusion. Multiple RCTs have found kava relatively safe and effective for treating anxiety symptoms. Caution is necessary, however, because of reports of liver toxicity associated with its use. Physician oversight and monitoring of kava use are appropriate.
Inositol. Evidence from RCTs suggests inositol, a natural isomer of glucose and a precursor in the phosphatidylinositol cycle, can significantly improve panic disorder symptoms.25-28 In 1 trial, efficacy and side effects were comparable to fluvoxamine.28 Effective doses ranged from 12 g/d to 18 g/d. Researchers tested inositol as monotherapy or augmentation to SSRIs for patients with mild-to-moderate OCD. In small double-blind crossover RCTs, inositol monotherapy significantly reduced Yale-Brown Obsessive Compulsive Scale scores compared with placebo26 but inositol augmentation added nothing to the effects of SSRIs.27
Conclusion. Inositol appears to be effective in improving symptoms of panic disorder. Its use for other anxiety disorders is unproven.
Table 4
Dietary supplements for anxiety disorders
Study | Design | Results |
---|---|---|
Kava | ||
Pittler et al, 200322 | Meta-analysis of 11 RCTs with a total of 645 GAD patients | Compared with placebo, kava significantly reduced anxiety as measured by total HAM-A score |
Witte et al, 200523 | Meta-analysis of 6 RCTs using kava extract WS1490 in patients with nonpsychotic anxiety disorders | Kava reduced HAM-A score more than placebo and seemed to be more effective in women and younger adults |
Sarris et al, 200924 | 60 adults with ≥1 month of elevated generalized anxiety randomly assigned to an aqueous extract of kava | Aqueous-extract kava was significantly more effective than placebo in reducing HAM-A score |
Inositol | ||
Benjamin et al, 199525 | 21 patients with panic disorder with or without agoraphobia randomly assigned to inositol, 12 g/d, or placebo | Inositol significantly reduced frequency and severity of panic attacks and severity of agoraphobia compared with placebo |
Fux et al, 199626 | 13 OCD patients randomly assigned to inositol,18 g/d, or placebo for 6 weeks | Patients taking inositol had significantly lower Y-BOCS scores compared with those receiving placebo |
Fux et al, 199927 | 10 OCD patients receiving an SSRI randomly assigned to augmentation with inositol, 18 g/d, or placebo for 6 weeks | No significant differences between treatments |
Palatnik et al, 200128 | In a crossover trial, 20 panic disorder patients completed 1 month of inositol, up to 18 g/d, and 1 month of fluvoxamine, up to 150 mg/d | Improvements in HAM-A, CGI, and agoraphobia scores were similar for both treatments |
CGI: Clinical Global Impression scale; GAD: generalized anxiety disorder; HAM-A: Hamilton Anxiety Rating scale; OCD: obsessive-compulsive disorder; RCTs: randomized controlled trials; SSRI: selective serotonin reuptake inhibitor; Y-BOCS: Yale-Brown Obsessive Compulsive Scale |
Supervision is recommended
The evidence base for most CAM interventions commonly used for anxiety is relatively poor and recent systematic reviews found few methodologically rigorous studies. This has not, however, diminished CAM treatments’ popularity. Despite a paucity of high-quality studies regarding CAM for anxiety disorders, there is enough data supporting yoga, exercise, bibliotherapy, kava, and inositol to allow psychiatrists to collaborate with patients who wish to try these treatments. Advise patients that they may need physician supervision similar to that used with standard psychiatric treatments.
Related Resources
- National Center for Complementary and Alternative Medicine. http://nccam.nih.gov.
- The Journal of Alternative and Complementary Medicine. www.liebertonline.com/loi/acm.
- MedlinePlus: Complementary and Alternative Medicine. www.nlm.nih.gov/medlineplus/complementaryandalternativemedicine.html.
Drug Brand Names
- Amitriptyline • Elavil
- Chlordiazepoxide • Librium
- Clomipramine • Anafranil
- Diazepam • Valium
- Fluvoxamine • Luvox
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Kessler RC, Soukup J, Davis RB, et al. The use of complementary and alternative therapies to treat anxiety and depression in the United States. Am J Psychiatry. 2001;158(2):289-294.
2. Wahlström M, Sihvo S, Haukkala A, et al. Use of mental health services and complementary and alternative medicine in persons with common mental disorders. Acta Psychiatr Scand. 2008;118(1):73-80.
3. Saeed SA, Bloch RM, Antonacci DJ, et al. CAM for your depressed patient: 6 recommended options. Current Psychiatry. 2009;8(10):39-47.
4. Pilkington K. Searching for CAM evidence: an evaluation of therapy-specific search strategies. J Altern Complement Med. 2007;13(4):451-459.
5. van der Watt G, Laugharne J, Janca A. Complementary and alternative medicine in the treatment of anxiety and depression. Curr Opin Psychiatry. 2008;21(1):37-42.
6. Kirkwood G, Rampes H, Tuffrey V, et al. Yoga for anxiety: a systematic review of the research evidence. Br J Sports Med. 2005;39(12):884-891.
7. Vahia NS, Doongaji DR, Jeste DV, et al. Psychophysiologic therapy based on the concepts of Patanjali. A new approach to the treatment of neurotic and psychosomatic disorders. Am J Psychother. 1973;27(4):557-565.
8. Vahia NS, Doongaji DR, Jeste DV, et al. Further experience with the therapy based upon concepts of Patanjali in the treatment of psychiatric disorders. Indian J Psychiatry. 1973;15(1):32-37.
9. Sahasi G, Mohan D, Kacker C. Effectiveness of yogic techniques in the management of anxiety. J Pers Clin Stud. 1989;5(1):51-55.
10. Sharma I, Azmi SA, Settiwar RM. Evaluation of the effect of pranayama in anxiety state. Alternative Medicine. 1991;3:227-235.
11. Shannahoff-Khalsa DS, Ray LE, Levine S, et al. Randomized controlled trial of yogic meditation techniques for patients with obsessive-compulsive disorder. CNS Spectr. 1999;4(12):34-47.
12. Gupta N, Khera S, Vempati RP, et al. Effect of yoga based lifestyle intervention on state and trait anxiety. Indian J Physiol Pharmacol. 2006;50(1):41-47.
13. Smith C, Hancock H, Blake-Mortimer J, et al. A randomised comparative trial of yoga and relaxation to reduce stress and anxiety. Complement Ther Med. 2007;15(2):77-83.
14. Descilo T, Vedamurtachar A, Gerbarg PL, et al. Effects of a yoga breath intervention alone and in combination with an exposure therapy for post-traumatic stress disorder and depression in survivors of the 2004 South-East Asia tsunami. Acta Psychiatr Scand. 2010;121(4):289-300.
15. Broocks A, Bandelow B, Pekrun G, et al. Comparison of aerobic exercise, clomipramine, and placebo in the treatment of panic disorder. Am J Psychiatry. 1998;155(5):603-609.
16. Merom D, Phongsavan P, Wagner R, et al. Promoting walking as an adjunct intervention to group cognitive behavioral therapy for anxiety disorders—a pilot group randomized trial. J Anxiety Disord. 2008;22(6):959-968.
17. Abrantes AM, Strong DR, Cohn A, et al. Acute changes in obsessions and compulsions following moderate-intensity aerobic exercise among patients with obsessive-compulsive disorder. J Anxiety Disord. 2009;23(7):923-927.
18. Lidren DM, Watkins PL, Gould RA, et al. A comparison of bibliotherapy and group therapy in the treatment of panic disorder. J Consult Clin Psychol. 1994;62(4):865-869.
19. Rapee RM, Abbott MJ, Lyneham HJ. Bibliotherapy for children with anxiety disorders using written materials for parents: a randomized controlled trial. J Consult Clin Psychol. 2006;74(3):436-444.
20. Abramowitz JS, Moore EL, Braddock AE, et al. Self-help cognitive-behavioral therapy with minimal therapist contact for social phobia: a controlled trial. J Behav Ther Exp Psychiatry. 2009;40(1):98-105.
21. Saeed SA, Bloch RM, Antonacci DJ. Herbal and dietary supplements for treatment of anxiety disorders. Am Fam Physician. 2007;76(4):549-556.
22. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-
23. Witte S, Loew D, Gaus W. Meta-analysis of the efficacy of the acetonic kava-kava extract WS1490 in patients with non-psychotic anxiety disorders. Phytother Res. 2005;19(3):183-188.
24. Sarris J, Kavanagh DJ, Byrne G, et al. The Kava Anxiety Depression Spectrum Study (KADSS): a randomized, placebo-controlled crossover trial using an aqueous extract of piper methysticum. Psychopharmacology (Berl). 2009;205:399-407.
25. Benjamin J, Levine J, Fux M, et al. Double-blind, placebo-controlled, crossover trial of inositol treatment for panic disorder. Am J Psychiatry. 1995;152(7):1084-1086.
26. Fux M, Levine J, Aviv A, et al. Inositol treatment of obsessive-compulsive disorder. Am J Psychiatry. 1996;153(9):1219-1221.
27. Fux M, Benjamin J, Belmaker RH. Inositol versus placebo augmentation of serotonin reuptake inhibitors in the treatment of obsessive-compulsive disorder: a double-blind cross-over study. Int J Neuropsychopharmcol. 1999;2(3):193-195.
28. Palatnik A, Frolov K, Fux M, et al. Double-blind, controlled, crossover trial of inositol versus fluvoxamine for the treatment of panic disorder. J Clin Psychopharmacol. 2001;21(3):335-339.
29. US Food and Drug Administration. Consumer advisory: kava-containing dietary supplements may be associated with severe injury. Available at: http://www.fda.gov/Food/ResourcesForYou/Consumers/ucm085482.htm. Accessed August 25, 2010.
30. Dragull K, Yoshida WY, Tang CS. Piperidine alkaloids from piper methysticum. Phytochemistry. 2003;63(2):193-198.
31. Simkins A, Thurston D, Colyar M, et al. Nature’s wrath? A closer look at complications with five popular herbs. Adv Nurse Pract. 2005;13(6):55-56.
32. Teschke R, Genthner A, Wolff A. Kava hepatotoxicity: comparison of aqueous, ethanolic, acetonic kava extracts and kava-herbs mixtures. J Ethnopharmacol. 2009;123(3):378-384.
33. Christl SU, Seifert A, Seeler D. Toxic hepatitis after consumption of traditional kava preparation. J Travel Med. 2009;16(1):55-56.
1. Kessler RC, Soukup J, Davis RB, et al. The use of complementary and alternative therapies to treat anxiety and depression in the United States. Am J Psychiatry. 2001;158(2):289-294.
2. Wahlström M, Sihvo S, Haukkala A, et al. Use of mental health services and complementary and alternative medicine in persons with common mental disorders. Acta Psychiatr Scand. 2008;118(1):73-80.
3. Saeed SA, Bloch RM, Antonacci DJ, et al. CAM for your depressed patient: 6 recommended options. Current Psychiatry. 2009;8(10):39-47.
4. Pilkington K. Searching for CAM evidence: an evaluation of therapy-specific search strategies. J Altern Complement Med. 2007;13(4):451-459.
5. van der Watt G, Laugharne J, Janca A. Complementary and alternative medicine in the treatment of anxiety and depression. Curr Opin Psychiatry. 2008;21(1):37-42.
6. Kirkwood G, Rampes H, Tuffrey V, et al. Yoga for anxiety: a systematic review of the research evidence. Br J Sports Med. 2005;39(12):884-891.
7. Vahia NS, Doongaji DR, Jeste DV, et al. Psychophysiologic therapy based on the concepts of Patanjali. A new approach to the treatment of neurotic and psychosomatic disorders. Am J Psychother. 1973;27(4):557-565.
8. Vahia NS, Doongaji DR, Jeste DV, et al. Further experience with the therapy based upon concepts of Patanjali in the treatment of psychiatric disorders. Indian J Psychiatry. 1973;15(1):32-37.
9. Sahasi G, Mohan D, Kacker C. Effectiveness of yogic techniques in the management of anxiety. J Pers Clin Stud. 1989;5(1):51-55.
10. Sharma I, Azmi SA, Settiwar RM. Evaluation of the effect of pranayama in anxiety state. Alternative Medicine. 1991;3:227-235.
11. Shannahoff-Khalsa DS, Ray LE, Levine S, et al. Randomized controlled trial of yogic meditation techniques for patients with obsessive-compulsive disorder. CNS Spectr. 1999;4(12):34-47.
12. Gupta N, Khera S, Vempati RP, et al. Effect of yoga based lifestyle intervention on state and trait anxiety. Indian J Physiol Pharmacol. 2006;50(1):41-47.
13. Smith C, Hancock H, Blake-Mortimer J, et al. A randomised comparative trial of yoga and relaxation to reduce stress and anxiety. Complement Ther Med. 2007;15(2):77-83.
14. Descilo T, Vedamurtachar A, Gerbarg PL, et al. Effects of a yoga breath intervention alone and in combination with an exposure therapy for post-traumatic stress disorder and depression in survivors of the 2004 South-East Asia tsunami. Acta Psychiatr Scand. 2010;121(4):289-300.
15. Broocks A, Bandelow B, Pekrun G, et al. Comparison of aerobic exercise, clomipramine, and placebo in the treatment of panic disorder. Am J Psychiatry. 1998;155(5):603-609.
16. Merom D, Phongsavan P, Wagner R, et al. Promoting walking as an adjunct intervention to group cognitive behavioral therapy for anxiety disorders—a pilot group randomized trial. J Anxiety Disord. 2008;22(6):959-968.
17. Abrantes AM, Strong DR, Cohn A, et al. Acute changes in obsessions and compulsions following moderate-intensity aerobic exercise among patients with obsessive-compulsive disorder. J Anxiety Disord. 2009;23(7):923-927.
18. Lidren DM, Watkins PL, Gould RA, et al. A comparison of bibliotherapy and group therapy in the treatment of panic disorder. J Consult Clin Psychol. 1994;62(4):865-869.
19. Rapee RM, Abbott MJ, Lyneham HJ. Bibliotherapy for children with anxiety disorders using written materials for parents: a randomized controlled trial. J Consult Clin Psychol. 2006;74(3):436-444.
20. Abramowitz JS, Moore EL, Braddock AE, et al. Self-help cognitive-behavioral therapy with minimal therapist contact for social phobia: a controlled trial. J Behav Ther Exp Psychiatry. 2009;40(1):98-105.
21. Saeed SA, Bloch RM, Antonacci DJ. Herbal and dietary supplements for treatment of anxiety disorders. Am Fam Physician. 2007;76(4):549-556.
22. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-
23. Witte S, Loew D, Gaus W. Meta-analysis of the efficacy of the acetonic kava-kava extract WS1490 in patients with non-psychotic anxiety disorders. Phytother Res. 2005;19(3):183-188.
24. Sarris J, Kavanagh DJ, Byrne G, et al. The Kava Anxiety Depression Spectrum Study (KADSS): a randomized, placebo-controlled crossover trial using an aqueous extract of piper methysticum. Psychopharmacology (Berl). 2009;205:399-407.
25. Benjamin J, Levine J, Fux M, et al. Double-blind, placebo-controlled, crossover trial of inositol treatment for panic disorder. Am J Psychiatry. 1995;152(7):1084-1086.
26. Fux M, Levine J, Aviv A, et al. Inositol treatment of obsessive-compulsive disorder. Am J Psychiatry. 1996;153(9):1219-1221.
27. Fux M, Benjamin J, Belmaker RH. Inositol versus placebo augmentation of serotonin reuptake inhibitors in the treatment of obsessive-compulsive disorder: a double-blind cross-over study. Int J Neuropsychopharmcol. 1999;2(3):193-195.
28. Palatnik A, Frolov K, Fux M, et al. Double-blind, controlled, crossover trial of inositol versus fluvoxamine for the treatment of panic disorder. J Clin Psychopharmacol. 2001;21(3):335-339.
29. US Food and Drug Administration. Consumer advisory: kava-containing dietary supplements may be associated with severe injury. Available at: http://www.fda.gov/Food/ResourcesForYou/Consumers/ucm085482.htm. Accessed August 25, 2010.
30. Dragull K, Yoshida WY, Tang CS. Piperidine alkaloids from piper methysticum. Phytochemistry. 2003;63(2):193-198.
31. Simkins A, Thurston D, Colyar M, et al. Nature’s wrath? A closer look at complications with five popular herbs. Adv Nurse Pract. 2005;13(6):55-56.
32. Teschke R, Genthner A, Wolff A. Kava hepatotoxicity: comparison of aqueous, ethanolic, acetonic kava extracts and kava-herbs mixtures. J Ethnopharmacol. 2009;123(3):378-384.
33. Christl SU, Seifert A, Seeler D. Toxic hepatitis after consumption of traditional kava preparation. J Travel Med. 2009;16(1):55-56.
Schizophrenia in older adults
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/schizophrenia-in-older-adults.html#comments
Ms. M, age 68, seeks treatment for stress and anxiety after her sister has a stroke. Ms. M has chronic paranoid schizophrenia, and her sister has been Ms. M’s primary support since the onset of illness in her late 20s. Ms. M lives in a supported housing community. Her last psychiatric hospitalization was 16 years ago; for the past 15 years she has been stable on haloperidol, 20 mg/d. Ms. M also takes diphenhydramine, 50 mg at bedtime, to help her sleep.
Ms. M is hypertensive but does not have diabetes, obesity, or metabolic syndrome. She has mild executive dysfunction and mild extrapyramidal symptoms (EPS) but no tardive dyskinesia (TD). There is no evidence of delusions or hallucinations, although Ms. M is mildly paranoid about her neighbors. In the last year, she has been experiencing tremors and has fallen twice.
The number of older adults (age ≥65) who developed schizophrenia before age 45 is expected to double in the next 2 decades; the 1-year prevalence of schizophrenia among older adults is approximately 0.6%.1,2 This article reviews how positive, negative, and cognitive symptoms and social functioning change over decades and discusses strategies for reducing the impact of long-term antipsychotic use on neurologic and physical health. Although some patients experience schizophrenia onset later in life, in this article we focus on older adults who developed the illness before age 45.
Symptoms change with age
Positive symptoms of schizophrenia—hallucinations, delusions, and disorganized or catatonic behavior—do not “burn out” in most older adults.3 The severity of “day-to-day” psychotic symptoms appears reduced in patients with schizophrenia who have not had recent severe psychotic episodes. Aging-associated decrease in dopaminergic and other monoaminergic activities may explain this.
Some older adults experience sustained remission of positive symptoms and may no longer need antipsychotics.4 Factors that contribute to a better prognosis include:
- female sex
- developing the illness later in life (eg, fourth decade instead of second or third decade)
- being married
- obtaining appropriate treatment early in the illness.2
With treatment, positive symptoms can remit in 40% to 50% of older adults, especially those who have greater social support and fewer lifetime traumatic events.3,5
Negative symptoms—flat affect, social withdrawal, and decreased motivation—may become worse in older adults with a history of poor functioning (especially institutionalized patients) as they age.2,6 Changes in negative symptoms are more closely correlated with symptom chronicity, functional and cognitive impairment, soft neurologic signs such as impaired fine motor coordination, and institutionalization than with the patient’s age.7
Generalized cognitive deficits are ubiquitous in patients with schizophrenia and substantially impact community functioning.1,8 Cognitive function may worsen in older schizophrenia patients with a history of poor functioning—especially institutionalized patients—as they age.9 Most older adults with schizophrenia who reside in the community have persistent, but generally not progressive, cognitive deficits. Low education levels, poor premorbid function, and more severe positive symptoms at baseline are associated with worse cognitive functioning at all ages.2 Older adults with schizophrenia and TD have greater global cognitive deficits and greater deficits in learning than age-, education-, and subtype-matched schizophrenia patients without TD.1
Differences and similarities in cognitive impairment in older adults with schizophrenia compared with those who have Alzheimer’s disease (AD) are listed in Table 1. The course of cognitive deficits appears to be the most sensitive measure for determining whether a patient with long-standing schizophrenia has developed concomitant AD. Individuals with AD experience a more precipitous and progressive decline in cognitive function compared with patients with schizophrenia. Neuropsychological testing is recommended to accurately diagnose AD in older schizophrenia patients as early as possible.
Table 1
Cognitive impairment: Schizophrenia vs Alzheimer’s disease
Older patients with schizophrenia | Patients with AD |
---|---|
|
|
Common to both | |
| |
AD: Alzheimer’s disease; MMSE: Mini-Mental State Examination |
Depressive symptoms
More than two-fifths of older adults with schizophrenia show signs of clinical depression.10 Depression in this population is linked to positive symptoms, poor physical health, low income, and diminished network support. Routinely screen for depressive symptoms in older schizophrenia patients and institute prompt treatment as required. Assess these patients for suicide. Although suicide rates in schizophrenia patients decrease with age, they remain considerably higher than those of age-matched persons without schizophrenia.11
Social functioning
Improvement or deterioration in social functioning is possible as patients with schizophrenia age.11 Compared with age-matched patients in the general population and those with bipolar disorder, older adults with schizophrenia need more help with activities of daily living (eg, looking after the home, using public transportation).11
Cognitive impairment seems to be the most important predictor of social functioning in patients with schizophrenia at any age. Impaired social functioning also is associated with negative symptoms and movement disorders. On community integration measures (how well the person lives, participates, and socializes in his or her community), older adults with schizophrenia do roughly half as well as their age-matched peers in the general community.3 Older schizophrenia patients’ social networks seem to be smaller than those of their age-matched peers,1 but they may experience fewer discordant interactions, such as situations with high expressed emotions. Increased psychological resilience may help older adults better adapt to changes as they age (Box).1,12
Psychological resilience factors—such as coping skills and self-efficacy—play an important role in an individual’s ability to adapt to life stressors associated with schizophrenia and old age. One study found that a strategy of fighting back unwanted thoughts was negatively related to age, whereas acceptance and diversion were positively correlated with age, which suggests increased resilience in older adults with schizophrenia.12 Coping skills seem to improve with aging and older patients may become more active participants in their recovery.1 Routine clinical care of older adults with schizophrenia should focus on identifying and supporting factors that promote resilience in addition to the standard “problems-centered” approach that focuses on treating positive symptoms.
A complex assessment
Older adults with schizophrenia have an increased prevalence of:
- obesity
- diabetes
- hyperlipidemia
- coronary artery disease
- myocardial infarction
- limited mobility
- illnesses related to smoking or substance abuse.13,14
The severity of these conditions often is greater in older adults with schizophrenia compared with age-matched controls. Older adults with schizophrenia also have poorer access to and use of health care services and compliance with treatment regimens, and receive a lower quality of care. The incidence of physical health and neuropsychiatric problems increases with age and older adults with schizophrenia with poor functioning may be less able to recognize and report symptoms to health care providers.
Because these patients have complex health care concerns, we recommend using a checklist to help make routine visits thorough and identify and treat problems. Click here for a downloadable assessment checklist. Ideally, the initial assessment should use an interdisciplinary approach that includes the patient, family/knowledgeable informants, psychiatrist, psychiatric nurse practitioner/physician assistant, social worker, caseworker, chaplain (if appropriate), and a nurse. The initial assessment may take 2 to 3 visits to complete.
Adapting treatment
Older adults with schizophrenia can benefit from the psychopharmacologic and psychosocial interventions used for younger patients (Table 2).15,16 However, you may need to adapt these treatments to accommodate cognitive impairment, medical comorbidities, or hearing and vision deficits. The most appropriate goal may not be recovery or rehabilitation, but making life more meaningful and satisfying for the patient and his or her family.
Pharmacotherapy. The 2009 schizophrenia Patient Outcomes Research Team (PORT) psychopharmacology treatment recommendations may be used for older adults.15 Most adverse effects of antipsychotics (except for dystonia) are more prevalent in older adults than in their younger counterparts. In general, second-generation antipsychotics (SGAs) are preferred over first-generation antipsychotics (FGAs) for treating positive symptoms in older patients because of SGAs’ lower risk of TD and EPS despite the increased risk of metabolic disorders.
Aripiprazole and ziprasidone are associated with significantly less risk of metabolic disorders and may be preferred in older adults who have diabetes, obesity, or hyperlipidemia. Aripiprazole has the lowest risk of QTc prolongation and may be preferred in patients who have prolonged QTc interval.15 Quetiapine and clozapine are associated with the lowest risk of EPS. Among SGAs, aripiprazole is associated with the lowest risk of prolactin elevation and sexual side effects and may be preferred in older adults who complain of sexual dysfunction or have osteoporosis.
Because rates of EPS and TDs may exceed 50% among older patients, many experts encourage clinicians to taper anti-psychotic dosages in patients with stable chronic symptoms. Tapering dosages may be critically important because EPS may affect functional performance more than positive or negative symptoms or duration of psychoses.
The severity of TD in older adults with schizophrenia may be masked because many patients receive high doses of FGAs. When a patient’s FGA dosage is reduced to manage EPS, subclinical TD may manifest for the first time, and existing TD may become noticeably worse. Consider clozapine for long-term management of older adults who have TD; however, burdens of its use—such as weekly blood draws and anticholinergic adverse effects—may limit its use in older adults.
Lowering the anticholinergic load by reducing the dosage of drugs with anti-cholinergic activity or discontinuing anti-cholinergic medications when possible is a key component of treating older adults with schizophrenia. Doing so may improve not only patients’ cognitive function but also their quality of life by reducing other anticholinergic adverse effects, such as constipation, blurred vision, and urinary retention.
Psychosocial interventions. The 2009 schizophrenia PORT psychosocial treatment recommendations and summary statements may be followed for older adults. Recommended interventions include:
- assertive community treatment
- supported employment
- cognitive-behavioral therapy (CBT)
- family-based services
- token economy
- skills training.16
Social skills training with or without CBT can be successfully adapted for older adults. Such interventions can improve social functioning and everyday living skills. Environmental modifications—such as removing decorative mirrors from the home of a delusional patient who believes people are living in the walls—may alleviate distress.
In addition, social contact and structured activities such as group exercises may benefit older patients. Educate care-givers about ways they can work with older adults, such as distracting them or not directly challenging false beliefs. Comprehensive psychosocial interventions also can improve health care management skills.
Preliminary data indicate that CBT and skills training with role-plays, structured feedback, and homework assignments can improve quality of life of older adults with schizophrenia.17,18 Functional Adaptation Skills Training focuses on medication management, social skills, communication skills, organization, planning, and financial management.19 Those who received this training showed improvement in their functional skills that persisted for at least 3 months after treatment ended.20
Poor adherence to medication is common in older schizophrenia patients and has devastating consequences. Adherence problems are complex and often have multiple causes, which requires customized interventions that target specific causes. Patients who receive a combination of psychosocial treatment and antipsychotics are more likely to be compliant with their medication and less likely to relapse or be hospitalized.16
Addressing the social stigma associated with schizophrenia may help reduce the social isolation and loneliness that many older adults experience. Psychiatrists can help fight stigma by participating in community educational programs and encouraging patients’ families to become involved in support and advocacy organizations.
Table 2
Interventions for older adults with schizophrenia
Symptom/problem | Intervention(s) |
---|---|
Positive symptoms | Second-generation antipsychotics, CBT, caregiver education |
Negative symptoms | Second-generation antipsychotics, caregiver education, token economy |
Cognitive symptoms | Reducing anticholinergic load, cognitive remediation |
Social deficits | Social skills training, FAST |
Depression | Antidepressants, CBT |
Mobility limitations | Gait and balance strengthening exercises, physical therapy |
Vascular risk factors | Second-generation antipsychotic with lowest risk of weight gain and hyperlipidemia, such as aripiprazole or ziprasidone |
Cigarette smoking | Smoking cessation program |
Severe tardive dyskinesia | Consider clozapine |
Extrapyramidal symptoms | Second-generation antipsychotics with lowest risk of extrapyramidal symptoms, such as quetiapine or clozapine |
Homelessness | Supported housing |
Progressive cognitive decline | Dementia workup |
Treatment nonadherence | Caregiver education, FAST, ACT |
Caregiver stress | Caregiver education, support groups, psychotherapy |
ACT: assertive community treatment; CBT: cognitive-behavioral therapy; FAST: functional adaptation skills training | |
Source: References 15,16 |
CASE CONTINUED: Medication changes
Ms. M’s psychiatrist tells her that her problems with tremors and falls are most likely caused by haloperidol and recommends a slow dosage reduction and discontinuing diphenhydramine. Haloperidol is decreased to 10 mg/d for 1 month and then to 5 mg/d. Diphenhydramine is decreased to 25 mg at bedtime for 7 days and then stopped.
Ms. M declines physical therapy but agrees to participate in strength and balance training offered at the supported housing community 3 times a week for 4 weeks. Tremors resolve over the next month and Ms. M has not fallen since.
Ms. M complains of insomnia and is reluctant to further decrease haloperidol unless she is prescribed a different antipsychotic and given something to help her sleep. Ms. M is started on quetiapine, 25 mg/d at bedtime. Over 3 weeks, the dosage is increased to 100 mg/d. Ms. M tolerates quetiapine well and her sleep improves. Haloperidol is then decreased to 2.5 mg/d for 1 month and then discontinued. Ms. M also is offered supportive psychotherapy every 2 weeks to address her paranoia and stress. She continues to do well on quetiapine and supportive psychotherapy.
Related Resources
- Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc; 2003.
- Mental health: a report of the Surgeon General. Chapter 5: other mental disorders in older adults; schizophrenia in late life. www.surgeongeneral.gov/library/mentalhealth/chapter5/sec5.html.
Drug Brand Names
- Aripiprazole • Abilify
- Clozapine • Clozaril
- Diphenhydramine • Sominex, Unisom, others
- Haloperidol • Haldol
- Quetiapine • Seroquel
- Ziprasidone • Geodon
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
1. Cohen CI, Cohen GD, Blank K, et al. Schizophrenia and older adults. An overview: directions for research and policy. Am J Geriatr Psychiatry. 2000;8:19-28.
2. Vahia I, Bankole AO, Diwan S, et al. Schizophrenia in late life. Aging Health. 2007;3:393-396.
3. Cohen CI, Pathak R, Ramirez PM, et al. Outcome among community dwelling older adults with schizophrenia: results using five conceptual models. Community Ment Health J. 2009;45:151-156.
4. Ausland LA, Jeste DV. Sustained remission of schizophrenia among community-dwelling older outpatients. Am J Psychiatry. 2004;161:1490-1493.
5. Bankole A, Cohen CI, Vahia I, et al. Symptomatic remission in a multiracial urban population of older adults with schizophrenia. Am J Geriatr Psychiatry. 2008;16(12):966-973.
6. Harding CM. Changes in schizophrenia across time. In: Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc.; 2003: 19–41.
7. Harvey PD, Howanitz E, Parrella M, et al. Symptoms, cognitive functioning, and adaptive skills in geriatric patients with lifelong schizophrenia: a comparison across treatment sites. Am J Psychiatry. 1998;155:1080-1086.
8. Morrison G, O’Carroll,, McCreadie R. Long-term course of cognitive impairment in schizophrenia. Br J Psychiatry. 2006;189:556-557.
9. Bowie CR, Tsapelas I, Friedman J, et al. The longitudinal course of thought disorder in geriatric patients with chronic schizophrenia. Am J Psychiatry. 2005;162(4):793-795.
10. Diwan S, Cohen CI, Bankole AO, et al. Depression in older adults with schizophrenia spectrum disorders: prevalence and associated factors. Am J Geriatr Psychiatry. 2007;15:991-998.
11. Berry K, Barrowclough C. The needs of older adults with schizophrenia. Implications for psychological interventions. Clin Psych Review. 2009;29:68-76.
12. Cohen CI. Age-related correlations in patient symptom management strategies in schizophrenia: an exploratory study. Int J Geriatr Psychiatry. 1993;8:211-213.
13. Leucht S, Burkard T, Henderson J, et al. Physical illness and schizophrenia: a review of the literature. Acta Psychiatr Scand. 2007;116(5):317-333.
14. Cohen CI, Vahia I, Reyes P, et al. Schizophrenia in later life: clinical symptoms and social well-being. Psychiatr Serv. 2008;59:232-234.
15. Buchanan RW, Kreyenbuhl J, Kelly DL, et al. The 2009 Schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36:71-93.
16. Dixon LB, Dickerson F, Bellack AS, et al. The 2009 Schizophrenia PORT psychosocial treatment recommendations and summary statements. Schizophr Bull. 2010;36:48-70.
17. McQuaid JR, Granholm E, McClure FS, et al. Development of an integrated cognitive-behavioral and social skills training intervention for older patients with schizophrenia. J Psychother Pract Res. 2000;9:149-156.
18. Granholm E, McQuaid JR, McClure FS, et al. A randomized controlled pilot study of cognitive behavioral social skills training for older patients with schizophrenia. Schizophr Res. 2002;53:167-169.
19. Patterson TL, McKibbin C, Taylor M, et al. Functional adaptation skills training (FAST): a pilot psychosocial intervention study in middle-aged and older patients with chronic psychiatric disorders. Am J Geriatr Psychiatry. 2003;11:17-23.
20. Patterson TL, Goldman S, McKibbin CL, et al. UCSD performance-based skills assessment: development of a new measure of everyday functioning for severely mentally ill adults. Schizophr Bull. 2001;27:235-245.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/schizophrenia-in-older-adults.html#comments
Ms. M, age 68, seeks treatment for stress and anxiety after her sister has a stroke. Ms. M has chronic paranoid schizophrenia, and her sister has been Ms. M’s primary support since the onset of illness in her late 20s. Ms. M lives in a supported housing community. Her last psychiatric hospitalization was 16 years ago; for the past 15 years she has been stable on haloperidol, 20 mg/d. Ms. M also takes diphenhydramine, 50 mg at bedtime, to help her sleep.
Ms. M is hypertensive but does not have diabetes, obesity, or metabolic syndrome. She has mild executive dysfunction and mild extrapyramidal symptoms (EPS) but no tardive dyskinesia (TD). There is no evidence of delusions or hallucinations, although Ms. M is mildly paranoid about her neighbors. In the last year, she has been experiencing tremors and has fallen twice.
The number of older adults (age ≥65) who developed schizophrenia before age 45 is expected to double in the next 2 decades; the 1-year prevalence of schizophrenia among older adults is approximately 0.6%.1,2 This article reviews how positive, negative, and cognitive symptoms and social functioning change over decades and discusses strategies for reducing the impact of long-term antipsychotic use on neurologic and physical health. Although some patients experience schizophrenia onset later in life, in this article we focus on older adults who developed the illness before age 45.
Symptoms change with age
Positive symptoms of schizophrenia—hallucinations, delusions, and disorganized or catatonic behavior—do not “burn out” in most older adults.3 The severity of “day-to-day” psychotic symptoms appears reduced in patients with schizophrenia who have not had recent severe psychotic episodes. Aging-associated decrease in dopaminergic and other monoaminergic activities may explain this.
Some older adults experience sustained remission of positive symptoms and may no longer need antipsychotics.4 Factors that contribute to a better prognosis include:
- female sex
- developing the illness later in life (eg, fourth decade instead of second or third decade)
- being married
- obtaining appropriate treatment early in the illness.2
With treatment, positive symptoms can remit in 40% to 50% of older adults, especially those who have greater social support and fewer lifetime traumatic events.3,5
Negative symptoms—flat affect, social withdrawal, and decreased motivation—may become worse in older adults with a history of poor functioning (especially institutionalized patients) as they age.2,6 Changes in negative symptoms are more closely correlated with symptom chronicity, functional and cognitive impairment, soft neurologic signs such as impaired fine motor coordination, and institutionalization than with the patient’s age.7
Generalized cognitive deficits are ubiquitous in patients with schizophrenia and substantially impact community functioning.1,8 Cognitive function may worsen in older schizophrenia patients with a history of poor functioning—especially institutionalized patients—as they age.9 Most older adults with schizophrenia who reside in the community have persistent, but generally not progressive, cognitive deficits. Low education levels, poor premorbid function, and more severe positive symptoms at baseline are associated with worse cognitive functioning at all ages.2 Older adults with schizophrenia and TD have greater global cognitive deficits and greater deficits in learning than age-, education-, and subtype-matched schizophrenia patients without TD.1
Differences and similarities in cognitive impairment in older adults with schizophrenia compared with those who have Alzheimer’s disease (AD) are listed in Table 1. The course of cognitive deficits appears to be the most sensitive measure for determining whether a patient with long-standing schizophrenia has developed concomitant AD. Individuals with AD experience a more precipitous and progressive decline in cognitive function compared with patients with schizophrenia. Neuropsychological testing is recommended to accurately diagnose AD in older schizophrenia patients as early as possible.
Table 1
Cognitive impairment: Schizophrenia vs Alzheimer’s disease
Older patients with schizophrenia | Patients with AD |
---|---|
|
|
Common to both | |
| |
AD: Alzheimer’s disease; MMSE: Mini-Mental State Examination |
Depressive symptoms
More than two-fifths of older adults with schizophrenia show signs of clinical depression.10 Depression in this population is linked to positive symptoms, poor physical health, low income, and diminished network support. Routinely screen for depressive symptoms in older schizophrenia patients and institute prompt treatment as required. Assess these patients for suicide. Although suicide rates in schizophrenia patients decrease with age, they remain considerably higher than those of age-matched persons without schizophrenia.11
Social functioning
Improvement or deterioration in social functioning is possible as patients with schizophrenia age.11 Compared with age-matched patients in the general population and those with bipolar disorder, older adults with schizophrenia need more help with activities of daily living (eg, looking after the home, using public transportation).11
Cognitive impairment seems to be the most important predictor of social functioning in patients with schizophrenia at any age. Impaired social functioning also is associated with negative symptoms and movement disorders. On community integration measures (how well the person lives, participates, and socializes in his or her community), older adults with schizophrenia do roughly half as well as their age-matched peers in the general community.3 Older schizophrenia patients’ social networks seem to be smaller than those of their age-matched peers,1 but they may experience fewer discordant interactions, such as situations with high expressed emotions. Increased psychological resilience may help older adults better adapt to changes as they age (Box).1,12
Psychological resilience factors—such as coping skills and self-efficacy—play an important role in an individual’s ability to adapt to life stressors associated with schizophrenia and old age. One study found that a strategy of fighting back unwanted thoughts was negatively related to age, whereas acceptance and diversion were positively correlated with age, which suggests increased resilience in older adults with schizophrenia.12 Coping skills seem to improve with aging and older patients may become more active participants in their recovery.1 Routine clinical care of older adults with schizophrenia should focus on identifying and supporting factors that promote resilience in addition to the standard “problems-centered” approach that focuses on treating positive symptoms.
A complex assessment
Older adults with schizophrenia have an increased prevalence of:
- obesity
- diabetes
- hyperlipidemia
- coronary artery disease
- myocardial infarction
- limited mobility
- illnesses related to smoking or substance abuse.13,14
The severity of these conditions often is greater in older adults with schizophrenia compared with age-matched controls. Older adults with schizophrenia also have poorer access to and use of health care services and compliance with treatment regimens, and receive a lower quality of care. The incidence of physical health and neuropsychiatric problems increases with age and older adults with schizophrenia with poor functioning may be less able to recognize and report symptoms to health care providers.
Because these patients have complex health care concerns, we recommend using a checklist to help make routine visits thorough and identify and treat problems. Click here for a downloadable assessment checklist. Ideally, the initial assessment should use an interdisciplinary approach that includes the patient, family/knowledgeable informants, psychiatrist, psychiatric nurse practitioner/physician assistant, social worker, caseworker, chaplain (if appropriate), and a nurse. The initial assessment may take 2 to 3 visits to complete.
Adapting treatment
Older adults with schizophrenia can benefit from the psychopharmacologic and psychosocial interventions used for younger patients (Table 2).15,16 However, you may need to adapt these treatments to accommodate cognitive impairment, medical comorbidities, or hearing and vision deficits. The most appropriate goal may not be recovery or rehabilitation, but making life more meaningful and satisfying for the patient and his or her family.
Pharmacotherapy. The 2009 schizophrenia Patient Outcomes Research Team (PORT) psychopharmacology treatment recommendations may be used for older adults.15 Most adverse effects of antipsychotics (except for dystonia) are more prevalent in older adults than in their younger counterparts. In general, second-generation antipsychotics (SGAs) are preferred over first-generation antipsychotics (FGAs) for treating positive symptoms in older patients because of SGAs’ lower risk of TD and EPS despite the increased risk of metabolic disorders.
Aripiprazole and ziprasidone are associated with significantly less risk of metabolic disorders and may be preferred in older adults who have diabetes, obesity, or hyperlipidemia. Aripiprazole has the lowest risk of QTc prolongation and may be preferred in patients who have prolonged QTc interval.15 Quetiapine and clozapine are associated with the lowest risk of EPS. Among SGAs, aripiprazole is associated with the lowest risk of prolactin elevation and sexual side effects and may be preferred in older adults who complain of sexual dysfunction or have osteoporosis.
Because rates of EPS and TDs may exceed 50% among older patients, many experts encourage clinicians to taper anti-psychotic dosages in patients with stable chronic symptoms. Tapering dosages may be critically important because EPS may affect functional performance more than positive or negative symptoms or duration of psychoses.
The severity of TD in older adults with schizophrenia may be masked because many patients receive high doses of FGAs. When a patient’s FGA dosage is reduced to manage EPS, subclinical TD may manifest for the first time, and existing TD may become noticeably worse. Consider clozapine for long-term management of older adults who have TD; however, burdens of its use—such as weekly blood draws and anticholinergic adverse effects—may limit its use in older adults.
Lowering the anticholinergic load by reducing the dosage of drugs with anti-cholinergic activity or discontinuing anti-cholinergic medications when possible is a key component of treating older adults with schizophrenia. Doing so may improve not only patients’ cognitive function but also their quality of life by reducing other anticholinergic adverse effects, such as constipation, blurred vision, and urinary retention.
Psychosocial interventions. The 2009 schizophrenia PORT psychosocial treatment recommendations and summary statements may be followed for older adults. Recommended interventions include:
- assertive community treatment
- supported employment
- cognitive-behavioral therapy (CBT)
- family-based services
- token economy
- skills training.16
Social skills training with or without CBT can be successfully adapted for older adults. Such interventions can improve social functioning and everyday living skills. Environmental modifications—such as removing decorative mirrors from the home of a delusional patient who believes people are living in the walls—may alleviate distress.
In addition, social contact and structured activities such as group exercises may benefit older patients. Educate care-givers about ways they can work with older adults, such as distracting them or not directly challenging false beliefs. Comprehensive psychosocial interventions also can improve health care management skills.
Preliminary data indicate that CBT and skills training with role-plays, structured feedback, and homework assignments can improve quality of life of older adults with schizophrenia.17,18 Functional Adaptation Skills Training focuses on medication management, social skills, communication skills, organization, planning, and financial management.19 Those who received this training showed improvement in their functional skills that persisted for at least 3 months after treatment ended.20
Poor adherence to medication is common in older schizophrenia patients and has devastating consequences. Adherence problems are complex and often have multiple causes, which requires customized interventions that target specific causes. Patients who receive a combination of psychosocial treatment and antipsychotics are more likely to be compliant with their medication and less likely to relapse or be hospitalized.16
Addressing the social stigma associated with schizophrenia may help reduce the social isolation and loneliness that many older adults experience. Psychiatrists can help fight stigma by participating in community educational programs and encouraging patients’ families to become involved in support and advocacy organizations.
Table 2
Interventions for older adults with schizophrenia
Symptom/problem | Intervention(s) |
---|---|
Positive symptoms | Second-generation antipsychotics, CBT, caregiver education |
Negative symptoms | Second-generation antipsychotics, caregiver education, token economy |
Cognitive symptoms | Reducing anticholinergic load, cognitive remediation |
Social deficits | Social skills training, FAST |
Depression | Antidepressants, CBT |
Mobility limitations | Gait and balance strengthening exercises, physical therapy |
Vascular risk factors | Second-generation antipsychotic with lowest risk of weight gain and hyperlipidemia, such as aripiprazole or ziprasidone |
Cigarette smoking | Smoking cessation program |
Severe tardive dyskinesia | Consider clozapine |
Extrapyramidal symptoms | Second-generation antipsychotics with lowest risk of extrapyramidal symptoms, such as quetiapine or clozapine |
Homelessness | Supported housing |
Progressive cognitive decline | Dementia workup |
Treatment nonadherence | Caregiver education, FAST, ACT |
Caregiver stress | Caregiver education, support groups, psychotherapy |
ACT: assertive community treatment; CBT: cognitive-behavioral therapy; FAST: functional adaptation skills training | |
Source: References 15,16 |
CASE CONTINUED: Medication changes
Ms. M’s psychiatrist tells her that her problems with tremors and falls are most likely caused by haloperidol and recommends a slow dosage reduction and discontinuing diphenhydramine. Haloperidol is decreased to 10 mg/d for 1 month and then to 5 mg/d. Diphenhydramine is decreased to 25 mg at bedtime for 7 days and then stopped.
Ms. M declines physical therapy but agrees to participate in strength and balance training offered at the supported housing community 3 times a week for 4 weeks. Tremors resolve over the next month and Ms. M has not fallen since.
Ms. M complains of insomnia and is reluctant to further decrease haloperidol unless she is prescribed a different antipsychotic and given something to help her sleep. Ms. M is started on quetiapine, 25 mg/d at bedtime. Over 3 weeks, the dosage is increased to 100 mg/d. Ms. M tolerates quetiapine well and her sleep improves. Haloperidol is then decreased to 2.5 mg/d for 1 month and then discontinued. Ms. M also is offered supportive psychotherapy every 2 weeks to address her paranoia and stress. She continues to do well on quetiapine and supportive psychotherapy.
Related Resources
- Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc; 2003.
- Mental health: a report of the Surgeon General. Chapter 5: other mental disorders in older adults; schizophrenia in late life. www.surgeongeneral.gov/library/mentalhealth/chapter5/sec5.html.
Drug Brand Names
- Aripiprazole • Abilify
- Clozapine • Clozaril
- Diphenhydramine • Sominex, Unisom, others
- Haloperidol • Haldol
- Quetiapine • Seroquel
- Ziprasidone • Geodon
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/schizophrenia-in-older-adults.html#comments
Ms. M, age 68, seeks treatment for stress and anxiety after her sister has a stroke. Ms. M has chronic paranoid schizophrenia, and her sister has been Ms. M’s primary support since the onset of illness in her late 20s. Ms. M lives in a supported housing community. Her last psychiatric hospitalization was 16 years ago; for the past 15 years she has been stable on haloperidol, 20 mg/d. Ms. M also takes diphenhydramine, 50 mg at bedtime, to help her sleep.
Ms. M is hypertensive but does not have diabetes, obesity, or metabolic syndrome. She has mild executive dysfunction and mild extrapyramidal symptoms (EPS) but no tardive dyskinesia (TD). There is no evidence of delusions or hallucinations, although Ms. M is mildly paranoid about her neighbors. In the last year, she has been experiencing tremors and has fallen twice.
The number of older adults (age ≥65) who developed schizophrenia before age 45 is expected to double in the next 2 decades; the 1-year prevalence of schizophrenia among older adults is approximately 0.6%.1,2 This article reviews how positive, negative, and cognitive symptoms and social functioning change over decades and discusses strategies for reducing the impact of long-term antipsychotic use on neurologic and physical health. Although some patients experience schizophrenia onset later in life, in this article we focus on older adults who developed the illness before age 45.
Symptoms change with age
Positive symptoms of schizophrenia—hallucinations, delusions, and disorganized or catatonic behavior—do not “burn out” in most older adults.3 The severity of “day-to-day” psychotic symptoms appears reduced in patients with schizophrenia who have not had recent severe psychotic episodes. Aging-associated decrease in dopaminergic and other monoaminergic activities may explain this.
Some older adults experience sustained remission of positive symptoms and may no longer need antipsychotics.4 Factors that contribute to a better prognosis include:
- female sex
- developing the illness later in life (eg, fourth decade instead of second or third decade)
- being married
- obtaining appropriate treatment early in the illness.2
With treatment, positive symptoms can remit in 40% to 50% of older adults, especially those who have greater social support and fewer lifetime traumatic events.3,5
Negative symptoms—flat affect, social withdrawal, and decreased motivation—may become worse in older adults with a history of poor functioning (especially institutionalized patients) as they age.2,6 Changes in negative symptoms are more closely correlated with symptom chronicity, functional and cognitive impairment, soft neurologic signs such as impaired fine motor coordination, and institutionalization than with the patient’s age.7
Generalized cognitive deficits are ubiquitous in patients with schizophrenia and substantially impact community functioning.1,8 Cognitive function may worsen in older schizophrenia patients with a history of poor functioning—especially institutionalized patients—as they age.9 Most older adults with schizophrenia who reside in the community have persistent, but generally not progressive, cognitive deficits. Low education levels, poor premorbid function, and more severe positive symptoms at baseline are associated with worse cognitive functioning at all ages.2 Older adults with schizophrenia and TD have greater global cognitive deficits and greater deficits in learning than age-, education-, and subtype-matched schizophrenia patients without TD.1
Differences and similarities in cognitive impairment in older adults with schizophrenia compared with those who have Alzheimer’s disease (AD) are listed in Table 1. The course of cognitive deficits appears to be the most sensitive measure for determining whether a patient with long-standing schizophrenia has developed concomitant AD. Individuals with AD experience a more precipitous and progressive decline in cognitive function compared with patients with schizophrenia. Neuropsychological testing is recommended to accurately diagnose AD in older schizophrenia patients as early as possible.
Table 1
Cognitive impairment: Schizophrenia vs Alzheimer’s disease
Older patients with schizophrenia | Patients with AD |
---|---|
|
|
Common to both | |
| |
AD: Alzheimer’s disease; MMSE: Mini-Mental State Examination |
Depressive symptoms
More than two-fifths of older adults with schizophrenia show signs of clinical depression.10 Depression in this population is linked to positive symptoms, poor physical health, low income, and diminished network support. Routinely screen for depressive symptoms in older schizophrenia patients and institute prompt treatment as required. Assess these patients for suicide. Although suicide rates in schizophrenia patients decrease with age, they remain considerably higher than those of age-matched persons without schizophrenia.11
Social functioning
Improvement or deterioration in social functioning is possible as patients with schizophrenia age.11 Compared with age-matched patients in the general population and those with bipolar disorder, older adults with schizophrenia need more help with activities of daily living (eg, looking after the home, using public transportation).11
Cognitive impairment seems to be the most important predictor of social functioning in patients with schizophrenia at any age. Impaired social functioning also is associated with negative symptoms and movement disorders. On community integration measures (how well the person lives, participates, and socializes in his or her community), older adults with schizophrenia do roughly half as well as their age-matched peers in the general community.3 Older schizophrenia patients’ social networks seem to be smaller than those of their age-matched peers,1 but they may experience fewer discordant interactions, such as situations with high expressed emotions. Increased psychological resilience may help older adults better adapt to changes as they age (Box).1,12
Psychological resilience factors—such as coping skills and self-efficacy—play an important role in an individual’s ability to adapt to life stressors associated with schizophrenia and old age. One study found that a strategy of fighting back unwanted thoughts was negatively related to age, whereas acceptance and diversion were positively correlated with age, which suggests increased resilience in older adults with schizophrenia.12 Coping skills seem to improve with aging and older patients may become more active participants in their recovery.1 Routine clinical care of older adults with schizophrenia should focus on identifying and supporting factors that promote resilience in addition to the standard “problems-centered” approach that focuses on treating positive symptoms.
A complex assessment
Older adults with schizophrenia have an increased prevalence of:
- obesity
- diabetes
- hyperlipidemia
- coronary artery disease
- myocardial infarction
- limited mobility
- illnesses related to smoking or substance abuse.13,14
The severity of these conditions often is greater in older adults with schizophrenia compared with age-matched controls. Older adults with schizophrenia also have poorer access to and use of health care services and compliance with treatment regimens, and receive a lower quality of care. The incidence of physical health and neuropsychiatric problems increases with age and older adults with schizophrenia with poor functioning may be less able to recognize and report symptoms to health care providers.
Because these patients have complex health care concerns, we recommend using a checklist to help make routine visits thorough and identify and treat problems. Click here for a downloadable assessment checklist. Ideally, the initial assessment should use an interdisciplinary approach that includes the patient, family/knowledgeable informants, psychiatrist, psychiatric nurse practitioner/physician assistant, social worker, caseworker, chaplain (if appropriate), and a nurse. The initial assessment may take 2 to 3 visits to complete.
Adapting treatment
Older adults with schizophrenia can benefit from the psychopharmacologic and psychosocial interventions used for younger patients (Table 2).15,16 However, you may need to adapt these treatments to accommodate cognitive impairment, medical comorbidities, or hearing and vision deficits. The most appropriate goal may not be recovery or rehabilitation, but making life more meaningful and satisfying for the patient and his or her family.
Pharmacotherapy. The 2009 schizophrenia Patient Outcomes Research Team (PORT) psychopharmacology treatment recommendations may be used for older adults.15 Most adverse effects of antipsychotics (except for dystonia) are more prevalent in older adults than in their younger counterparts. In general, second-generation antipsychotics (SGAs) are preferred over first-generation antipsychotics (FGAs) for treating positive symptoms in older patients because of SGAs’ lower risk of TD and EPS despite the increased risk of metabolic disorders.
Aripiprazole and ziprasidone are associated with significantly less risk of metabolic disorders and may be preferred in older adults who have diabetes, obesity, or hyperlipidemia. Aripiprazole has the lowest risk of QTc prolongation and may be preferred in patients who have prolonged QTc interval.15 Quetiapine and clozapine are associated with the lowest risk of EPS. Among SGAs, aripiprazole is associated with the lowest risk of prolactin elevation and sexual side effects and may be preferred in older adults who complain of sexual dysfunction or have osteoporosis.
Because rates of EPS and TDs may exceed 50% among older patients, many experts encourage clinicians to taper anti-psychotic dosages in patients with stable chronic symptoms. Tapering dosages may be critically important because EPS may affect functional performance more than positive or negative symptoms or duration of psychoses.
The severity of TD in older adults with schizophrenia may be masked because many patients receive high doses of FGAs. When a patient’s FGA dosage is reduced to manage EPS, subclinical TD may manifest for the first time, and existing TD may become noticeably worse. Consider clozapine for long-term management of older adults who have TD; however, burdens of its use—such as weekly blood draws and anticholinergic adverse effects—may limit its use in older adults.
Lowering the anticholinergic load by reducing the dosage of drugs with anti-cholinergic activity or discontinuing anti-cholinergic medications when possible is a key component of treating older adults with schizophrenia. Doing so may improve not only patients’ cognitive function but also their quality of life by reducing other anticholinergic adverse effects, such as constipation, blurred vision, and urinary retention.
Psychosocial interventions. The 2009 schizophrenia PORT psychosocial treatment recommendations and summary statements may be followed for older adults. Recommended interventions include:
- assertive community treatment
- supported employment
- cognitive-behavioral therapy (CBT)
- family-based services
- token economy
- skills training.16
Social skills training with or without CBT can be successfully adapted for older adults. Such interventions can improve social functioning and everyday living skills. Environmental modifications—such as removing decorative mirrors from the home of a delusional patient who believes people are living in the walls—may alleviate distress.
In addition, social contact and structured activities such as group exercises may benefit older patients. Educate care-givers about ways they can work with older adults, such as distracting them or not directly challenging false beliefs. Comprehensive psychosocial interventions also can improve health care management skills.
Preliminary data indicate that CBT and skills training with role-plays, structured feedback, and homework assignments can improve quality of life of older adults with schizophrenia.17,18 Functional Adaptation Skills Training focuses on medication management, social skills, communication skills, organization, planning, and financial management.19 Those who received this training showed improvement in their functional skills that persisted for at least 3 months after treatment ended.20
Poor adherence to medication is common in older schizophrenia patients and has devastating consequences. Adherence problems are complex and often have multiple causes, which requires customized interventions that target specific causes. Patients who receive a combination of psychosocial treatment and antipsychotics are more likely to be compliant with their medication and less likely to relapse or be hospitalized.16
Addressing the social stigma associated with schizophrenia may help reduce the social isolation and loneliness that many older adults experience. Psychiatrists can help fight stigma by participating in community educational programs and encouraging patients’ families to become involved in support and advocacy organizations.
Table 2
Interventions for older adults with schizophrenia
Symptom/problem | Intervention(s) |
---|---|
Positive symptoms | Second-generation antipsychotics, CBT, caregiver education |
Negative symptoms | Second-generation antipsychotics, caregiver education, token economy |
Cognitive symptoms | Reducing anticholinergic load, cognitive remediation |
Social deficits | Social skills training, FAST |
Depression | Antidepressants, CBT |
Mobility limitations | Gait and balance strengthening exercises, physical therapy |
Vascular risk factors | Second-generation antipsychotic with lowest risk of weight gain and hyperlipidemia, such as aripiprazole or ziprasidone |
Cigarette smoking | Smoking cessation program |
Severe tardive dyskinesia | Consider clozapine |
Extrapyramidal symptoms | Second-generation antipsychotics with lowest risk of extrapyramidal symptoms, such as quetiapine or clozapine |
Homelessness | Supported housing |
Progressive cognitive decline | Dementia workup |
Treatment nonadherence | Caregiver education, FAST, ACT |
Caregiver stress | Caregiver education, support groups, psychotherapy |
ACT: assertive community treatment; CBT: cognitive-behavioral therapy; FAST: functional adaptation skills training | |
Source: References 15,16 |
CASE CONTINUED: Medication changes
Ms. M’s psychiatrist tells her that her problems with tremors and falls are most likely caused by haloperidol and recommends a slow dosage reduction and discontinuing diphenhydramine. Haloperidol is decreased to 10 mg/d for 1 month and then to 5 mg/d. Diphenhydramine is decreased to 25 mg at bedtime for 7 days and then stopped.
Ms. M declines physical therapy but agrees to participate in strength and balance training offered at the supported housing community 3 times a week for 4 weeks. Tremors resolve over the next month and Ms. M has not fallen since.
Ms. M complains of insomnia and is reluctant to further decrease haloperidol unless she is prescribed a different antipsychotic and given something to help her sleep. Ms. M is started on quetiapine, 25 mg/d at bedtime. Over 3 weeks, the dosage is increased to 100 mg/d. Ms. M tolerates quetiapine well and her sleep improves. Haloperidol is then decreased to 2.5 mg/d for 1 month and then discontinued. Ms. M also is offered supportive psychotherapy every 2 weeks to address her paranoia and stress. She continues to do well on quetiapine and supportive psychotherapy.
Related Resources
- Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc; 2003.
- Mental health: a report of the Surgeon General. Chapter 5: other mental disorders in older adults; schizophrenia in late life. www.surgeongeneral.gov/library/mentalhealth/chapter5/sec5.html.
Drug Brand Names
- Aripiprazole • Abilify
- Clozapine • Clozaril
- Diphenhydramine • Sominex, Unisom, others
- Haloperidol • Haldol
- Quetiapine • Seroquel
- Ziprasidone • Geodon
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.
1. Cohen CI, Cohen GD, Blank K, et al. Schizophrenia and older adults. An overview: directions for research and policy. Am J Geriatr Psychiatry. 2000;8:19-28.
2. Vahia I, Bankole AO, Diwan S, et al. Schizophrenia in late life. Aging Health. 2007;3:393-396.
3. Cohen CI, Pathak R, Ramirez PM, et al. Outcome among community dwelling older adults with schizophrenia: results using five conceptual models. Community Ment Health J. 2009;45:151-156.
4. Ausland LA, Jeste DV. Sustained remission of schizophrenia among community-dwelling older outpatients. Am J Psychiatry. 2004;161:1490-1493.
5. Bankole A, Cohen CI, Vahia I, et al. Symptomatic remission in a multiracial urban population of older adults with schizophrenia. Am J Geriatr Psychiatry. 2008;16(12):966-973.
6. Harding CM. Changes in schizophrenia across time. In: Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc.; 2003: 19–41.
7. Harvey PD, Howanitz E, Parrella M, et al. Symptoms, cognitive functioning, and adaptive skills in geriatric patients with lifelong schizophrenia: a comparison across treatment sites. Am J Psychiatry. 1998;155:1080-1086.
8. Morrison G, O’Carroll,, McCreadie R. Long-term course of cognitive impairment in schizophrenia. Br J Psychiatry. 2006;189:556-557.
9. Bowie CR, Tsapelas I, Friedman J, et al. The longitudinal course of thought disorder in geriatric patients with chronic schizophrenia. Am J Psychiatry. 2005;162(4):793-795.
10. Diwan S, Cohen CI, Bankole AO, et al. Depression in older adults with schizophrenia spectrum disorders: prevalence and associated factors. Am J Geriatr Psychiatry. 2007;15:991-998.
11. Berry K, Barrowclough C. The needs of older adults with schizophrenia. Implications for psychological interventions. Clin Psych Review. 2009;29:68-76.
12. Cohen CI. Age-related correlations in patient symptom management strategies in schizophrenia: an exploratory study. Int J Geriatr Psychiatry. 1993;8:211-213.
13. Leucht S, Burkard T, Henderson J, et al. Physical illness and schizophrenia: a review of the literature. Acta Psychiatr Scand. 2007;116(5):317-333.
14. Cohen CI, Vahia I, Reyes P, et al. Schizophrenia in later life: clinical symptoms and social well-being. Psychiatr Serv. 2008;59:232-234.
15. Buchanan RW, Kreyenbuhl J, Kelly DL, et al. The 2009 Schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36:71-93.
16. Dixon LB, Dickerson F, Bellack AS, et al. The 2009 Schizophrenia PORT psychosocial treatment recommendations and summary statements. Schizophr Bull. 2010;36:48-70.
17. McQuaid JR, Granholm E, McClure FS, et al. Development of an integrated cognitive-behavioral and social skills training intervention for older patients with schizophrenia. J Psychother Pract Res. 2000;9:149-156.
18. Granholm E, McQuaid JR, McClure FS, et al. A randomized controlled pilot study of cognitive behavioral social skills training for older patients with schizophrenia. Schizophr Res. 2002;53:167-169.
19. Patterson TL, McKibbin C, Taylor M, et al. Functional adaptation skills training (FAST): a pilot psychosocial intervention study in middle-aged and older patients with chronic psychiatric disorders. Am J Geriatr Psychiatry. 2003;11:17-23.
20. Patterson TL, Goldman S, McKibbin CL, et al. UCSD performance-based skills assessment: development of a new measure of everyday functioning for severely mentally ill adults. Schizophr Bull. 2001;27:235-245.
1. Cohen CI, Cohen GD, Blank K, et al. Schizophrenia and older adults. An overview: directions for research and policy. Am J Geriatr Psychiatry. 2000;8:19-28.
2. Vahia I, Bankole AO, Diwan S, et al. Schizophrenia in late life. Aging Health. 2007;3:393-396.
3. Cohen CI, Pathak R, Ramirez PM, et al. Outcome among community dwelling older adults with schizophrenia: results using five conceptual models. Community Ment Health J. 2009;45:151-156.
4. Ausland LA, Jeste DV. Sustained remission of schizophrenia among community-dwelling older outpatients. Am J Psychiatry. 2004;161:1490-1493.
5. Bankole A, Cohen CI, Vahia I, et al. Symptomatic remission in a multiracial urban population of older adults with schizophrenia. Am J Geriatr Psychiatry. 2008;16(12):966-973.
6. Harding CM. Changes in schizophrenia across time. In: Cohen CI, ed. Schizophrenia into later life: treatment, research, and policy. Arlington, VA: American Psychiatric Publishing, Inc.; 2003: 19–41.
7. Harvey PD, Howanitz E, Parrella M, et al. Symptoms, cognitive functioning, and adaptive skills in geriatric patients with lifelong schizophrenia: a comparison across treatment sites. Am J Psychiatry. 1998;155:1080-1086.
8. Morrison G, O’Carroll,, McCreadie R. Long-term course of cognitive impairment in schizophrenia. Br J Psychiatry. 2006;189:556-557.
9. Bowie CR, Tsapelas I, Friedman J, et al. The longitudinal course of thought disorder in geriatric patients with chronic schizophrenia. Am J Psychiatry. 2005;162(4):793-795.
10. Diwan S, Cohen CI, Bankole AO, et al. Depression in older adults with schizophrenia spectrum disorders: prevalence and associated factors. Am J Geriatr Psychiatry. 2007;15:991-998.
11. Berry K, Barrowclough C. The needs of older adults with schizophrenia. Implications for psychological interventions. Clin Psych Review. 2009;29:68-76.
12. Cohen CI. Age-related correlations in patient symptom management strategies in schizophrenia: an exploratory study. Int J Geriatr Psychiatry. 1993;8:211-213.
13. Leucht S, Burkard T, Henderson J, et al. Physical illness and schizophrenia: a review of the literature. Acta Psychiatr Scand. 2007;116(5):317-333.
14. Cohen CI, Vahia I, Reyes P, et al. Schizophrenia in later life: clinical symptoms and social well-being. Psychiatr Serv. 2008;59:232-234.
15. Buchanan RW, Kreyenbuhl J, Kelly DL, et al. The 2009 Schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36:71-93.
16. Dixon LB, Dickerson F, Bellack AS, et al. The 2009 Schizophrenia PORT psychosocial treatment recommendations and summary statements. Schizophr Bull. 2010;36:48-70.
17. McQuaid JR, Granholm E, McClure FS, et al. Development of an integrated cognitive-behavioral and social skills training intervention for older patients with schizophrenia. J Psychother Pract Res. 2000;9:149-156.
18. Granholm E, McQuaid JR, McClure FS, et al. A randomized controlled pilot study of cognitive behavioral social skills training for older patients with schizophrenia. Schizophr Res. 2002;53:167-169.
19. Patterson TL, McKibbin C, Taylor M, et al. Functional adaptation skills training (FAST): a pilot psychosocial intervention study in middle-aged and older patients with chronic psychiatric disorders. Am J Geriatr Psychiatry. 2003;11:17-23.
20. Patterson TL, Goldman S, McKibbin CL, et al. UCSD performance-based skills assessment: development of a new measure of everyday functioning for severely mentally ill adults. Schizophr Bull. 2001;27:235-245.
Vaccine for cocaine addiction: A promising new immunotherapy
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/vaccine-for-cocaine-addiction.html#comments
Unlike opioid or alcohol abuse, for cocaine dependence there are no FDA-approved pharmacotherapies, which leaves psychosocial treatment as the standard of care for the estimated 1.6 million individuals in the United States who abuse cocaine.1 However, researchers are developing a novel way to help cocaine-dependent patients reduce their drug use. Therapy for addiction–cocaine addiction (TA-CD) is thought to curb cocaine use by engaging the body’s immune reaction and stopping cocaine molecules from reaching the brain, thereby reducing the drug’s pleasurable effects. One researcher working on this vaccine, Eugene Somoza, MD, PhD—the principal investigator of the Ohio Valley Node of the National Institute on Drug Abuse clinical trials network of 16 universities and treatment programs—discusses with Current Psychiatry Section Editor Robert M. Anthenelli, MD, how TA-CD works and how it might be used in clinical practice.
DR. ANTHENELLI: How is immunotherapy being applied to treating cocaine addiction and how does TA-CD work?
DR. SOMOZA: Our bodies have a very efficient immune system that can recognize foreign proteins and other complex molecules and develop specific antibodies against them that join irreversibly to these molecules to make them inactive. Immunotherapy usually is used to treat disorders that involve very complex molecules. Cocaine is a very simple molecule, but you can attach a simple molecule to a complex molecule and still trigger the immune system. You can use this method to develop antibodies to cocaine. When an individual uses cocaine, the antibodies will bind to the cocaine in the blood stream and the drug never reaches the brain because the molecule is now too large to pass the blood-brain barrier. The reinforcing properties of addictive agents depend on how fast they get into the brain. By slowing down or even stopping this process, you decrease the pleasurable effect individuals get from cocaine.
The cocaine vaccine that is being tested makes use of the B subunit of the cholera toxin molecule. It is highly immunogenic, and a recombinant of it is available in large quantities.2 Cocaine molecules are connected to various areas of this complex cholera toxin subunit with covalent bonding. This makes the cocaine a larger target for an antibody response.
The interesting aspect of this process is that the vaccine acts outside of the brain. Other pharmacotherapies being tested, such as modafinil and disulfiram, target receptors or enzymes within the brain, which means that these 2 types of treatment would be synergistic. An early article on cocaine vaccines by Fox et al3 emphasized that this therapy is compatible with other treatments.
DR. ANTHENELLI: After receiving the vaccine, how long does it take for antibody levels to be high enough to produce a therapeutic effect?
DR. SOMOZA: Typically about 8 weeks.
DR. ANTHENELLI: Some trials have shown that patients display high variability in antibody levels. Only 38% of subjects in a 24-week, randomized, double-blind, placebo-controlled trial by Martell et al4 achieved high antibody levels (≥43 μ/mL). Are there ways to predict who will achieve the higher antibody titers and to increase the percentage of people who might develop the antibodies?
DR. SOMOZA: Right now there are not. In the Martell study, subjects’ antibody response curves—the increase and subsequent decrease in antibody concentration—were very different from individual to individual. We estimate that 40% of patients receiving the vaccine will develop ≥40 μ/mL of antibodies; this level is necessary for heavy cocaine users. However, not all patients take large amounts of cocaine, so we expect that even if a patient develops 30 μ/mL of antibodies, the amount of cocaine reaching the brain will be reduced— although the process may be slower—and using will not be as enticing.5
DR. ANTHENELLI: How long will the effects of TA-CD last, and how often will patients need to receive booster shots to keep antibody titers high?
DR. SOMOZA: The antibodies stay high for approximately 10 to 30 weeks, so you have to give boosters periodically. We need to carefully study if one can give a patient a booster every few months and, if so, how many booster shots would be required.
DR. ANTHENELLI: What are the known side effects of the vaccine?
DR. SOMOZA: In phase 1 and phase 2 studies, there haven’t been any problems at all.6 Theoretically, we could see some reaction at the injection site such as bruising or red or inflamed skin. In some cases with protein vaccines they’ve seen systemic reactions like fever. There’s also a risk of serum sickness, but this is theoretical based on other protein-based vaccines.7
DR. ANTHENELLI: Are there any data that address the safety of long-term TA-CD use?
DR. SOMOZA: We do not have any data on long-term use, but we know what happens over several months. When this project began 12 years ago, investigators worried that if the vaccine prevents cocaine from getting to the brain, cocaine-dependent individuals would just take more and more of the drug and suffer serious consequences. However, in preliminary studies, people have taken as much as 10 times their “normal” amount of cocaine with no adverse events. It looks like the vaccine may ameliorate some of cocaine’s effects on the heart. We’re certainly not encouraging study subjects to try to override the vaccine blockade, but these preliminary data at least minimize some of those concerns.
DR. ANTHENELLI: In clinical trials of TA-CD, during the 8-week ramp-up period where you’re waiting for patients’ antibody titers to get high enough to have a therapeutic effect, do trial participants receive other treatment?
DR. SOMOZA: Participants receive state-of-the-art cognitive-behavioral therapy (CBT) once a week. We do this to help patients look for triggers to cocaine use and how to handle them, but also to encourage them to stay in the study. It’s important that people who enroll in our trials are motivated to stop using. Many patients who have been using cocaine for years haven’t been able to own a house, get married, or even buy a car because all of their money is spent on cocaine. Eventually they decide it’s not a good idea to keep using forever. These are the participants we’d like to find.
There are other ways of increasing retention, such as rewarding patients for coming to appointments, providing urine for toxicology screens, or getting the boosters. We’re hoping contingency management will help keep patients in the trial.
How TA-CD will aid treatment
DR. ANTHENELLI: How do you envision TA-CD could be used in clinical practice?
DR. SOMOZA: It could become another tool in our armamentarium for treating cocaine dependence. Currently, there are no FDA-approved medications for cocaine dependence, although some pharmacologic treatments are being studied (Table).8-13 When a patient comes in to be vaccinated, he or she also could receive other treatments if they are available, and the effect potentially would be additive. We would also use CBT because cocaine dependence is a very complex disorder. In CBT patients identify triggers that cause them to want to use and learn how to combat them and make better decisions.
DR. ANTHENELLI: Some research shows that TA-CD doesn’t stop cocaine use altogether but reduces use. Will that be a deterrent for clinicians who wish to help patients achieve absteinence?
DR. SOMOZA: That’s true about any medication we develop for addictions. I think it is magical thinking to say that you can give patients a pill and they will be abstinent for the rest of their lives. If you look at tobacco or alcohol, in practice abstinence is an end point that one has to approximate successively. In addition, permanent abstinence from cocaine is virtually impossible to measure. Because the half-life of benzoylecgonine (BE), the principle metabolite of cocaine, is 6 to 8 hours, this limits the effectiveness of urine toxicology screens in monitoring abstinence. Cocaine-dependent patients might not have used the drug the day before a urine toxicology screen. If a patient says he is abstaining from cocaine, it would be difficult to document it quantitatively without obtaining urine BE levels every day or every other day.
I think clinicians need to get used to the fact that we have to treat cocaine dependence in an incremental manner. A pharmacotherapy that would reduce use and hopefully limit the problems people are having as a result of cocaine use would be a positive step.
DR. ANTHENELLI: If TA-CD is found to be effective, what is the earliest it might come into clinical use?
DR. SOMOZA: I would speculate that it would be 7 to 10 years.
DR. ANTHENELLI: What other kinds of research are going on as far as vaccines for cocaine?
DR. SOMOZA: There is a strain of transgenic mice that when stimulated produce human, as well as mice, antibodies. At the University of Cincinnati, Andrew Norman, PhD, was able to immunize these mice and they generated human antibodies against cocaine (Box 1).14,15 Then you’ll have vials of monoclonal antibodies that you can administer to your patient. However, this is still in early testing.
DR. ANTHENELLI: We’ve talked about immunotherapy and how it might work for the treatment of cocaine addiction. How might these types of vaccines be used for treating other substances of abuse?
DR. SOMOZA: Investigators are currently working on a vaccine for nicotine dependence (Box 2)4,16-20 and there’s a vaccine being developed for methamphetamine,21 but it is not as advanced as cocaine. A similar methodology has been used for some time to treat digitalis overdose. There is no antidote for digitalis toxicity, so researchers have developed an antibody—digoxin immune fab—that attaches to the drug, which is then excreted through the kidneys. I fully expect that this methodology eventually will work for cocaine, meth-amphetamine, and nicotine dependence. My hunch is that producing human antibody in industrial quantities would be the most sensible way to eventually make this work.
Table
Pharmacotherapy for cocaine dependence: Most evidence is weak
Study | Design | Results |
---|---|---|
Disulfiram | ||
Pani et al, 20108 | Meta-analysis of 7 studies with 492 cocaine-dependent patients | Researchers found ‘low evidence’ supporting disulfiram for treating cocaine dependence |
Modafinil | ||
Dackis et al, 20059 | 62 cocaine-dependent patients randomized to modafinil, 400 mg/d, or placebo for 8 weeks | Patients receiving modafinil provided significantly more BE-negative urine samples and were significantly more likely to achieve ≥3 weeks of cocaine abstinence |
Anderson et al, 200910 | 210 cocaine-dependent patients randomized to modafinil, 200 mg/d or 400 mg/d, or placebo for 12 weeks | Modafinil significantly reduced cocaine craving but did not significantly improve the average weekly percentage of cocaine non-use days |
Tiagabine | ||
Winhusen et al, 200711 | 141 cocaine-dependent patients randomized to tiagabine, 20 mg/d, or placebo for 12 weeks | No significant changes in cocaine use vs placebo as measured by self-report and urine BE |
Baclofen | ||
Kahn et al, 200912 | Cocaine-dependent patients randomized to baclofen, 60 mg/d, or placebo for 8 weeks | No significant difference between groups in cocaine use as measured by urine BE |
Ondansetron | ||
Johnson et al, 200613 | 63 cocaine-dependent patients randomized to ondansetron, 0.25 mg, 1 mg, or 4 mg twice daily, or placebo for 10 weeks | The odansetron 4 mg group had a significantly greater rate of improvement in percentage of patients with a cocaine-free week compared with the placebo group |
BE: benzoylecgonine |
Promising clinical trials of therapy for addiction–cocaine addiction (TA-CD) and nicotine conjugate vaccines show that immunotherapy may be effective for addictive disorders. However, immune response varies among patients and the vaccines are effective only in those who produce high concentrations of anti-drug antibodies. Our multidisciplinary translational research project has generated a predominantly human sequence monoclonal antibody (mAb) with high affinity (Kd = 4 nM) for cocaine and specificity over cocaine’s inactive metabolites. This mAb (preclinical designation, 2E2) is at an advanced stage of preclinical development for preventing relapse in treatment-seeking cocaine abusers.
Development of 2E2 has met several key safety and efficacy milestones. Because the structure of mAb is mostly human, repeated treatments should be safe and should confer long-term efficacy. 2E2 binds to and sequesters cocaine in the peripheral circulation and dramatically lowers brain cocaine concentrations in mice.14 Furthermore, 2E2 decreases the effect of cocaine in a rat model of relapse.15 In FDA-required safety tests, there was no apparent cross-reactivity of 2E2 with an extensive panel of human tissues in vitro, indicating that 2E2 likely is safe for patients. The genes encoding the mAb have been cloned and slightly re-engineered to make them even closer to a human sequence and the expressed recombinant protein retains the identical affinity and specificity for cocaine. We continue to work with our industry collaborator, Vybion Inc., to develop a stably transfected mammalian cell line capable of high-level production of 2E2, which is necessary to support in vivo toxicology studies required for an FDA Investigational New Drug application and subsequent clinical trials. This anti-cocaine mAb should be a useful adjunct to TA-CD by supplementing concentrations of vaccine-generated anti-cocaine antibodies.
Andrew B. Norman, PhD
Department of psychiatry and behavioral neuroscience
William J. Ball, PhD
Department of pharmacology and cell biophysics
University of Cincinnati College of Medicine
Cincinnati, OH
Dependence on nicotine—the main addictive agent in cigarette smoke and other tobacco products—also is being targeted with vaccines. Like cocaine, the nicotine molecule is too small to provoke an immune response by itself. Therefore, nicotine derivatives linked to virus-like particles16 or detoxified bacteria-derived proteins17 are immunogenic enough to stimulate an antibody response. With once-monthly vaccinations of these conjugated nicotine compounds, patients can produce sufficient antibodies to sequester nicotine in the peripheral bloodstream before it crosses the blood-brain barrier to produce its rewarding effects.
Animal and human studies have found proof that this concept may work. These immunotherapies do not seem to provoke acute nicotine withdrawal and patients do not increase their smoking rates to try to counteract the antibodies’ nicotine-scavenging effects.16-19 As was the case with the cocaine trial,4 smoking cessation efficacy is positively correlated with the individual’s antibody titer response. Published phase I and II trials indicate that these vaccines may be safe and well-tolerated; mild reactions at the intramuscular injection site are the most commonly reported adverse event.16-18 Larger phase III clinical trials are underway.20
Robert M. Anthenelli, MD
Related Resource
- Martell BA, Mitchell E, Poling J, et al. Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry. 2005;58(2):158-164.
Drug Brand Names
- Baclofen • Lioresal
- Digoxin • Lanoxin
- Digoxin immune fab • Digibind
- Disulfiram • Antabuse
- Modafinil • Provigil
- Ondansetron • Zofran
- Tiagabine • Gabitril
Disclosure
Dr. Anthenelli receives grant/research support from Eli Lilly and Company, Nabi Biopharmaceuticals, and Pfizer Inc., and is a consultant to Pfizer Inc.
Dr. Somoza receives grant/research support from the National Institute on Drug Abuse.
Drs. Norman and Ball receive grant/research support from the National Institutes of Health and the National Institute on Drug Abuse and are consultants to Vybion, Inc.
Treatment adherence
DR. ANTHENELLI: We know from working in the addiction field that compliance with medication regimens is a big challenge. What are the data regarding adherence to TA-CD?
DR. SOMOZA: We don’t have any specific data about adherence to the vaccine, but it is probably similar to any other medication for addiction. Remember that cocaine-dependent patients often are erratic and don’t use planners to set up their day. If you look at clinical trials over the past 20 years, if you get 75% retention you’re doing really good, but quite often you see 50% or 25% retention. With TA-CD, retention is going to be worse because you have to wait 8 weeks before patients build up enough antibodies to have therapeutic effect. I’m hoping we can convince the FDA to look at the relationship between antibody generation and improvement in treatment efficacy. Obviously if patients don’t develop antibodies they’re not going to get better.
Patient characteristics
DR. ANTHENELLI: I know it’s a little early, but if you had to use your crystal ball, what type of patient do you think that TA-CD might work best for?
DR. SOMOZA: Certainly it would be for people that are motivated to stop using. If they really don’t want to stop using cocaine, probably nothing will work. These patients could get the vaccine and boosters and it won’t do them any good. They’ll take it and nothing happens.
Future research
DR. ANTHENELLI: One of the things you have discussed is who will achieve enough antibody titers to make TA-CD effective. Are there other kinds of research you think will be related to this?
DR. SOMOZA: Increasing the serum concentration of the antibodies is one. Another would be to increase the fraction of people who develop high levels of antibodies. One wonders if we could use a different protein that would increase the immunogenicity of the vaccine. If we use 2 different proteins, perhaps the effects would be additive. In an early study of mice, Fox and colleaguesa used a blood protein, not a cholera toxin.
References
a. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
1. Substance Abuse and Mental Health Services Administration. 2007 national survey on drug use and health. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2008.
2. Jertborn M, Svennerholm AM, Holmgren J. Safety and immunogenicity of an oral recombinant cholera B subunit-whole cell vaccine in Swedish volunteers. Vaccine. 1992;10:130-132.
3. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
4. Martell BA, Orson FM, Poling J, et al. Cocaine vaccine for the treatment of cocaine dependence in methadone-maintained patients: a randomized, double-blind, placebo-controlled efficacy trial. Arch Gen Psychiatry. 2009;66(10):1116-1123.
5. Haney M, Gunderson EW, Jiang H, et al. Cocaine-specific antibodies blunt the subjective effects of smoked cocaine in humans. Biol Psychiatry. 2010;67(1):59-65.
6. Kosten TR, Rosen M, Bond J, et al. Human therapeutic cocaine vaccine: safety and immunogenicity. Vaccine. 2002;20:1196-1204.
7. Grabenstein JD. ImmunoFacts: vaccines and immunological drugs. St. Louis, MO: Facts and Comparisons, Inc.; 1994:487b.
8. Pani PP, Trogu E, Vacca R, et al. Disulfiram for the treatment of cocaine dependence. Cochrane Database Syst Rev. 2010;(1):CD007024.-
9. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology. 2005;30(1):205-211.
10. Anderson AL, Reid MS, Li SH, et al. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend. 2009;104(1-2):133-139.
11. Winhusen T, Somoza E, Ciraulo DA, et al. A double-blind, placebo-controlled trial of tiagabine for the treatment of cocaine dependence. Drug Alcohol Depend. 2007;91 (2-3):141-148.
12. Kahn R, Biswas K, Childress AR, et al. Multi-center trial of baclofen for abstinence initiation in severe cocaine-dependent individuals. Drug Alcohol Depend. 2009;103 (1-2):59-64.
13. Johnson BA, Roache JD, Ait-Daoud N, et al. A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of cocaine dependence. Drug Alcohol Depend. 2006;84(3):256-263.
14. Norman AB, Tabet MR, Norman MK, et al. A chimeric human/murine anticocaine monoclonal antibody inhibits the distribution of cocaine to the brain in mice. J Pharmacol Exp Ther. 2007;320:145-153.
15. Norman AB, Norman MK, Buesing WR, et al. The effect of a chimeric human/murine anti-cocaine monoclonal antibody on cocaine self-administration in rats. J Pharmacol Exp Ther. 2009;328:873-881.
16. Cornuz J, Zwahlen S, Jungi WF, et al. A vaccine against nicotine for smoking cessation: a randomized controlled trial. PLoS One. 2008;3(6):e2547.-
17. Wagena EJ, de Vos A, Horwith G, et al. The immunogenicity and safety of a nicotine vaccine in smokers and nonsmokers: results of a randomized, placebo-controlled phase 1/2 trial. Nicotine Tob Res. 2008;10(1):213-218.
18. Hatsukami DK, Rennard S, Jorenby D, et al. Safety and immunogenicity of a nicotine conjugate vaccine in current smokers. Clin Pharmacol Ther. 2005;78(5):456-467.
19. Maurer P, Bachmann MF. Vaccination against nicotine: an emerging therapy for tobacco dependence. Expert Opin Investig Drugs. 2007;16(11):1775-1783.
20. Volkow ND. Message from the director on ARRA funding for the development of a nicotine vaccine. National Institute on Drug Abuse Web site. Available at: http://drugabuse.gov/about/welcome/nicotinevaccine909.html. Accessed August 13, 2010.
21. Laurenzana EM, Hendrickson HP, Carpenter D, et al. Functional and biological determinants affecting the duration of action and efficacy of anti-(+)-methamphetamine monoclonal antibodies in rats. Vaccine. 2009;27(50):7011-7020.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/vaccine-for-cocaine-addiction.html#comments
Unlike opioid or alcohol abuse, for cocaine dependence there are no FDA-approved pharmacotherapies, which leaves psychosocial treatment as the standard of care for the estimated 1.6 million individuals in the United States who abuse cocaine.1 However, researchers are developing a novel way to help cocaine-dependent patients reduce their drug use. Therapy for addiction–cocaine addiction (TA-CD) is thought to curb cocaine use by engaging the body’s immune reaction and stopping cocaine molecules from reaching the brain, thereby reducing the drug’s pleasurable effects. One researcher working on this vaccine, Eugene Somoza, MD, PhD—the principal investigator of the Ohio Valley Node of the National Institute on Drug Abuse clinical trials network of 16 universities and treatment programs—discusses with Current Psychiatry Section Editor Robert M. Anthenelli, MD, how TA-CD works and how it might be used in clinical practice.
DR. ANTHENELLI: How is immunotherapy being applied to treating cocaine addiction and how does TA-CD work?
DR. SOMOZA: Our bodies have a very efficient immune system that can recognize foreign proteins and other complex molecules and develop specific antibodies against them that join irreversibly to these molecules to make them inactive. Immunotherapy usually is used to treat disorders that involve very complex molecules. Cocaine is a very simple molecule, but you can attach a simple molecule to a complex molecule and still trigger the immune system. You can use this method to develop antibodies to cocaine. When an individual uses cocaine, the antibodies will bind to the cocaine in the blood stream and the drug never reaches the brain because the molecule is now too large to pass the blood-brain barrier. The reinforcing properties of addictive agents depend on how fast they get into the brain. By slowing down or even stopping this process, you decrease the pleasurable effect individuals get from cocaine.
The cocaine vaccine that is being tested makes use of the B subunit of the cholera toxin molecule. It is highly immunogenic, and a recombinant of it is available in large quantities.2 Cocaine molecules are connected to various areas of this complex cholera toxin subunit with covalent bonding. This makes the cocaine a larger target for an antibody response.
The interesting aspect of this process is that the vaccine acts outside of the brain. Other pharmacotherapies being tested, such as modafinil and disulfiram, target receptors or enzymes within the brain, which means that these 2 types of treatment would be synergistic. An early article on cocaine vaccines by Fox et al3 emphasized that this therapy is compatible with other treatments.
DR. ANTHENELLI: After receiving the vaccine, how long does it take for antibody levels to be high enough to produce a therapeutic effect?
DR. SOMOZA: Typically about 8 weeks.
DR. ANTHENELLI: Some trials have shown that patients display high variability in antibody levels. Only 38% of subjects in a 24-week, randomized, double-blind, placebo-controlled trial by Martell et al4 achieved high antibody levels (≥43 μ/mL). Are there ways to predict who will achieve the higher antibody titers and to increase the percentage of people who might develop the antibodies?
DR. SOMOZA: Right now there are not. In the Martell study, subjects’ antibody response curves—the increase and subsequent decrease in antibody concentration—were very different from individual to individual. We estimate that 40% of patients receiving the vaccine will develop ≥40 μ/mL of antibodies; this level is necessary for heavy cocaine users. However, not all patients take large amounts of cocaine, so we expect that even if a patient develops 30 μ/mL of antibodies, the amount of cocaine reaching the brain will be reduced— although the process may be slower—and using will not be as enticing.5
DR. ANTHENELLI: How long will the effects of TA-CD last, and how often will patients need to receive booster shots to keep antibody titers high?
DR. SOMOZA: The antibodies stay high for approximately 10 to 30 weeks, so you have to give boosters periodically. We need to carefully study if one can give a patient a booster every few months and, if so, how many booster shots would be required.
DR. ANTHENELLI: What are the known side effects of the vaccine?
DR. SOMOZA: In phase 1 and phase 2 studies, there haven’t been any problems at all.6 Theoretically, we could see some reaction at the injection site such as bruising or red or inflamed skin. In some cases with protein vaccines they’ve seen systemic reactions like fever. There’s also a risk of serum sickness, but this is theoretical based on other protein-based vaccines.7
DR. ANTHENELLI: Are there any data that address the safety of long-term TA-CD use?
DR. SOMOZA: We do not have any data on long-term use, but we know what happens over several months. When this project began 12 years ago, investigators worried that if the vaccine prevents cocaine from getting to the brain, cocaine-dependent individuals would just take more and more of the drug and suffer serious consequences. However, in preliminary studies, people have taken as much as 10 times their “normal” amount of cocaine with no adverse events. It looks like the vaccine may ameliorate some of cocaine’s effects on the heart. We’re certainly not encouraging study subjects to try to override the vaccine blockade, but these preliminary data at least minimize some of those concerns.
DR. ANTHENELLI: In clinical trials of TA-CD, during the 8-week ramp-up period where you’re waiting for patients’ antibody titers to get high enough to have a therapeutic effect, do trial participants receive other treatment?
DR. SOMOZA: Participants receive state-of-the-art cognitive-behavioral therapy (CBT) once a week. We do this to help patients look for triggers to cocaine use and how to handle them, but also to encourage them to stay in the study. It’s important that people who enroll in our trials are motivated to stop using. Many patients who have been using cocaine for years haven’t been able to own a house, get married, or even buy a car because all of their money is spent on cocaine. Eventually they decide it’s not a good idea to keep using forever. These are the participants we’d like to find.
There are other ways of increasing retention, such as rewarding patients for coming to appointments, providing urine for toxicology screens, or getting the boosters. We’re hoping contingency management will help keep patients in the trial.
How TA-CD will aid treatment
DR. ANTHENELLI: How do you envision TA-CD could be used in clinical practice?
DR. SOMOZA: It could become another tool in our armamentarium for treating cocaine dependence. Currently, there are no FDA-approved medications for cocaine dependence, although some pharmacologic treatments are being studied (Table).8-13 When a patient comes in to be vaccinated, he or she also could receive other treatments if they are available, and the effect potentially would be additive. We would also use CBT because cocaine dependence is a very complex disorder. In CBT patients identify triggers that cause them to want to use and learn how to combat them and make better decisions.
DR. ANTHENELLI: Some research shows that TA-CD doesn’t stop cocaine use altogether but reduces use. Will that be a deterrent for clinicians who wish to help patients achieve absteinence?
DR. SOMOZA: That’s true about any medication we develop for addictions. I think it is magical thinking to say that you can give patients a pill and they will be abstinent for the rest of their lives. If you look at tobacco or alcohol, in practice abstinence is an end point that one has to approximate successively. In addition, permanent abstinence from cocaine is virtually impossible to measure. Because the half-life of benzoylecgonine (BE), the principle metabolite of cocaine, is 6 to 8 hours, this limits the effectiveness of urine toxicology screens in monitoring abstinence. Cocaine-dependent patients might not have used the drug the day before a urine toxicology screen. If a patient says he is abstaining from cocaine, it would be difficult to document it quantitatively without obtaining urine BE levels every day or every other day.
I think clinicians need to get used to the fact that we have to treat cocaine dependence in an incremental manner. A pharmacotherapy that would reduce use and hopefully limit the problems people are having as a result of cocaine use would be a positive step.
DR. ANTHENELLI: If TA-CD is found to be effective, what is the earliest it might come into clinical use?
DR. SOMOZA: I would speculate that it would be 7 to 10 years.
DR. ANTHENELLI: What other kinds of research are going on as far as vaccines for cocaine?
DR. SOMOZA: There is a strain of transgenic mice that when stimulated produce human, as well as mice, antibodies. At the University of Cincinnati, Andrew Norman, PhD, was able to immunize these mice and they generated human antibodies against cocaine (Box 1).14,15 Then you’ll have vials of monoclonal antibodies that you can administer to your patient. However, this is still in early testing.
DR. ANTHENELLI: We’ve talked about immunotherapy and how it might work for the treatment of cocaine addiction. How might these types of vaccines be used for treating other substances of abuse?
DR. SOMOZA: Investigators are currently working on a vaccine for nicotine dependence (Box 2)4,16-20 and there’s a vaccine being developed for methamphetamine,21 but it is not as advanced as cocaine. A similar methodology has been used for some time to treat digitalis overdose. There is no antidote for digitalis toxicity, so researchers have developed an antibody—digoxin immune fab—that attaches to the drug, which is then excreted through the kidneys. I fully expect that this methodology eventually will work for cocaine, meth-amphetamine, and nicotine dependence. My hunch is that producing human antibody in industrial quantities would be the most sensible way to eventually make this work.
Table
Pharmacotherapy for cocaine dependence: Most evidence is weak
Study | Design | Results |
---|---|---|
Disulfiram | ||
Pani et al, 20108 | Meta-analysis of 7 studies with 492 cocaine-dependent patients | Researchers found ‘low evidence’ supporting disulfiram for treating cocaine dependence |
Modafinil | ||
Dackis et al, 20059 | 62 cocaine-dependent patients randomized to modafinil, 400 mg/d, or placebo for 8 weeks | Patients receiving modafinil provided significantly more BE-negative urine samples and were significantly more likely to achieve ≥3 weeks of cocaine abstinence |
Anderson et al, 200910 | 210 cocaine-dependent patients randomized to modafinil, 200 mg/d or 400 mg/d, or placebo for 12 weeks | Modafinil significantly reduced cocaine craving but did not significantly improve the average weekly percentage of cocaine non-use days |
Tiagabine | ||
Winhusen et al, 200711 | 141 cocaine-dependent patients randomized to tiagabine, 20 mg/d, or placebo for 12 weeks | No significant changes in cocaine use vs placebo as measured by self-report and urine BE |
Baclofen | ||
Kahn et al, 200912 | Cocaine-dependent patients randomized to baclofen, 60 mg/d, or placebo for 8 weeks | No significant difference between groups in cocaine use as measured by urine BE |
Ondansetron | ||
Johnson et al, 200613 | 63 cocaine-dependent patients randomized to ondansetron, 0.25 mg, 1 mg, or 4 mg twice daily, or placebo for 10 weeks | The odansetron 4 mg group had a significantly greater rate of improvement in percentage of patients with a cocaine-free week compared with the placebo group |
BE: benzoylecgonine |
Promising clinical trials of therapy for addiction–cocaine addiction (TA-CD) and nicotine conjugate vaccines show that immunotherapy may be effective for addictive disorders. However, immune response varies among patients and the vaccines are effective only in those who produce high concentrations of anti-drug antibodies. Our multidisciplinary translational research project has generated a predominantly human sequence monoclonal antibody (mAb) with high affinity (Kd = 4 nM) for cocaine and specificity over cocaine’s inactive metabolites. This mAb (preclinical designation, 2E2) is at an advanced stage of preclinical development for preventing relapse in treatment-seeking cocaine abusers.
Development of 2E2 has met several key safety and efficacy milestones. Because the structure of mAb is mostly human, repeated treatments should be safe and should confer long-term efficacy. 2E2 binds to and sequesters cocaine in the peripheral circulation and dramatically lowers brain cocaine concentrations in mice.14 Furthermore, 2E2 decreases the effect of cocaine in a rat model of relapse.15 In FDA-required safety tests, there was no apparent cross-reactivity of 2E2 with an extensive panel of human tissues in vitro, indicating that 2E2 likely is safe for patients. The genes encoding the mAb have been cloned and slightly re-engineered to make them even closer to a human sequence and the expressed recombinant protein retains the identical affinity and specificity for cocaine. We continue to work with our industry collaborator, Vybion Inc., to develop a stably transfected mammalian cell line capable of high-level production of 2E2, which is necessary to support in vivo toxicology studies required for an FDA Investigational New Drug application and subsequent clinical trials. This anti-cocaine mAb should be a useful adjunct to TA-CD by supplementing concentrations of vaccine-generated anti-cocaine antibodies.
Andrew B. Norman, PhD
Department of psychiatry and behavioral neuroscience
William J. Ball, PhD
Department of pharmacology and cell biophysics
University of Cincinnati College of Medicine
Cincinnati, OH
Dependence on nicotine—the main addictive agent in cigarette smoke and other tobacco products—also is being targeted with vaccines. Like cocaine, the nicotine molecule is too small to provoke an immune response by itself. Therefore, nicotine derivatives linked to virus-like particles16 or detoxified bacteria-derived proteins17 are immunogenic enough to stimulate an antibody response. With once-monthly vaccinations of these conjugated nicotine compounds, patients can produce sufficient antibodies to sequester nicotine in the peripheral bloodstream before it crosses the blood-brain barrier to produce its rewarding effects.
Animal and human studies have found proof that this concept may work. These immunotherapies do not seem to provoke acute nicotine withdrawal and patients do not increase their smoking rates to try to counteract the antibodies’ nicotine-scavenging effects.16-19 As was the case with the cocaine trial,4 smoking cessation efficacy is positively correlated with the individual’s antibody titer response. Published phase I and II trials indicate that these vaccines may be safe and well-tolerated; mild reactions at the intramuscular injection site are the most commonly reported adverse event.16-18 Larger phase III clinical trials are underway.20
Robert M. Anthenelli, MD
Related Resource
- Martell BA, Mitchell E, Poling J, et al. Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry. 2005;58(2):158-164.
Drug Brand Names
- Baclofen • Lioresal
- Digoxin • Lanoxin
- Digoxin immune fab • Digibind
- Disulfiram • Antabuse
- Modafinil • Provigil
- Ondansetron • Zofran
- Tiagabine • Gabitril
Disclosure
Dr. Anthenelli receives grant/research support from Eli Lilly and Company, Nabi Biopharmaceuticals, and Pfizer Inc., and is a consultant to Pfizer Inc.
Dr. Somoza receives grant/research support from the National Institute on Drug Abuse.
Drs. Norman and Ball receive grant/research support from the National Institutes of Health and the National Institute on Drug Abuse and are consultants to Vybion, Inc.
Treatment adherence
DR. ANTHENELLI: We know from working in the addiction field that compliance with medication regimens is a big challenge. What are the data regarding adherence to TA-CD?
DR. SOMOZA: We don’t have any specific data about adherence to the vaccine, but it is probably similar to any other medication for addiction. Remember that cocaine-dependent patients often are erratic and don’t use planners to set up their day. If you look at clinical trials over the past 20 years, if you get 75% retention you’re doing really good, but quite often you see 50% or 25% retention. With TA-CD, retention is going to be worse because you have to wait 8 weeks before patients build up enough antibodies to have therapeutic effect. I’m hoping we can convince the FDA to look at the relationship between antibody generation and improvement in treatment efficacy. Obviously if patients don’t develop antibodies they’re not going to get better.
Patient characteristics
DR. ANTHENELLI: I know it’s a little early, but if you had to use your crystal ball, what type of patient do you think that TA-CD might work best for?
DR. SOMOZA: Certainly it would be for people that are motivated to stop using. If they really don’t want to stop using cocaine, probably nothing will work. These patients could get the vaccine and boosters and it won’t do them any good. They’ll take it and nothing happens.
Future research
DR. ANTHENELLI: One of the things you have discussed is who will achieve enough antibody titers to make TA-CD effective. Are there other kinds of research you think will be related to this?
DR. SOMOZA: Increasing the serum concentration of the antibodies is one. Another would be to increase the fraction of people who develop high levels of antibodies. One wonders if we could use a different protein that would increase the immunogenicity of the vaccine. If we use 2 different proteins, perhaps the effects would be additive. In an early study of mice, Fox and colleaguesa used a blood protein, not a cholera toxin.
References
a. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/09/vaccine-for-cocaine-addiction.html#comments
Unlike opioid or alcohol abuse, for cocaine dependence there are no FDA-approved pharmacotherapies, which leaves psychosocial treatment as the standard of care for the estimated 1.6 million individuals in the United States who abuse cocaine.1 However, researchers are developing a novel way to help cocaine-dependent patients reduce their drug use. Therapy for addiction–cocaine addiction (TA-CD) is thought to curb cocaine use by engaging the body’s immune reaction and stopping cocaine molecules from reaching the brain, thereby reducing the drug’s pleasurable effects. One researcher working on this vaccine, Eugene Somoza, MD, PhD—the principal investigator of the Ohio Valley Node of the National Institute on Drug Abuse clinical trials network of 16 universities and treatment programs—discusses with Current Psychiatry Section Editor Robert M. Anthenelli, MD, how TA-CD works and how it might be used in clinical practice.
DR. ANTHENELLI: How is immunotherapy being applied to treating cocaine addiction and how does TA-CD work?
DR. SOMOZA: Our bodies have a very efficient immune system that can recognize foreign proteins and other complex molecules and develop specific antibodies against them that join irreversibly to these molecules to make them inactive. Immunotherapy usually is used to treat disorders that involve very complex molecules. Cocaine is a very simple molecule, but you can attach a simple molecule to a complex molecule and still trigger the immune system. You can use this method to develop antibodies to cocaine. When an individual uses cocaine, the antibodies will bind to the cocaine in the blood stream and the drug never reaches the brain because the molecule is now too large to pass the blood-brain barrier. The reinforcing properties of addictive agents depend on how fast they get into the brain. By slowing down or even stopping this process, you decrease the pleasurable effect individuals get from cocaine.
The cocaine vaccine that is being tested makes use of the B subunit of the cholera toxin molecule. It is highly immunogenic, and a recombinant of it is available in large quantities.2 Cocaine molecules are connected to various areas of this complex cholera toxin subunit with covalent bonding. This makes the cocaine a larger target for an antibody response.
The interesting aspect of this process is that the vaccine acts outside of the brain. Other pharmacotherapies being tested, such as modafinil and disulfiram, target receptors or enzymes within the brain, which means that these 2 types of treatment would be synergistic. An early article on cocaine vaccines by Fox et al3 emphasized that this therapy is compatible with other treatments.
DR. ANTHENELLI: After receiving the vaccine, how long does it take for antibody levels to be high enough to produce a therapeutic effect?
DR. SOMOZA: Typically about 8 weeks.
DR. ANTHENELLI: Some trials have shown that patients display high variability in antibody levels. Only 38% of subjects in a 24-week, randomized, double-blind, placebo-controlled trial by Martell et al4 achieved high antibody levels (≥43 μ/mL). Are there ways to predict who will achieve the higher antibody titers and to increase the percentage of people who might develop the antibodies?
DR. SOMOZA: Right now there are not. In the Martell study, subjects’ antibody response curves—the increase and subsequent decrease in antibody concentration—were very different from individual to individual. We estimate that 40% of patients receiving the vaccine will develop ≥40 μ/mL of antibodies; this level is necessary for heavy cocaine users. However, not all patients take large amounts of cocaine, so we expect that even if a patient develops 30 μ/mL of antibodies, the amount of cocaine reaching the brain will be reduced— although the process may be slower—and using will not be as enticing.5
DR. ANTHENELLI: How long will the effects of TA-CD last, and how often will patients need to receive booster shots to keep antibody titers high?
DR. SOMOZA: The antibodies stay high for approximately 10 to 30 weeks, so you have to give boosters periodically. We need to carefully study if one can give a patient a booster every few months and, if so, how many booster shots would be required.
DR. ANTHENELLI: What are the known side effects of the vaccine?
DR. SOMOZA: In phase 1 and phase 2 studies, there haven’t been any problems at all.6 Theoretically, we could see some reaction at the injection site such as bruising or red or inflamed skin. In some cases with protein vaccines they’ve seen systemic reactions like fever. There’s also a risk of serum sickness, but this is theoretical based on other protein-based vaccines.7
DR. ANTHENELLI: Are there any data that address the safety of long-term TA-CD use?
DR. SOMOZA: We do not have any data on long-term use, but we know what happens over several months. When this project began 12 years ago, investigators worried that if the vaccine prevents cocaine from getting to the brain, cocaine-dependent individuals would just take more and more of the drug and suffer serious consequences. However, in preliminary studies, people have taken as much as 10 times their “normal” amount of cocaine with no adverse events. It looks like the vaccine may ameliorate some of cocaine’s effects on the heart. We’re certainly not encouraging study subjects to try to override the vaccine blockade, but these preliminary data at least minimize some of those concerns.
DR. ANTHENELLI: In clinical trials of TA-CD, during the 8-week ramp-up period where you’re waiting for patients’ antibody titers to get high enough to have a therapeutic effect, do trial participants receive other treatment?
DR. SOMOZA: Participants receive state-of-the-art cognitive-behavioral therapy (CBT) once a week. We do this to help patients look for triggers to cocaine use and how to handle them, but also to encourage them to stay in the study. It’s important that people who enroll in our trials are motivated to stop using. Many patients who have been using cocaine for years haven’t been able to own a house, get married, or even buy a car because all of their money is spent on cocaine. Eventually they decide it’s not a good idea to keep using forever. These are the participants we’d like to find.
There are other ways of increasing retention, such as rewarding patients for coming to appointments, providing urine for toxicology screens, or getting the boosters. We’re hoping contingency management will help keep patients in the trial.
How TA-CD will aid treatment
DR. ANTHENELLI: How do you envision TA-CD could be used in clinical practice?
DR. SOMOZA: It could become another tool in our armamentarium for treating cocaine dependence. Currently, there are no FDA-approved medications for cocaine dependence, although some pharmacologic treatments are being studied (Table).8-13 When a patient comes in to be vaccinated, he or she also could receive other treatments if they are available, and the effect potentially would be additive. We would also use CBT because cocaine dependence is a very complex disorder. In CBT patients identify triggers that cause them to want to use and learn how to combat them and make better decisions.
DR. ANTHENELLI: Some research shows that TA-CD doesn’t stop cocaine use altogether but reduces use. Will that be a deterrent for clinicians who wish to help patients achieve absteinence?
DR. SOMOZA: That’s true about any medication we develop for addictions. I think it is magical thinking to say that you can give patients a pill and they will be abstinent for the rest of their lives. If you look at tobacco or alcohol, in practice abstinence is an end point that one has to approximate successively. In addition, permanent abstinence from cocaine is virtually impossible to measure. Because the half-life of benzoylecgonine (BE), the principle metabolite of cocaine, is 6 to 8 hours, this limits the effectiveness of urine toxicology screens in monitoring abstinence. Cocaine-dependent patients might not have used the drug the day before a urine toxicology screen. If a patient says he is abstaining from cocaine, it would be difficult to document it quantitatively without obtaining urine BE levels every day or every other day.
I think clinicians need to get used to the fact that we have to treat cocaine dependence in an incremental manner. A pharmacotherapy that would reduce use and hopefully limit the problems people are having as a result of cocaine use would be a positive step.
DR. ANTHENELLI: If TA-CD is found to be effective, what is the earliest it might come into clinical use?
DR. SOMOZA: I would speculate that it would be 7 to 10 years.
DR. ANTHENELLI: What other kinds of research are going on as far as vaccines for cocaine?
DR. SOMOZA: There is a strain of transgenic mice that when stimulated produce human, as well as mice, antibodies. At the University of Cincinnati, Andrew Norman, PhD, was able to immunize these mice and they generated human antibodies against cocaine (Box 1).14,15 Then you’ll have vials of monoclonal antibodies that you can administer to your patient. However, this is still in early testing.
DR. ANTHENELLI: We’ve talked about immunotherapy and how it might work for the treatment of cocaine addiction. How might these types of vaccines be used for treating other substances of abuse?
DR. SOMOZA: Investigators are currently working on a vaccine for nicotine dependence (Box 2)4,16-20 and there’s a vaccine being developed for methamphetamine,21 but it is not as advanced as cocaine. A similar methodology has been used for some time to treat digitalis overdose. There is no antidote for digitalis toxicity, so researchers have developed an antibody—digoxin immune fab—that attaches to the drug, which is then excreted through the kidneys. I fully expect that this methodology eventually will work for cocaine, meth-amphetamine, and nicotine dependence. My hunch is that producing human antibody in industrial quantities would be the most sensible way to eventually make this work.
Table
Pharmacotherapy for cocaine dependence: Most evidence is weak
Study | Design | Results |
---|---|---|
Disulfiram | ||
Pani et al, 20108 | Meta-analysis of 7 studies with 492 cocaine-dependent patients | Researchers found ‘low evidence’ supporting disulfiram for treating cocaine dependence |
Modafinil | ||
Dackis et al, 20059 | 62 cocaine-dependent patients randomized to modafinil, 400 mg/d, or placebo for 8 weeks | Patients receiving modafinil provided significantly more BE-negative urine samples and were significantly more likely to achieve ≥3 weeks of cocaine abstinence |
Anderson et al, 200910 | 210 cocaine-dependent patients randomized to modafinil, 200 mg/d or 400 mg/d, or placebo for 12 weeks | Modafinil significantly reduced cocaine craving but did not significantly improve the average weekly percentage of cocaine non-use days |
Tiagabine | ||
Winhusen et al, 200711 | 141 cocaine-dependent patients randomized to tiagabine, 20 mg/d, or placebo for 12 weeks | No significant changes in cocaine use vs placebo as measured by self-report and urine BE |
Baclofen | ||
Kahn et al, 200912 | Cocaine-dependent patients randomized to baclofen, 60 mg/d, or placebo for 8 weeks | No significant difference between groups in cocaine use as measured by urine BE |
Ondansetron | ||
Johnson et al, 200613 | 63 cocaine-dependent patients randomized to ondansetron, 0.25 mg, 1 mg, or 4 mg twice daily, or placebo for 10 weeks | The odansetron 4 mg group had a significantly greater rate of improvement in percentage of patients with a cocaine-free week compared with the placebo group |
BE: benzoylecgonine |
Promising clinical trials of therapy for addiction–cocaine addiction (TA-CD) and nicotine conjugate vaccines show that immunotherapy may be effective for addictive disorders. However, immune response varies among patients and the vaccines are effective only in those who produce high concentrations of anti-drug antibodies. Our multidisciplinary translational research project has generated a predominantly human sequence monoclonal antibody (mAb) with high affinity (Kd = 4 nM) for cocaine and specificity over cocaine’s inactive metabolites. This mAb (preclinical designation, 2E2) is at an advanced stage of preclinical development for preventing relapse in treatment-seeking cocaine abusers.
Development of 2E2 has met several key safety and efficacy milestones. Because the structure of mAb is mostly human, repeated treatments should be safe and should confer long-term efficacy. 2E2 binds to and sequesters cocaine in the peripheral circulation and dramatically lowers brain cocaine concentrations in mice.14 Furthermore, 2E2 decreases the effect of cocaine in a rat model of relapse.15 In FDA-required safety tests, there was no apparent cross-reactivity of 2E2 with an extensive panel of human tissues in vitro, indicating that 2E2 likely is safe for patients. The genes encoding the mAb have been cloned and slightly re-engineered to make them even closer to a human sequence and the expressed recombinant protein retains the identical affinity and specificity for cocaine. We continue to work with our industry collaborator, Vybion Inc., to develop a stably transfected mammalian cell line capable of high-level production of 2E2, which is necessary to support in vivo toxicology studies required for an FDA Investigational New Drug application and subsequent clinical trials. This anti-cocaine mAb should be a useful adjunct to TA-CD by supplementing concentrations of vaccine-generated anti-cocaine antibodies.
Andrew B. Norman, PhD
Department of psychiatry and behavioral neuroscience
William J. Ball, PhD
Department of pharmacology and cell biophysics
University of Cincinnati College of Medicine
Cincinnati, OH
Dependence on nicotine—the main addictive agent in cigarette smoke and other tobacco products—also is being targeted with vaccines. Like cocaine, the nicotine molecule is too small to provoke an immune response by itself. Therefore, nicotine derivatives linked to virus-like particles16 or detoxified bacteria-derived proteins17 are immunogenic enough to stimulate an antibody response. With once-monthly vaccinations of these conjugated nicotine compounds, patients can produce sufficient antibodies to sequester nicotine in the peripheral bloodstream before it crosses the blood-brain barrier to produce its rewarding effects.
Animal and human studies have found proof that this concept may work. These immunotherapies do not seem to provoke acute nicotine withdrawal and patients do not increase their smoking rates to try to counteract the antibodies’ nicotine-scavenging effects.16-19 As was the case with the cocaine trial,4 smoking cessation efficacy is positively correlated with the individual’s antibody titer response. Published phase I and II trials indicate that these vaccines may be safe and well-tolerated; mild reactions at the intramuscular injection site are the most commonly reported adverse event.16-18 Larger phase III clinical trials are underway.20
Robert M. Anthenelli, MD
Related Resource
- Martell BA, Mitchell E, Poling J, et al. Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry. 2005;58(2):158-164.
Drug Brand Names
- Baclofen • Lioresal
- Digoxin • Lanoxin
- Digoxin immune fab • Digibind
- Disulfiram • Antabuse
- Modafinil • Provigil
- Ondansetron • Zofran
- Tiagabine • Gabitril
Disclosure
Dr. Anthenelli receives grant/research support from Eli Lilly and Company, Nabi Biopharmaceuticals, and Pfizer Inc., and is a consultant to Pfizer Inc.
Dr. Somoza receives grant/research support from the National Institute on Drug Abuse.
Drs. Norman and Ball receive grant/research support from the National Institutes of Health and the National Institute on Drug Abuse and are consultants to Vybion, Inc.
Treatment adherence
DR. ANTHENELLI: We know from working in the addiction field that compliance with medication regimens is a big challenge. What are the data regarding adherence to TA-CD?
DR. SOMOZA: We don’t have any specific data about adherence to the vaccine, but it is probably similar to any other medication for addiction. Remember that cocaine-dependent patients often are erratic and don’t use planners to set up their day. If you look at clinical trials over the past 20 years, if you get 75% retention you’re doing really good, but quite often you see 50% or 25% retention. With TA-CD, retention is going to be worse because you have to wait 8 weeks before patients build up enough antibodies to have therapeutic effect. I’m hoping we can convince the FDA to look at the relationship between antibody generation and improvement in treatment efficacy. Obviously if patients don’t develop antibodies they’re not going to get better.
Patient characteristics
DR. ANTHENELLI: I know it’s a little early, but if you had to use your crystal ball, what type of patient do you think that TA-CD might work best for?
DR. SOMOZA: Certainly it would be for people that are motivated to stop using. If they really don’t want to stop using cocaine, probably nothing will work. These patients could get the vaccine and boosters and it won’t do them any good. They’ll take it and nothing happens.
Future research
DR. ANTHENELLI: One of the things you have discussed is who will achieve enough antibody titers to make TA-CD effective. Are there other kinds of research you think will be related to this?
DR. SOMOZA: Increasing the serum concentration of the antibodies is one. Another would be to increase the fraction of people who develop high levels of antibodies. One wonders if we could use a different protein that would increase the immunogenicity of the vaccine. If we use 2 different proteins, perhaps the effects would be additive. In an early study of mice, Fox and colleaguesa used a blood protein, not a cholera toxin.
References
a. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
1. Substance Abuse and Mental Health Services Administration. 2007 national survey on drug use and health. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2008.
2. Jertborn M, Svennerholm AM, Holmgren J. Safety and immunogenicity of an oral recombinant cholera B subunit-whole cell vaccine in Swedish volunteers. Vaccine. 1992;10:130-132.
3. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
4. Martell BA, Orson FM, Poling J, et al. Cocaine vaccine for the treatment of cocaine dependence in methadone-maintained patients: a randomized, double-blind, placebo-controlled efficacy trial. Arch Gen Psychiatry. 2009;66(10):1116-1123.
5. Haney M, Gunderson EW, Jiang H, et al. Cocaine-specific antibodies blunt the subjective effects of smoked cocaine in humans. Biol Psychiatry. 2010;67(1):59-65.
6. Kosten TR, Rosen M, Bond J, et al. Human therapeutic cocaine vaccine: safety and immunogenicity. Vaccine. 2002;20:1196-1204.
7. Grabenstein JD. ImmunoFacts: vaccines and immunological drugs. St. Louis, MO: Facts and Comparisons, Inc.; 1994:487b.
8. Pani PP, Trogu E, Vacca R, et al. Disulfiram for the treatment of cocaine dependence. Cochrane Database Syst Rev. 2010;(1):CD007024.-
9. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology. 2005;30(1):205-211.
10. Anderson AL, Reid MS, Li SH, et al. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend. 2009;104(1-2):133-139.
11. Winhusen T, Somoza E, Ciraulo DA, et al. A double-blind, placebo-controlled trial of tiagabine for the treatment of cocaine dependence. Drug Alcohol Depend. 2007;91 (2-3):141-148.
12. Kahn R, Biswas K, Childress AR, et al. Multi-center trial of baclofen for abstinence initiation in severe cocaine-dependent individuals. Drug Alcohol Depend. 2009;103 (1-2):59-64.
13. Johnson BA, Roache JD, Ait-Daoud N, et al. A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of cocaine dependence. Drug Alcohol Depend. 2006;84(3):256-263.
14. Norman AB, Tabet MR, Norman MK, et al. A chimeric human/murine anticocaine monoclonal antibody inhibits the distribution of cocaine to the brain in mice. J Pharmacol Exp Ther. 2007;320:145-153.
15. Norman AB, Norman MK, Buesing WR, et al. The effect of a chimeric human/murine anti-cocaine monoclonal antibody on cocaine self-administration in rats. J Pharmacol Exp Ther. 2009;328:873-881.
16. Cornuz J, Zwahlen S, Jungi WF, et al. A vaccine against nicotine for smoking cessation: a randomized controlled trial. PLoS One. 2008;3(6):e2547.-
17. Wagena EJ, de Vos A, Horwith G, et al. The immunogenicity and safety of a nicotine vaccine in smokers and nonsmokers: results of a randomized, placebo-controlled phase 1/2 trial. Nicotine Tob Res. 2008;10(1):213-218.
18. Hatsukami DK, Rennard S, Jorenby D, et al. Safety and immunogenicity of a nicotine conjugate vaccine in current smokers. Clin Pharmacol Ther. 2005;78(5):456-467.
19. Maurer P, Bachmann MF. Vaccination against nicotine: an emerging therapy for tobacco dependence. Expert Opin Investig Drugs. 2007;16(11):1775-1783.
20. Volkow ND. Message from the director on ARRA funding for the development of a nicotine vaccine. National Institute on Drug Abuse Web site. Available at: http://drugabuse.gov/about/welcome/nicotinevaccine909.html. Accessed August 13, 2010.
21. Laurenzana EM, Hendrickson HP, Carpenter D, et al. Functional and biological determinants affecting the duration of action and efficacy of anti-(+)-methamphetamine monoclonal antibodies in rats. Vaccine. 2009;27(50):7011-7020.
1. Substance Abuse and Mental Health Services Administration. 2007 national survey on drug use and health. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2008.
2. Jertborn M, Svennerholm AM, Holmgren J. Safety and immunogenicity of an oral recombinant cholera B subunit-whole cell vaccine in Swedish volunteers. Vaccine. 1992;10:130-132.
3. Fox BS, Kantak KM, Edwards MA, et al. Efficacy of a therapeutic cocaine vaccine in rodent models. Nat Med. 1996;2:1129-1132.
4. Martell BA, Orson FM, Poling J, et al. Cocaine vaccine for the treatment of cocaine dependence in methadone-maintained patients: a randomized, double-blind, placebo-controlled efficacy trial. Arch Gen Psychiatry. 2009;66(10):1116-1123.
5. Haney M, Gunderson EW, Jiang H, et al. Cocaine-specific antibodies blunt the subjective effects of smoked cocaine in humans. Biol Psychiatry. 2010;67(1):59-65.
6. Kosten TR, Rosen M, Bond J, et al. Human therapeutic cocaine vaccine: safety and immunogenicity. Vaccine. 2002;20:1196-1204.
7. Grabenstein JD. ImmunoFacts: vaccines and immunological drugs. St. Louis, MO: Facts and Comparisons, Inc.; 1994:487b.
8. Pani PP, Trogu E, Vacca R, et al. Disulfiram for the treatment of cocaine dependence. Cochrane Database Syst Rev. 2010;(1):CD007024.-
9. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology. 2005;30(1):205-211.
10. Anderson AL, Reid MS, Li SH, et al. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend. 2009;104(1-2):133-139.
11. Winhusen T, Somoza E, Ciraulo DA, et al. A double-blind, placebo-controlled trial of tiagabine for the treatment of cocaine dependence. Drug Alcohol Depend. 2007;91 (2-3):141-148.
12. Kahn R, Biswas K, Childress AR, et al. Multi-center trial of baclofen for abstinence initiation in severe cocaine-dependent individuals. Drug Alcohol Depend. 2009;103 (1-2):59-64.
13. Johnson BA, Roache JD, Ait-Daoud N, et al. A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of cocaine dependence. Drug Alcohol Depend. 2006;84(3):256-263.
14. Norman AB, Tabet MR, Norman MK, et al. A chimeric human/murine anticocaine monoclonal antibody inhibits the distribution of cocaine to the brain in mice. J Pharmacol Exp Ther. 2007;320:145-153.
15. Norman AB, Norman MK, Buesing WR, et al. The effect of a chimeric human/murine anti-cocaine monoclonal antibody on cocaine self-administration in rats. J Pharmacol Exp Ther. 2009;328:873-881.
16. Cornuz J, Zwahlen S, Jungi WF, et al. A vaccine against nicotine for smoking cessation: a randomized controlled trial. PLoS One. 2008;3(6):e2547.-
17. Wagena EJ, de Vos A, Horwith G, et al. The immunogenicity and safety of a nicotine vaccine in smokers and nonsmokers: results of a randomized, placebo-controlled phase 1/2 trial. Nicotine Tob Res. 2008;10(1):213-218.
18. Hatsukami DK, Rennard S, Jorenby D, et al. Safety and immunogenicity of a nicotine conjugate vaccine in current smokers. Clin Pharmacol Ther. 2005;78(5):456-467.
19. Maurer P, Bachmann MF. Vaccination against nicotine: an emerging therapy for tobacco dependence. Expert Opin Investig Drugs. 2007;16(11):1775-1783.
20. Volkow ND. Message from the director on ARRA funding for the development of a nicotine vaccine. National Institute on Drug Abuse Web site. Available at: http://drugabuse.gov/about/welcome/nicotinevaccine909.html. Accessed August 13, 2010.
21. Laurenzana EM, Hendrickson HP, Carpenter D, et al. Functional and biological determinants affecting the duration of action and efficacy of anti-(+)-methamphetamine monoclonal antibodies in rats. Vaccine. 2009;27(50):7011-7020.
Adolescents who self-harm: How to protect them from themselves
Discuss this article at http://currentpsychiatry.blogspot.com/2010/08/adolescents-who-self-harm.html#comments
Josh, age 16, gets poor grades in school and occasionally smokes marijuana and abuses inhalants. After his girlfriend breaks up with him, he cuts his wrist with a hunting knife. While bleeding profusely, Josh calls his mother at work, who calls 911. The cut is deep and requires sutures. Josh says he did not try to kill himself; he only wanted to carve his girlfriend’s initials into his wrist to show his love for her.
When treating teenagers with self-harming thoughts and behavior, it may be difficult to distinguish suicide attempts from self-injury without intent to die. Understanding adolescent self-harm, suicide risk assessment, and treatment options guides clinicians to appropriate interventions. Recognizing the need for aggressive treatment—including psychiatric hospitalization—is essential to keeping self-harming teenagers safe.
Suicidal vs nonsuicidal self-harm
Suicidal behavior involves intent to end one’s life and includes ideation (thoughts) and actions (non fatal or fatal attempts).1 Nonsuicidal self-injury (NSSI) involves socially unacceptable, self-inflicted harm to one’s body without intent to die.2
Suicide is the third leading cause of death among youths age 12 to 19, claiming almost 2,000 lives each year.3 Nearly 1 in 5 (17%) U.S. high school students has suicidal thoughts each year, and almost 1 in 10 (8%) attempts suicide.4
Studies report a 13% to 23% lifetime prevalence of NSSI.5 These behaviors often begin between age 13 to 15.6 Cutting and hitting are the most common forms of NSSI; other methods include burning, scratching, and interfering with wound healing. Most teens who harm themselves without suicidal intent report that they feel little or no pain during the act.5 Unlike suicide attempts, NSSI can be viewed as a means to stay alive. Many adolescents injure themselves to cope with overwhelming feelings that can produce suicidal thoughts. Self-injury may distract the adolescent from painful emotions, reduce tensions, or penetrate numbness.7
Teenagers who hurt themselves but do not intend to die are at high risk for suicide and suicide attempts. Adolescents who engage in NSSI are more likely to experience suicidal behaviors, and vice versa.8 In a large study, 70% of adolescents who engaged in NSSI had made at least 1 suicide attempt and 55% made multiple suicide attempts.2 Current suicidal ideation is a risk factor for suicide, and a past suicide attempt is the strongest predictor of future suicidal behavior.9
Risk factors for suicidal behavior and NSSI overlap (Table 1)2,5,6,10,11 and include:
- depression
- substance use
- anxiety
- impulsive aggression
- history of childhood trauma.
Many teens who engage in NSSI report depression.2 A history of psychiatric illness—especially depression—increases the likelihood of adolescent suicide.8 A study comparing adolescents who engaged in NSSI with those who attempted suicide found that both groups reported similar levels of suicidal ideation and depressive symptoms.6 However, adolescents with a history of NSSI and attempted suicide reported higher levels of suicidal ideation and fewer reasons for living than those who attempted suicide but have no history of NSSI.12
Factors that protect against suicidal behavior include:
- a good parent-child relationship
- strong cultural or religious values
- an intact family
- a sense of connection with peer group and community.13
No studies have determined protective factors for NSSI.
Table 1
Characteristics of teens who harm themselves
Factor | Comments |
---|---|
Older age | Both suicide attempts and NSSI are more common in mid-adolescence (age 13 to 15) |
Sex | Males complete suicide more often (4:1) but more females make attempts. Sex differences have not been consistently identified for NSSI |
Psychiatric illness | Diagnoses associated with adolescent suicide include major depression, substance abuse, and conduct disorder |
Psychosocial and situational risks (usually combined with psychiatric illness) | Recent loss or rejection, living alone (eg, running away or homeless), poor social supports, family conflicts, family suicidal behavior, poor communication with parents, availability of firearms, exposure to suicide in the community or media, academic difficulties, legal problems, gender identity conflicts, history of maltreatment, being bullied, and risky behaviors |
NSSI: nonsuicidal self-injury | |
Source: References 2,5,6,10,11 |
CASE CONTINUED: ‘No point in living’
As Josh becomes less guarded, he says that he sees “no point in living” without his girlfriend. He thought the only way to feel better was to “get high,” but this left him feeling even more despondent and anxious. He wrote a suicide note, but after cutting himself he was unsure he wanted to die. Josh says that when he feels depressed he can’t talk to his parents because “they wouldn’t understand and don’t care.”
Assessing self-harming adolescents
Distinguishing between suicidal behaviors and NSSI can be challenging (Table 2). Identifying risk factors for adolescent suicidal behavior must be coupled with a thorough psychiatric evaluation. If possible, interview the adolescent alone and obtain collateral information from parents, family members, teachers, caseworkers, probation officers, and others as needed. Also examine family interactions because conflicts and communication problems could undermine the teenager’s safety.
Consider using standardized measures of suicidal intentions such as the Scale for Suicidal Ideation (SSI).14 Although the SSI was developed for adults, a large case-control study validated the scale’s use in adolescent psychiatric outpatients and students.15 In addition to assessing subjective reports of suicidal intent, the SSI also takes into account objective indicators of increased risk, such as planning an attempt, hiding details from others, and making preparations for death.
Questions to ask. After a self-harm incident, it may be helpful to begin the psychiatric interview with a general question such as, “What happened that led you to hurt yourself?” This does not categorize the act as suicidal and allows the adolescent to describe it in his or her own words. Shea16 suggests normalizing the act by assuming that self-harm occurred, rather than making the patient admit to it. For example, an interviewer might say “Many people I know who are hurting inside also try to hurt their body; how often do you do that?”
Inquire about suicidal intent in a few ways. For example, first ask, “Do you ever wish you were dead?” and follow up with, “Would you ever do anything to try to make yourself dead?” Asking about suicidal thoughts does not increase suicidal thoughts or behavior.10,17
Reviewing thoughts and feelings leading up to a self-harm act can help identify triggers, coping difficulties, and issues to address in treatment. This behavioral analysis can be completed using the mnemonic ABC:
- Antecedents (situations or stressors leading to self-harming thoughts or actions)
- Behavior characteristics (frequency, intensity, and duration of self-harming)
- Consequences (eg, emotional relief, care and attention from others).
The results of this analysis could suggest treatment strategies, such as cognitive restructuring or techniques for decreasing feelings of distress.
The Risk of Suicide Questionnaire, which is designed for adolescents, asks:18
- Are you here because you tried to hurt yourself?
- In the past week, have you been having thoughts about killing yourself?
- Have you ever tried to hurt yourself in the past?
- Has something very stressful happened to you in the past few weeks?
Research has yet to determine whether this simple, rapid screen accurately identifies the need for psychiatric hospitalization or risk for suicidal outcomes. Although clinician- and self-administered suicide questionnaires may be useful for screening large populations, they are not a substitute for a thorough clinical assessment.
Inpatient or outpatient? When evaluating self-harming adolescents, first determine if they are in imminent danger of suicide and if more intensive services, such as hospitalization, are needed to maintain safety. Inpatient psychiatric services are appropriate for adolescents with suicidal thoughts or self-harm behaviors in addition to acute psychiatric disorders, significant substance abuse, serious medical issues, poor social supports, or inability to be managed safely as an outpatient.19 See the Table for a list of additional factors to consider.
Consent for treatment may be required because many self-harming adolescents do not present with life-threatening symptoms. Laws regarding consent vary among states. In some jurisdictions, patients age ≥15 can consent to mental health treatment without parents’ knowledge or consent. If an adolescent is in imminent danger and cannot voluntarily consent to treatment, physicians can initiate mental health “holds.” Some states allow registered nurses, psychologists, licensed social workers, and others to initiate mental health holds.
Table 2
Strategies for assessing adolescent self-harm
Complete a thorough psychiatric evaluation |
Interview the adolescent separately from parents |
Obtain collateral information from parents and family, teachers, caseworkers, and others as needed |
Use an empathic, nonjudgmental manner |
Note appearance and presence of scarring and bruises, and patient’s clothing style |
Ask about current and past self-harming thoughts and behavior:
|
Ask about acute stressors (eg, break-up, loss or rejection, conflict with parents) |
Inquire about thoughts, feelings, and events leading up to the self-harm episode |
Assess for psychosis and ask about homicidal thoughts. If yes, assess whether there is a duty to warn others |
Ask about drug/alcohol use and consider a urine toxicology screen to help clarify whether substance abuse problems may be contributing to self-harm |
Assess family interaction and communication style, noting conflicts that might impact safety |
Consider using a standardized measure, such as the Scale for Suicidal Ideation14,15 |
CASE CONTINUED: Inpatient treatment
After the interview Josh says he still feels that “there is no point in living” and he cannot develop an adequate safety plan with his family. He is hospitalized to maintain safety, improve his coping skills and communication with his family, and mobilize safety plans, social supports, and follow-up care.
Maintaining safety
Psychosocial treatments for suicidal behaviors and NSSI are similar because with both, the priority is to help the patient maintain safety. This may include:
- developing a collaborative safety plan with family
- increasing monitoring
- removing access to firearms or other lethal means
- helping the adolescent to develop alternate, safer coping methods.
Many clinicians rely on no-harm contracts or agreements; however, there is no evidence that they are effective.20 The American Psychiatric Association recommends against using no-harm contracts with patients who are new, in an emergency setting, using substances, agitated, psychotic, or impulsive.21 Instead, clinicians, adolescents, and families can discuss specific steps the patient can take to remain safe. This collaborative plan should identify situations likely to trigger self-harming impulses; adaptive ways the teenager can cope, such as taking a nap or jogging; methods for communicating distress to family members and other helpers; and places to go for help, such as an emergency room. These safety plans should draw on the patient’s internal and external resources.
CASE CONTINUED: Strengthening relationships
While in the hospital, Josh finds it helpful to use a 0-to-10 scale to measure his distress and let his family know the intensity of his feelings. He identifies situations when he felt like hurting himself, such as being humiliated in math class. Josh learns about cognitive distortions—such as “they don’t care” and “there is no point in living”—and discusses methods for managing his feelings if he encounters further disappointments. His parents become more attentive when Josh explains his feelings, which allows the family to develop a collaborative safety plan. Josh decides to strengthen friendships he had been neglecting and agrees to attend a substance abuse treatment program.
Psychosocial treatment
In addition to maintaining safety, treatment goals for self-harming adolescents include:
- managing underlying psychiatric disorders
- identifying triggers for self-injurious acts
- improving family relationships
- developing better communication and coping skills.
Improving affective language skills, acquiring frustration tolerance, and learning alternatives to self-injury are key to strengthening coping abilities. Address problem-solving skills because self-harming adolescents often lack these abilities.22
Treatment of self-harming adolescents often consists of cognitive-behavioral therapy (CBT)23 or dialectical behavior therapy (DBT).24 CBT involves examining cognitive distortions or otherwise unhealthy beliefs about oneself, others, and life in general, focusing specifically on thoughts the patient has immediately before engaging in self-harm. DBT also integrates emotion regulation training and mindfulness. A review of 28 studies found these therapies effectively reduced self-harm behaviors in adults.25 However, few studies have examined these therapies’ efficacy in self-harming adolescents.
Pharmacotherapy
Psychopharmacology should focus on treating underlying psychiatric disorders. No medications are specifically effective for treating suicidal thoughts, suicidal behaviors, or NSSI. Some evidence suggests that antidepressants may trigger suicidal thoughts in a small proportion of youth,26 but the benefits of antidepressants outweigh the risk of suicidal thoughts.27 When prescribing antidepressants, inform patients and their parents of possible adverse reactions and monitor the patient regularly.28
Take precautions when prescribing medication for self-harming adolescents. For example, benzodiazepines may cause disinhibition, and larger quantities of medication could be lethal in an overdose. If possible, arrange for parents or guardians to monitor medication use.
What to document
Good documentation is especially helpful when an adolescent requires involuntary commitment or is discharged home. Involuntary commitment is based on legal interpretation of 3 circumstances—danger to self, danger to others, or gravely disabled—in which safety concerns may override an individual’s civil rights. If involuntary commitment is needed, a physician must clarify how the youth meets ≥1 of these criteria. If the adolescent is discharged, document that the patient is not an imminent danger to self or others and why you made this determination. Also note that follow-up services and a safety plan are in place, a parent will monitor safety issues and remove firearms and other lethal means from the home, and acute conflicts have been resolved. Other details, such as using the patient’s words to describe reasons for living, can be helpful.
Related Resources
- National Alliance for the Mentally Ill. National Helpline. 800-950-6264.
- American Foundation for Suicide Prevention. www.afsp.org.
- American Association of Suicidology. www.suicidology.org.
- National Suicide Prevention Lifeline. 800-273-TALK (8255).
Acknowledgements
The authors wish to thank students Scott Schubert from Regis University, Denver, CO, and Emily Peterson from Beloit College, Beloit, WI, for their help in preparing the manuscript.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Table
Hospitalization or home? Acute crisis planning for self-harming youth
Hospitalization is more appropriate when ≥1 of the following is present with suicidal or self-injurious thoughts:
|
Home is more appropriate when:
|
1. O’Carroll PW, Berman AL, Maris RW, et al. Beyond the Tower of Babel: a nomenclature for suicidology. Suicide Life Threat Behav. 1996;26:237-252.
2. Nock MK, Joiner TE, Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
3. Centers for Disease Control and Prevention. Injury prevention and control: data and statistics (WISQARS). Available at: http://www.cdc.gov/injury/wisqars. Accessed June 22, 2010.
4. National Center for Health Statistics. Health, United States, 2006. Available at: http://www.cdc.gov/nchs/data/hus/hus06.pdf#062. Accessed May 28, 2010.
5. Jacobson CM, Gould M. The epidemiology and phenomenology of non-suicidal self-injurious behavior among adolescents: a critical review of the literature. Arch Suicide Res. 2007;11(2):129-147.
6. Muehlenkamp JJ, Gutierrez PM. An investigation of differences between self-injurious behavior and suicide attempts in a sample of adolescents. Suicide Life Threat Behav. 2004;34:12-23.
7. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41:1333-1341.
8. Whitlock J, Knox KL. The relationship between self-injurious behavior and suicide in a young adult population. Arch Pediatr Adolesc Med. 2007;161:634-640.
9. Bridge JA, Goldstein TR, Brent DA. Adolescent suicide and suicidal behavior. J Child Psychol Psychiatry. 2006;47:372-394.
10. American Academy of Child and Adolescent Psychiatry. Practice parameter for the assessment and treatment of children and adolescents with suicidal behavior. J Am Acad Child Adolesc Psychiatry. 2001;40(suppl 7):24S-51S.
11. Laye-Gindhu A, Schonert-Reichl KA. Nonsuicidal self-harm among community adolescents: understanding the “whats” and “whys” of self-harm. J Youth Adolesc. 2005;34:447-457.
12. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res. 2007;11:69-82.
13. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42:386-405.
14. Beck AT, Kovacs M, Weissmann A. Assessment of suicidal intention: the Scale for Suicide Ideation. J Consult Clin Psychol. 1979;46:343-352.
15. Holi MM, Pelkonen M, Karlsson L, et al. Psychometric properties and clinical utility of the Scale for Suicidal Ideation (SSI) in adolescents. BMC Psychiatry. 2005;5:8.-
16. Shea SC. The practical art of suicide assessment: a guide for mental health professionals and substance abuse counselors. New York, NY: John Wiley; 1999.
17. Gould MS, Marrocco FA, Kleinman M, et al. Evaluating iatrogenic risk of youth suicide screening programs. JAMA. 2005;293:1635-1643.
18. Horowitz LM, Wang PS, Koocher GP, et al. Detecting suicide risk in a pediatric emergency department: development of a brief screening tool. Pediatrics. 2001;107:1133-1137.
19. Kennedy SP, Baraff LJ, Suddath RL, et al. Emergency department management of suicidal adolescents. Ann Emerg Med. 2004;43:452-460.
20. Lewis LM. No-harm contracts: a review of what we know. Suicide Life Threat Behav. 2007;37:50-57.
21. American Psychiatric Association. Practice guideline for the assessment and treatment of patients with suicidal behavior. Am J Psychiatry. 2003;160(suppl 11):1-60.
22. Speckens AE, Hawton K. Social problem solving in adolescents with suicidal behavior: a systematic review. Suicide Life Threat Behav. 2005;35:365-387.
23. Henriques G, Beck AT, Brown GK. Cognitive therapy for adolescent and young adult suicide attempters. American Behavioral Scientist. 2003;46:1258-1268.
24. Rathus JH, Miller AL. Dialectical behavior therapy adapted for suicidal adolescents. Suicide Life Threat Behav. 2002;32:146-157.
25. Tarrier N, Taylor K, Gooding P. Cognitive-behavioral interventions to reduce suicide behavior: a systematic review and meta-analysis. Behav Modif. 2008;32:77-108.
26. Jick H, Kaye JA, Jick SS. Antidepressants and the risk of suicidal behaviors. JAMA. 2004;292:338-343.
27. 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. 2007;297(15):1683-1696.
28. Simon GE. The antidepressant quandary—considering suicide risk when treating adolescent depression. N Engl J Med. 2006;355:2722-2723.
Discuss this article at http://currentpsychiatry.blogspot.com/2010/08/adolescents-who-self-harm.html#comments
Josh, age 16, gets poor grades in school and occasionally smokes marijuana and abuses inhalants. After his girlfriend breaks up with him, he cuts his wrist with a hunting knife. While bleeding profusely, Josh calls his mother at work, who calls 911. The cut is deep and requires sutures. Josh says he did not try to kill himself; he only wanted to carve his girlfriend’s initials into his wrist to show his love for her.
When treating teenagers with self-harming thoughts and behavior, it may be difficult to distinguish suicide attempts from self-injury without intent to die. Understanding adolescent self-harm, suicide risk assessment, and treatment options guides clinicians to appropriate interventions. Recognizing the need for aggressive treatment—including psychiatric hospitalization—is essential to keeping self-harming teenagers safe.
Suicidal vs nonsuicidal self-harm
Suicidal behavior involves intent to end one’s life and includes ideation (thoughts) and actions (non fatal or fatal attempts).1 Nonsuicidal self-injury (NSSI) involves socially unacceptable, self-inflicted harm to one’s body without intent to die.2
Suicide is the third leading cause of death among youths age 12 to 19, claiming almost 2,000 lives each year.3 Nearly 1 in 5 (17%) U.S. high school students has suicidal thoughts each year, and almost 1 in 10 (8%) attempts suicide.4
Studies report a 13% to 23% lifetime prevalence of NSSI.5 These behaviors often begin between age 13 to 15.6 Cutting and hitting are the most common forms of NSSI; other methods include burning, scratching, and interfering with wound healing. Most teens who harm themselves without suicidal intent report that they feel little or no pain during the act.5 Unlike suicide attempts, NSSI can be viewed as a means to stay alive. Many adolescents injure themselves to cope with overwhelming feelings that can produce suicidal thoughts. Self-injury may distract the adolescent from painful emotions, reduce tensions, or penetrate numbness.7
Teenagers who hurt themselves but do not intend to die are at high risk for suicide and suicide attempts. Adolescents who engage in NSSI are more likely to experience suicidal behaviors, and vice versa.8 In a large study, 70% of adolescents who engaged in NSSI had made at least 1 suicide attempt and 55% made multiple suicide attempts.2 Current suicidal ideation is a risk factor for suicide, and a past suicide attempt is the strongest predictor of future suicidal behavior.9
Risk factors for suicidal behavior and NSSI overlap (Table 1)2,5,6,10,11 and include:
- depression
- substance use
- anxiety
- impulsive aggression
- history of childhood trauma.
Many teens who engage in NSSI report depression.2 A history of psychiatric illness—especially depression—increases the likelihood of adolescent suicide.8 A study comparing adolescents who engaged in NSSI with those who attempted suicide found that both groups reported similar levels of suicidal ideation and depressive symptoms.6 However, adolescents with a history of NSSI and attempted suicide reported higher levels of suicidal ideation and fewer reasons for living than those who attempted suicide but have no history of NSSI.12
Factors that protect against suicidal behavior include:
- a good parent-child relationship
- strong cultural or religious values
- an intact family
- a sense of connection with peer group and community.13
No studies have determined protective factors for NSSI.
Table 1
Characteristics of teens who harm themselves
Factor | Comments |
---|---|
Older age | Both suicide attempts and NSSI are more common in mid-adolescence (age 13 to 15) |
Sex | Males complete suicide more often (4:1) but more females make attempts. Sex differences have not been consistently identified for NSSI |
Psychiatric illness | Diagnoses associated with adolescent suicide include major depression, substance abuse, and conduct disorder |
Psychosocial and situational risks (usually combined with psychiatric illness) | Recent loss or rejection, living alone (eg, running away or homeless), poor social supports, family conflicts, family suicidal behavior, poor communication with parents, availability of firearms, exposure to suicide in the community or media, academic difficulties, legal problems, gender identity conflicts, history of maltreatment, being bullied, and risky behaviors |
NSSI: nonsuicidal self-injury | |
Source: References 2,5,6,10,11 |
CASE CONTINUED: ‘No point in living’
As Josh becomes less guarded, he says that he sees “no point in living” without his girlfriend. He thought the only way to feel better was to “get high,” but this left him feeling even more despondent and anxious. He wrote a suicide note, but after cutting himself he was unsure he wanted to die. Josh says that when he feels depressed he can’t talk to his parents because “they wouldn’t understand and don’t care.”
Assessing self-harming adolescents
Distinguishing between suicidal behaviors and NSSI can be challenging (Table 2). Identifying risk factors for adolescent suicidal behavior must be coupled with a thorough psychiatric evaluation. If possible, interview the adolescent alone and obtain collateral information from parents, family members, teachers, caseworkers, probation officers, and others as needed. Also examine family interactions because conflicts and communication problems could undermine the teenager’s safety.
Consider using standardized measures of suicidal intentions such as the Scale for Suicidal Ideation (SSI).14 Although the SSI was developed for adults, a large case-control study validated the scale’s use in adolescent psychiatric outpatients and students.15 In addition to assessing subjective reports of suicidal intent, the SSI also takes into account objective indicators of increased risk, such as planning an attempt, hiding details from others, and making preparations for death.
Questions to ask. After a self-harm incident, it may be helpful to begin the psychiatric interview with a general question such as, “What happened that led you to hurt yourself?” This does not categorize the act as suicidal and allows the adolescent to describe it in his or her own words. Shea16 suggests normalizing the act by assuming that self-harm occurred, rather than making the patient admit to it. For example, an interviewer might say “Many people I know who are hurting inside also try to hurt their body; how often do you do that?”
Inquire about suicidal intent in a few ways. For example, first ask, “Do you ever wish you were dead?” and follow up with, “Would you ever do anything to try to make yourself dead?” Asking about suicidal thoughts does not increase suicidal thoughts or behavior.10,17
Reviewing thoughts and feelings leading up to a self-harm act can help identify triggers, coping difficulties, and issues to address in treatment. This behavioral analysis can be completed using the mnemonic ABC:
- Antecedents (situations or stressors leading to self-harming thoughts or actions)
- Behavior characteristics (frequency, intensity, and duration of self-harming)
- Consequences (eg, emotional relief, care and attention from others).
The results of this analysis could suggest treatment strategies, such as cognitive restructuring or techniques for decreasing feelings of distress.
The Risk of Suicide Questionnaire, which is designed for adolescents, asks:18
- Are you here because you tried to hurt yourself?
- In the past week, have you been having thoughts about killing yourself?
- Have you ever tried to hurt yourself in the past?
- Has something very stressful happened to you in the past few weeks?
Research has yet to determine whether this simple, rapid screen accurately identifies the need for psychiatric hospitalization or risk for suicidal outcomes. Although clinician- and self-administered suicide questionnaires may be useful for screening large populations, they are not a substitute for a thorough clinical assessment.
Inpatient or outpatient? When evaluating self-harming adolescents, first determine if they are in imminent danger of suicide and if more intensive services, such as hospitalization, are needed to maintain safety. Inpatient psychiatric services are appropriate for adolescents with suicidal thoughts or self-harm behaviors in addition to acute psychiatric disorders, significant substance abuse, serious medical issues, poor social supports, or inability to be managed safely as an outpatient.19 See the Table for a list of additional factors to consider.
Consent for treatment may be required because many self-harming adolescents do not present with life-threatening symptoms. Laws regarding consent vary among states. In some jurisdictions, patients age ≥15 can consent to mental health treatment without parents’ knowledge or consent. If an adolescent is in imminent danger and cannot voluntarily consent to treatment, physicians can initiate mental health “holds.” Some states allow registered nurses, psychologists, licensed social workers, and others to initiate mental health holds.
Table 2
Strategies for assessing adolescent self-harm
Complete a thorough psychiatric evaluation |
Interview the adolescent separately from parents |
Obtain collateral information from parents and family, teachers, caseworkers, and others as needed |
Use an empathic, nonjudgmental manner |
Note appearance and presence of scarring and bruises, and patient’s clothing style |
Ask about current and past self-harming thoughts and behavior:
|
Ask about acute stressors (eg, break-up, loss or rejection, conflict with parents) |
Inquire about thoughts, feelings, and events leading up to the self-harm episode |
Assess for psychosis and ask about homicidal thoughts. If yes, assess whether there is a duty to warn others |
Ask about drug/alcohol use and consider a urine toxicology screen to help clarify whether substance abuse problems may be contributing to self-harm |
Assess family interaction and communication style, noting conflicts that might impact safety |
Consider using a standardized measure, such as the Scale for Suicidal Ideation14,15 |
CASE CONTINUED: Inpatient treatment
After the interview Josh says he still feels that “there is no point in living” and he cannot develop an adequate safety plan with his family. He is hospitalized to maintain safety, improve his coping skills and communication with his family, and mobilize safety plans, social supports, and follow-up care.
Maintaining safety
Psychosocial treatments for suicidal behaviors and NSSI are similar because with both, the priority is to help the patient maintain safety. This may include:
- developing a collaborative safety plan with family
- increasing monitoring
- removing access to firearms or other lethal means
- helping the adolescent to develop alternate, safer coping methods.
Many clinicians rely on no-harm contracts or agreements; however, there is no evidence that they are effective.20 The American Psychiatric Association recommends against using no-harm contracts with patients who are new, in an emergency setting, using substances, agitated, psychotic, or impulsive.21 Instead, clinicians, adolescents, and families can discuss specific steps the patient can take to remain safe. This collaborative plan should identify situations likely to trigger self-harming impulses; adaptive ways the teenager can cope, such as taking a nap or jogging; methods for communicating distress to family members and other helpers; and places to go for help, such as an emergency room. These safety plans should draw on the patient’s internal and external resources.
CASE CONTINUED: Strengthening relationships
While in the hospital, Josh finds it helpful to use a 0-to-10 scale to measure his distress and let his family know the intensity of his feelings. He identifies situations when he felt like hurting himself, such as being humiliated in math class. Josh learns about cognitive distortions—such as “they don’t care” and “there is no point in living”—and discusses methods for managing his feelings if he encounters further disappointments. His parents become more attentive when Josh explains his feelings, which allows the family to develop a collaborative safety plan. Josh decides to strengthen friendships he had been neglecting and agrees to attend a substance abuse treatment program.
Psychosocial treatment
In addition to maintaining safety, treatment goals for self-harming adolescents include:
- managing underlying psychiatric disorders
- identifying triggers for self-injurious acts
- improving family relationships
- developing better communication and coping skills.
Improving affective language skills, acquiring frustration tolerance, and learning alternatives to self-injury are key to strengthening coping abilities. Address problem-solving skills because self-harming adolescents often lack these abilities.22
Treatment of self-harming adolescents often consists of cognitive-behavioral therapy (CBT)23 or dialectical behavior therapy (DBT).24 CBT involves examining cognitive distortions or otherwise unhealthy beliefs about oneself, others, and life in general, focusing specifically on thoughts the patient has immediately before engaging in self-harm. DBT also integrates emotion regulation training and mindfulness. A review of 28 studies found these therapies effectively reduced self-harm behaviors in adults.25 However, few studies have examined these therapies’ efficacy in self-harming adolescents.
Pharmacotherapy
Psychopharmacology should focus on treating underlying psychiatric disorders. No medications are specifically effective for treating suicidal thoughts, suicidal behaviors, or NSSI. Some evidence suggests that antidepressants may trigger suicidal thoughts in a small proportion of youth,26 but the benefits of antidepressants outweigh the risk of suicidal thoughts.27 When prescribing antidepressants, inform patients and their parents of possible adverse reactions and monitor the patient regularly.28
Take precautions when prescribing medication for self-harming adolescents. For example, benzodiazepines may cause disinhibition, and larger quantities of medication could be lethal in an overdose. If possible, arrange for parents or guardians to monitor medication use.
What to document
Good documentation is especially helpful when an adolescent requires involuntary commitment or is discharged home. Involuntary commitment is based on legal interpretation of 3 circumstances—danger to self, danger to others, or gravely disabled—in which safety concerns may override an individual’s civil rights. If involuntary commitment is needed, a physician must clarify how the youth meets ≥1 of these criteria. If the adolescent is discharged, document that the patient is not an imminent danger to self or others and why you made this determination. Also note that follow-up services and a safety plan are in place, a parent will monitor safety issues and remove firearms and other lethal means from the home, and acute conflicts have been resolved. Other details, such as using the patient’s words to describe reasons for living, can be helpful.
Related Resources
- National Alliance for the Mentally Ill. National Helpline. 800-950-6264.
- American Foundation for Suicide Prevention. www.afsp.org.
- American Association of Suicidology. www.suicidology.org.
- National Suicide Prevention Lifeline. 800-273-TALK (8255).
Acknowledgements
The authors wish to thank students Scott Schubert from Regis University, Denver, CO, and Emily Peterson from Beloit College, Beloit, WI, for their help in preparing the manuscript.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Table
Hospitalization or home? Acute crisis planning for self-harming youth
Hospitalization is more appropriate when ≥1 of the following is present with suicidal or self-injurious thoughts:
|
Home is more appropriate when:
|
Discuss this article at http://currentpsychiatry.blogspot.com/2010/08/adolescents-who-self-harm.html#comments
Josh, age 16, gets poor grades in school and occasionally smokes marijuana and abuses inhalants. After his girlfriend breaks up with him, he cuts his wrist with a hunting knife. While bleeding profusely, Josh calls his mother at work, who calls 911. The cut is deep and requires sutures. Josh says he did not try to kill himself; he only wanted to carve his girlfriend’s initials into his wrist to show his love for her.
When treating teenagers with self-harming thoughts and behavior, it may be difficult to distinguish suicide attempts from self-injury without intent to die. Understanding adolescent self-harm, suicide risk assessment, and treatment options guides clinicians to appropriate interventions. Recognizing the need for aggressive treatment—including psychiatric hospitalization—is essential to keeping self-harming teenagers safe.
Suicidal vs nonsuicidal self-harm
Suicidal behavior involves intent to end one’s life and includes ideation (thoughts) and actions (non fatal or fatal attempts).1 Nonsuicidal self-injury (NSSI) involves socially unacceptable, self-inflicted harm to one’s body without intent to die.2
Suicide is the third leading cause of death among youths age 12 to 19, claiming almost 2,000 lives each year.3 Nearly 1 in 5 (17%) U.S. high school students has suicidal thoughts each year, and almost 1 in 10 (8%) attempts suicide.4
Studies report a 13% to 23% lifetime prevalence of NSSI.5 These behaviors often begin between age 13 to 15.6 Cutting and hitting are the most common forms of NSSI; other methods include burning, scratching, and interfering with wound healing. Most teens who harm themselves without suicidal intent report that they feel little or no pain during the act.5 Unlike suicide attempts, NSSI can be viewed as a means to stay alive. Many adolescents injure themselves to cope with overwhelming feelings that can produce suicidal thoughts. Self-injury may distract the adolescent from painful emotions, reduce tensions, or penetrate numbness.7
Teenagers who hurt themselves but do not intend to die are at high risk for suicide and suicide attempts. Adolescents who engage in NSSI are more likely to experience suicidal behaviors, and vice versa.8 In a large study, 70% of adolescents who engaged in NSSI had made at least 1 suicide attempt and 55% made multiple suicide attempts.2 Current suicidal ideation is a risk factor for suicide, and a past suicide attempt is the strongest predictor of future suicidal behavior.9
Risk factors for suicidal behavior and NSSI overlap (Table 1)2,5,6,10,11 and include:
- depression
- substance use
- anxiety
- impulsive aggression
- history of childhood trauma.
Many teens who engage in NSSI report depression.2 A history of psychiatric illness—especially depression—increases the likelihood of adolescent suicide.8 A study comparing adolescents who engaged in NSSI with those who attempted suicide found that both groups reported similar levels of suicidal ideation and depressive symptoms.6 However, adolescents with a history of NSSI and attempted suicide reported higher levels of suicidal ideation and fewer reasons for living than those who attempted suicide but have no history of NSSI.12
Factors that protect against suicidal behavior include:
- a good parent-child relationship
- strong cultural or religious values
- an intact family
- a sense of connection with peer group and community.13
No studies have determined protective factors for NSSI.
Table 1
Characteristics of teens who harm themselves
Factor | Comments |
---|---|
Older age | Both suicide attempts and NSSI are more common in mid-adolescence (age 13 to 15) |
Sex | Males complete suicide more often (4:1) but more females make attempts. Sex differences have not been consistently identified for NSSI |
Psychiatric illness | Diagnoses associated with adolescent suicide include major depression, substance abuse, and conduct disorder |
Psychosocial and situational risks (usually combined with psychiatric illness) | Recent loss or rejection, living alone (eg, running away or homeless), poor social supports, family conflicts, family suicidal behavior, poor communication with parents, availability of firearms, exposure to suicide in the community or media, academic difficulties, legal problems, gender identity conflicts, history of maltreatment, being bullied, and risky behaviors |
NSSI: nonsuicidal self-injury | |
Source: References 2,5,6,10,11 |
CASE CONTINUED: ‘No point in living’
As Josh becomes less guarded, he says that he sees “no point in living” without his girlfriend. He thought the only way to feel better was to “get high,” but this left him feeling even more despondent and anxious. He wrote a suicide note, but after cutting himself he was unsure he wanted to die. Josh says that when he feels depressed he can’t talk to his parents because “they wouldn’t understand and don’t care.”
Assessing self-harming adolescents
Distinguishing between suicidal behaviors and NSSI can be challenging (Table 2). Identifying risk factors for adolescent suicidal behavior must be coupled with a thorough psychiatric evaluation. If possible, interview the adolescent alone and obtain collateral information from parents, family members, teachers, caseworkers, probation officers, and others as needed. Also examine family interactions because conflicts and communication problems could undermine the teenager’s safety.
Consider using standardized measures of suicidal intentions such as the Scale for Suicidal Ideation (SSI).14 Although the SSI was developed for adults, a large case-control study validated the scale’s use in adolescent psychiatric outpatients and students.15 In addition to assessing subjective reports of suicidal intent, the SSI also takes into account objective indicators of increased risk, such as planning an attempt, hiding details from others, and making preparations for death.
Questions to ask. After a self-harm incident, it may be helpful to begin the psychiatric interview with a general question such as, “What happened that led you to hurt yourself?” This does not categorize the act as suicidal and allows the adolescent to describe it in his or her own words. Shea16 suggests normalizing the act by assuming that self-harm occurred, rather than making the patient admit to it. For example, an interviewer might say “Many people I know who are hurting inside also try to hurt their body; how often do you do that?”
Inquire about suicidal intent in a few ways. For example, first ask, “Do you ever wish you were dead?” and follow up with, “Would you ever do anything to try to make yourself dead?” Asking about suicidal thoughts does not increase suicidal thoughts or behavior.10,17
Reviewing thoughts and feelings leading up to a self-harm act can help identify triggers, coping difficulties, and issues to address in treatment. This behavioral analysis can be completed using the mnemonic ABC:
- Antecedents (situations or stressors leading to self-harming thoughts or actions)
- Behavior characteristics (frequency, intensity, and duration of self-harming)
- Consequences (eg, emotional relief, care and attention from others).
The results of this analysis could suggest treatment strategies, such as cognitive restructuring or techniques for decreasing feelings of distress.
The Risk of Suicide Questionnaire, which is designed for adolescents, asks:18
- Are you here because you tried to hurt yourself?
- In the past week, have you been having thoughts about killing yourself?
- Have you ever tried to hurt yourself in the past?
- Has something very stressful happened to you in the past few weeks?
Research has yet to determine whether this simple, rapid screen accurately identifies the need for psychiatric hospitalization or risk for suicidal outcomes. Although clinician- and self-administered suicide questionnaires may be useful for screening large populations, they are not a substitute for a thorough clinical assessment.
Inpatient or outpatient? When evaluating self-harming adolescents, first determine if they are in imminent danger of suicide and if more intensive services, such as hospitalization, are needed to maintain safety. Inpatient psychiatric services are appropriate for adolescents with suicidal thoughts or self-harm behaviors in addition to acute psychiatric disorders, significant substance abuse, serious medical issues, poor social supports, or inability to be managed safely as an outpatient.19 See the Table for a list of additional factors to consider.
Consent for treatment may be required because many self-harming adolescents do not present with life-threatening symptoms. Laws regarding consent vary among states. In some jurisdictions, patients age ≥15 can consent to mental health treatment without parents’ knowledge or consent. If an adolescent is in imminent danger and cannot voluntarily consent to treatment, physicians can initiate mental health “holds.” Some states allow registered nurses, psychologists, licensed social workers, and others to initiate mental health holds.
Table 2
Strategies for assessing adolescent self-harm
Complete a thorough psychiatric evaluation |
Interview the adolescent separately from parents |
Obtain collateral information from parents and family, teachers, caseworkers, and others as needed |
Use an empathic, nonjudgmental manner |
Note appearance and presence of scarring and bruises, and patient’s clothing style |
Ask about current and past self-harming thoughts and behavior:
|
Ask about acute stressors (eg, break-up, loss or rejection, conflict with parents) |
Inquire about thoughts, feelings, and events leading up to the self-harm episode |
Assess for psychosis and ask about homicidal thoughts. If yes, assess whether there is a duty to warn others |
Ask about drug/alcohol use and consider a urine toxicology screen to help clarify whether substance abuse problems may be contributing to self-harm |
Assess family interaction and communication style, noting conflicts that might impact safety |
Consider using a standardized measure, such as the Scale for Suicidal Ideation14,15 |
CASE CONTINUED: Inpatient treatment
After the interview Josh says he still feels that “there is no point in living” and he cannot develop an adequate safety plan with his family. He is hospitalized to maintain safety, improve his coping skills and communication with his family, and mobilize safety plans, social supports, and follow-up care.
Maintaining safety
Psychosocial treatments for suicidal behaviors and NSSI are similar because with both, the priority is to help the patient maintain safety. This may include:
- developing a collaborative safety plan with family
- increasing monitoring
- removing access to firearms or other lethal means
- helping the adolescent to develop alternate, safer coping methods.
Many clinicians rely on no-harm contracts or agreements; however, there is no evidence that they are effective.20 The American Psychiatric Association recommends against using no-harm contracts with patients who are new, in an emergency setting, using substances, agitated, psychotic, or impulsive.21 Instead, clinicians, adolescents, and families can discuss specific steps the patient can take to remain safe. This collaborative plan should identify situations likely to trigger self-harming impulses; adaptive ways the teenager can cope, such as taking a nap or jogging; methods for communicating distress to family members and other helpers; and places to go for help, such as an emergency room. These safety plans should draw on the patient’s internal and external resources.
CASE CONTINUED: Strengthening relationships
While in the hospital, Josh finds it helpful to use a 0-to-10 scale to measure his distress and let his family know the intensity of his feelings. He identifies situations when he felt like hurting himself, such as being humiliated in math class. Josh learns about cognitive distortions—such as “they don’t care” and “there is no point in living”—and discusses methods for managing his feelings if he encounters further disappointments. His parents become more attentive when Josh explains his feelings, which allows the family to develop a collaborative safety plan. Josh decides to strengthen friendships he had been neglecting and agrees to attend a substance abuse treatment program.
Psychosocial treatment
In addition to maintaining safety, treatment goals for self-harming adolescents include:
- managing underlying psychiatric disorders
- identifying triggers for self-injurious acts
- improving family relationships
- developing better communication and coping skills.
Improving affective language skills, acquiring frustration tolerance, and learning alternatives to self-injury are key to strengthening coping abilities. Address problem-solving skills because self-harming adolescents often lack these abilities.22
Treatment of self-harming adolescents often consists of cognitive-behavioral therapy (CBT)23 or dialectical behavior therapy (DBT).24 CBT involves examining cognitive distortions or otherwise unhealthy beliefs about oneself, others, and life in general, focusing specifically on thoughts the patient has immediately before engaging in self-harm. DBT also integrates emotion regulation training and mindfulness. A review of 28 studies found these therapies effectively reduced self-harm behaviors in adults.25 However, few studies have examined these therapies’ efficacy in self-harming adolescents.
Pharmacotherapy
Psychopharmacology should focus on treating underlying psychiatric disorders. No medications are specifically effective for treating suicidal thoughts, suicidal behaviors, or NSSI. Some evidence suggests that antidepressants may trigger suicidal thoughts in a small proportion of youth,26 but the benefits of antidepressants outweigh the risk of suicidal thoughts.27 When prescribing antidepressants, inform patients and their parents of possible adverse reactions and monitor the patient regularly.28
Take precautions when prescribing medication for self-harming adolescents. For example, benzodiazepines may cause disinhibition, and larger quantities of medication could be lethal in an overdose. If possible, arrange for parents or guardians to monitor medication use.
What to document
Good documentation is especially helpful when an adolescent requires involuntary commitment or is discharged home. Involuntary commitment is based on legal interpretation of 3 circumstances—danger to self, danger to others, or gravely disabled—in which safety concerns may override an individual’s civil rights. If involuntary commitment is needed, a physician must clarify how the youth meets ≥1 of these criteria. If the adolescent is discharged, document that the patient is not an imminent danger to self or others and why you made this determination. Also note that follow-up services and a safety plan are in place, a parent will monitor safety issues and remove firearms and other lethal means from the home, and acute conflicts have been resolved. Other details, such as using the patient’s words to describe reasons for living, can be helpful.
Related Resources
- National Alliance for the Mentally Ill. National Helpline. 800-950-6264.
- American Foundation for Suicide Prevention. www.afsp.org.
- American Association of Suicidology. www.suicidology.org.
- National Suicide Prevention Lifeline. 800-273-TALK (8255).
Acknowledgements
The authors wish to thank students Scott Schubert from Regis University, Denver, CO, and Emily Peterson from Beloit College, Beloit, WI, for their help in preparing the manuscript.
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Table
Hospitalization or home? Acute crisis planning for self-harming youth
Hospitalization is more appropriate when ≥1 of the following is present with suicidal or self-injurious thoughts:
|
Home is more appropriate when:
|
1. O’Carroll PW, Berman AL, Maris RW, et al. Beyond the Tower of Babel: a nomenclature for suicidology. Suicide Life Threat Behav. 1996;26:237-252.
2. Nock MK, Joiner TE, Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
3. Centers for Disease Control and Prevention. Injury prevention and control: data and statistics (WISQARS). Available at: http://www.cdc.gov/injury/wisqars. Accessed June 22, 2010.
4. National Center for Health Statistics. Health, United States, 2006. Available at: http://www.cdc.gov/nchs/data/hus/hus06.pdf#062. Accessed May 28, 2010.
5. Jacobson CM, Gould M. The epidemiology and phenomenology of non-suicidal self-injurious behavior among adolescents: a critical review of the literature. Arch Suicide Res. 2007;11(2):129-147.
6. Muehlenkamp JJ, Gutierrez PM. An investigation of differences between self-injurious behavior and suicide attempts in a sample of adolescents. Suicide Life Threat Behav. 2004;34:12-23.
7. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41:1333-1341.
8. Whitlock J, Knox KL. The relationship between self-injurious behavior and suicide in a young adult population. Arch Pediatr Adolesc Med. 2007;161:634-640.
9. Bridge JA, Goldstein TR, Brent DA. Adolescent suicide and suicidal behavior. J Child Psychol Psychiatry. 2006;47:372-394.
10. American Academy of Child and Adolescent Psychiatry. Practice parameter for the assessment and treatment of children and adolescents with suicidal behavior. J Am Acad Child Adolesc Psychiatry. 2001;40(suppl 7):24S-51S.
11. Laye-Gindhu A, Schonert-Reichl KA. Nonsuicidal self-harm among community adolescents: understanding the “whats” and “whys” of self-harm. J Youth Adolesc. 2005;34:447-457.
12. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res. 2007;11:69-82.
13. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42:386-405.
14. Beck AT, Kovacs M, Weissmann A. Assessment of suicidal intention: the Scale for Suicide Ideation. J Consult Clin Psychol. 1979;46:343-352.
15. Holi MM, Pelkonen M, Karlsson L, et al. Psychometric properties and clinical utility of the Scale for Suicidal Ideation (SSI) in adolescents. BMC Psychiatry. 2005;5:8.-
16. Shea SC. The practical art of suicide assessment: a guide for mental health professionals and substance abuse counselors. New York, NY: John Wiley; 1999.
17. Gould MS, Marrocco FA, Kleinman M, et al. Evaluating iatrogenic risk of youth suicide screening programs. JAMA. 2005;293:1635-1643.
18. Horowitz LM, Wang PS, Koocher GP, et al. Detecting suicide risk in a pediatric emergency department: development of a brief screening tool. Pediatrics. 2001;107:1133-1137.
19. Kennedy SP, Baraff LJ, Suddath RL, et al. Emergency department management of suicidal adolescents. Ann Emerg Med. 2004;43:452-460.
20. Lewis LM. No-harm contracts: a review of what we know. Suicide Life Threat Behav. 2007;37:50-57.
21. American Psychiatric Association. Practice guideline for the assessment and treatment of patients with suicidal behavior. Am J Psychiatry. 2003;160(suppl 11):1-60.
22. Speckens AE, Hawton K. Social problem solving in adolescents with suicidal behavior: a systematic review. Suicide Life Threat Behav. 2005;35:365-387.
23. Henriques G, Beck AT, Brown GK. Cognitive therapy for adolescent and young adult suicide attempters. American Behavioral Scientist. 2003;46:1258-1268.
24. Rathus JH, Miller AL. Dialectical behavior therapy adapted for suicidal adolescents. Suicide Life Threat Behav. 2002;32:146-157.
25. Tarrier N, Taylor K, Gooding P. Cognitive-behavioral interventions to reduce suicide behavior: a systematic review and meta-analysis. Behav Modif. 2008;32:77-108.
26. Jick H, Kaye JA, Jick SS. Antidepressants and the risk of suicidal behaviors. JAMA. 2004;292:338-343.
27. 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. 2007;297(15):1683-1696.
28. Simon GE. The antidepressant quandary—considering suicide risk when treating adolescent depression. N Engl J Med. 2006;355:2722-2723.
1. O’Carroll PW, Berman AL, Maris RW, et al. Beyond the Tower of Babel: a nomenclature for suicidology. Suicide Life Threat Behav. 1996;26:237-252.
2. Nock MK, Joiner TE, Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
3. Centers for Disease Control and Prevention. Injury prevention and control: data and statistics (WISQARS). Available at: http://www.cdc.gov/injury/wisqars. Accessed June 22, 2010.
4. National Center for Health Statistics. Health, United States, 2006. Available at: http://www.cdc.gov/nchs/data/hus/hus06.pdf#062. Accessed May 28, 2010.
5. Jacobson CM, Gould M. The epidemiology and phenomenology of non-suicidal self-injurious behavior among adolescents: a critical review of the literature. Arch Suicide Res. 2007;11(2):129-147.
6. Muehlenkamp JJ, Gutierrez PM. An investigation of differences between self-injurious behavior and suicide attempts in a sample of adolescents. Suicide Life Threat Behav. 2004;34:12-23.
7. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41:1333-1341.
8. Whitlock J, Knox KL. The relationship between self-injurious behavior and suicide in a young adult population. Arch Pediatr Adolesc Med. 2007;161:634-640.
9. Bridge JA, Goldstein TR, Brent DA. Adolescent suicide and suicidal behavior. J Child Psychol Psychiatry. 2006;47:372-394.
10. American Academy of Child and Adolescent Psychiatry. Practice parameter for the assessment and treatment of children and adolescents with suicidal behavior. J Am Acad Child Adolesc Psychiatry. 2001;40(suppl 7):24S-51S.
11. Laye-Gindhu A, Schonert-Reichl KA. Nonsuicidal self-harm among community adolescents: understanding the “whats” and “whys” of self-harm. J Youth Adolesc. 2005;34:447-457.
12. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res. 2007;11:69-82.
13. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42:386-405.
14. Beck AT, Kovacs M, Weissmann A. Assessment of suicidal intention: the Scale for Suicide Ideation. J Consult Clin Psychol. 1979;46:343-352.
15. Holi MM, Pelkonen M, Karlsson L, et al. Psychometric properties and clinical utility of the Scale for Suicidal Ideation (SSI) in adolescents. BMC Psychiatry. 2005;5:8.-
16. Shea SC. The practical art of suicide assessment: a guide for mental health professionals and substance abuse counselors. New York, NY: John Wiley; 1999.
17. Gould MS, Marrocco FA, Kleinman M, et al. Evaluating iatrogenic risk of youth suicide screening programs. JAMA. 2005;293:1635-1643.
18. Horowitz LM, Wang PS, Koocher GP, et al. Detecting suicide risk in a pediatric emergency department: development of a brief screening tool. Pediatrics. 2001;107:1133-1137.
19. Kennedy SP, Baraff LJ, Suddath RL, et al. Emergency department management of suicidal adolescents. Ann Emerg Med. 2004;43:452-460.
20. Lewis LM. No-harm contracts: a review of what we know. Suicide Life Threat Behav. 2007;37:50-57.
21. American Psychiatric Association. Practice guideline for the assessment and treatment of patients with suicidal behavior. Am J Psychiatry. 2003;160(suppl 11):1-60.
22. Speckens AE, Hawton K. Social problem solving in adolescents with suicidal behavior: a systematic review. Suicide Life Threat Behav. 2005;35:365-387.
23. Henriques G, Beck AT, Brown GK. Cognitive therapy for adolescent and young adult suicide attempters. American Behavioral Scientist. 2003;46:1258-1268.
24. Rathus JH, Miller AL. Dialectical behavior therapy adapted for suicidal adolescents. Suicide Life Threat Behav. 2002;32:146-157.
25. Tarrier N, Taylor K, Gooding P. Cognitive-behavioral interventions to reduce suicide behavior: a systematic review and meta-analysis. Behav Modif. 2008;32:77-108.
26. Jick H, Kaye JA, Jick SS. Antidepressants and the risk of suicidal behaviors. JAMA. 2004;292:338-343.
27. 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. 2007;297(15):1683-1696.
28. Simon GE. The antidepressant quandary—considering suicide risk when treating adolescent depression. N Engl J Med. 2006;355:2722-2723.
Bipolar disorder and substance abuse: Overcome the challenges of ‘dual diagnosis’ patients
After testing positive for cocaine on a recent court-mandated urine drug screen, Mr. M, age 49, is referred by his parole officer for psychiatric and substance abuse treatment. Mr. M has bipolar I disorder and alcohol, cocaine, and opioid dependence. He says he has been hospitalized or incarcerated at least once each year for the past 22 years. Mr. M has seen numerous psychiatrists as an outpatient, but rarely for more than 2 to 3 months and has not taken any psychotropics for more than 5 months.
Approximately 1 week before his recent urine drug screen, Mr. M became euphoric, stopped sleeping for several days, and spent $2,000 on cocaine and “escorts.” He reports that each day he smokes 30 cigarettes and drinks 1 pint of liquor and prefers to use cocaine and opiates by IV injection. Several years ago Mr. M was diagnosed with hepatitis C virus (HCV) but received no further workup or treatment. Although he denies manic or psychotic symptoms, Mr. M is observed speaking to unseen others in the waiting room and has difficulty remaining still during his interview. His chief concern is insomnia, stating, “Doc, I just need something to help me sleep.”
The high prevalence of substance use disorders (SUDs) in persons with bipolar disorder (BD) is well documented.1,2 Up to 60% of bipolar patients develop an SUD at some point in their lives.3-5 Alcohol use disorders are particularly common among BD patients, with a lifetime prevalence of roughly 50%.2-5 Recent epidemiologic data indicate that 38% of persons with bipolar I disorder and 19% of those with bipolar II disorder meet criteria for alcohol dependence.5 Comorbid SUDs in patients with BD are associated with:
- poor treatment compliance
- longer and more frequent mood episodes
- more mixed episodes
- more hospitalizations
- more frequent suicide attempts.1,2
The impact of co-occurring SUDs on suicidality is particularly high among those with bipolar I disorder.6
Frequently referred to as “dual diagnosis” conditions, co-occurring BD and SUDs may be more accurately envisioned as multi-morbid, rather than comorbid, illnesses. Data from the Stanley Foundation Bipolar Network suggest that 42% of BD patients have a lifetime history of ≥2 comorbid axis I disorders.7 Rates of generalized anxiety disorder, panic disorder, and posttraumatic stress disorder are particularly high in BD patients with co-occurring SUDs.8,9 In addition, the presence of 1 SUD may mark the presence of other SUDs; for example, alcohol dependence is strongly associated with polysubstance abuse, especially in females with BD.8 Furthermore, medical comorbidities that impact treatment decisions also are highly prevalent in BD patients with comorbid SUDs.10 In particular, HCV rates are higher in persons with BD compared with the general population,11 and are >5 times as likely in bipolar patients with co-occurring SUDs.12
Unfortunately, limited treatment research guides clinical management of comorbid BD and SUDs.2,13,14 Clinical trials of medications for BD traditionally have excluded patients with SUDs, and persons with serious mental illness usually are ineligible for SUD treatment studies.13 Furthermore, the few randomized controlled trials (RCTs) conducted in persons with both illnesses have been constrained by relatively small sample sizes and low retention rates. In the absence of a definitive consensus for optimal treatment, this article outlines general clinical considerations and an integrated approach to assessing and managing this complex patient population.
Birds of a feather
Multiple hypotheses try to account for the high rate of SUDs in patients with BD (Table 1), but none fully explain the complex interaction observed clinically.14,15 Substance dependence and BD are chronic remitting/relapsing disorders with heterogeneous presentations and highly variable natural histories. As with SUDs, BD may go undiagnosed and untreated for years, coming to clinical attention only after substantial disease progression.16
The fluctuating illness course in BD and SUDs makes diagnosis and treatment difficult. Symptomatic periods often are interrupted by spontaneous remissions and longer—although usually temporary—periods of perceived control. Both BD and SUDs may be associated with profound mood instability, increased impulsivity, altered responsiveness to reward, and impaired executive function.17 Finally, the high degree of heritability in BD18 and many SUDs19 may make treatment engagement more difficult if either disorder is present in multiple family members because of:
- potential for greater clinical severity
- reduced psychosocial resources
- altered familial behavioral norms that may impede the patient’s recognition of illness.
Denial of illness is a critical symptom that may fluctuate with disease course in both disorders. Persons with BD or SUDs may be least likely to recognize that they are ill during periods of highest symptom severity. Accordingly, treatment adherence in patients with either disorder may be limited at baseline and decline further when the 2 illnesses co-occur.20
Involvement in the criminal justice system and medical comorbidities, particularly HCV, also complicate diagnosis and treatment of BD patients with SUDs. For more information about these topics, see Box1 and Box2.
Table 1
Why is substance abuse so prevalent among bipolar patients?
Proposed hypothesis | Selected limitations of this hypothesis |
---|---|
Self-medication: substance abuse occurs as an attempt to regulate mood | High rates of substance use during euthymia; high prevalence of alcohol/depressant use during depressive phase, stimulant use during manic phase |
Common neurobiologic or genetic risk factors | Specific evidence from linkage/association studies currently is lacking |
Substance use occurs as a symptom of bipolar disorder | High percentage of patients with bipolar disorder do not have SUDs; there is a poor correlation of onset, course of bipolar, and SUD symptoms |
Substance use unmasks bipolar disorder or a bipolar diathesis | Emergence of mania before SUD is common and predictive of more severe course of bipolar disorder |
High comorbidity rates are an artifact of misdiagnosis based on overlapping symptoms and poor diagnostic boundaries | Very high prevalence of SUDs also is observed in longitudinal studies of patients initially hospitalized for mania |
SUDs: substance use disorders | |
Source: References 14,15 |
Integrated clinical management
Assessment. Although not intended to be comprehensive, suggestions for routine assessment of patients with suspected SUDs and/or BD are listed in Table 2. Because clinicians may encounter dual diagnosis patients in general psychiatric clinics or specialty (addiction or mood disorder) clinics, it is useful to obtain a thorough substance use history in all patients with known or suspected BD as well as a thorough history of hypomania/mania and depression in all patients with addictive disorders. BD diagnoses by self-report or chart history in patients with SUDs should be considered cautiously because BD often is overdiagnosed in persons engaged in active substance abuse or experiencing withdrawal.21 If past or present mood symptoms and substance use have co-occurred, further focused assessment of mood symptoms before alcohol and drug use or during extended periods of abstinence are necessary to make the diagnosis of bipolar disorder with confidence. Family history of SUDs and/or BD are neither necessary nor sufficient for either diagnosis; however, collateral information from family or significant others could help make the diagnosis and may identify aids and obstacles to treatment planning and engagement.
When a patient’s clinical history strongly supports the diagnoses of BD and co-occurring SUDs, more detailed inquiry is warranted. Determining the patient’s age at onset of each disorder may have prognostic value because onset of mania before SUDs developed, especially in adolescence, may predict a more severe course of both illnesses.22 A complete alcohol use history should include routine questioning about past withdrawal. Previous withdrawal seizures in an actively drinking BD patient may tip the balance toward adding an anticonvulsant for mood stabilization. A thorough SUD history should elicit information about polysubstance abuse or dependence and include screening for injection drug use and other risk factors for HCV and human immunodeficiency virus (HIV), such as hypersexuality during manic or hypomanic episodes. Document the date of the last screening for HCV/ HIV in BD patients at high risk of infection. The U.S. Centers for Disease Control and Prevention recommends that all patients at high risk for HIV consider voluntary screening at least annually.23
Assess your patient’s historical and ongoing alcohol and other drug abuse at the initial visit, and continue to monitor substance use at all subsequent visits, especially in patients with HCV. When feasible, order urine drug screening and laboratory testing for alcohol use biomarkers such as carbohydrate-deficient transferrin and gamma-glutamyltransferase to supplement self-report data, especially in patients with poor insight or low motivation. Assess suicidal ideation and any changes in suicide risk factors at every visit.
Treatment. No biologic therapies have been FDA-approved for treating patients with co-occurring BD and SUDs. Comorbid SUDs in BD patients—as well as rapid cycling and mixed mood episodes, both of which are more common in patients with comorbid SUDs—predict poor response to lithium.17 However, the evidence base for optimal pharmacotherapy remains extremely limited. Published double-blind, placebo-controlled RCTs in persons with BD and co-morbid SUDs are limited to only 1 trial each of lithium, carbamazepine, quetiapine, and naltrexone, and 2 comparisons of lithium plus divalproex vs lithium alone (Table 3).24-29
Salloum et al24 reported that bipolar I disorder patients with alcohol dependence who received divalproex plus lithium as maintenance treatment had fewer heavy drinking days and fewer drinks per heavy drinking day than those receiving lithium plus placebo. However, the addition of divalproex did not improve manic or depressive symptoms, and depression remission rates remained relatively low in both groups. A recent 6-month study comparing lithium vs lithium plus divalproex in patients with SUDs and rapid-cycling BD found no additional benefit of divalproex over lithium monotherapy in retention, mood, or substance use outcomes.28 However, modest evidence that anticonvulsants such as valproic acid and carbamazepine may help treat acute alcohol withdrawal2 could support their preferential use as mood stabilizers over lithium in actively drinking BD patients.
Research underscores the difficulty in keeping dual diagnosis patients in treatment. Salloum et al24 reported that only one-third of randomized subjects completed the 24-week study. In the Kemp study,28 79% of 149 recruited subjects failed to complete the lead-in stabilization phase. Of the 31 remaining subjects who were randomized to lithium or lithium/divalproex combination, only 8 (26% of those randomized, 5% of those recruited) completed the 6-month trial.
Substance abuse is associated with significantly decreased treatment adherence in persons with BD20 and may affect medication choice. For example, caution may be warranted in the use of lamotrigine in substance-abusing patients with poor adherence because re-titration from the starting dose is recommended if the medicine has been missed for a consecutive period exceeding 5 half-lives of the drug.30
Notable progress has been made in developing psychosocial treatments for comorbid SUDs and BD. Integrated group therapy has been designed to address the 2 disorders simultaneously by emphasizing the relationship between the disorders and highlighting similarities in cognitive and behavioral change that promote recovery in both.31 In a recent well-controlled RCT, this approach reduced alcohol and other drug use to approximately one-half the levels observed in those who received only group drug counseling.31
Research suggests that an integrated approach that encompasses psychiatric, medical, psychosocial, and legal dimensions simultaneously may be most effective. For patients such as Mr. M, this would include aggressively treating mood symptoms while employing motivational interviewing techniques to improve engagement in substance dependence treatment. If possible, involving family members, parole officials, housing agencies, and other public assistance workers in the treatment plan may increase treatment adherence and reduce loss of contact during illness exacerbations. Stabilization of substance use and psychiatric morbidity should be accompanied by timely evaluation of HCV and other medical comorbidities in order to improve long-term prognosis.
Table 2
Assessing patients you suspect have comorbid BD and SUDs
Initial assessment |
---|
Thorough substance use history in all patients with known or suspected bipolar disorder:
|
Thorough evaluation of any history of hypomania/mania and depression in all patients with known or suspected SUDs:
|
Assess risk factors, screening status for hepatitis C, HIV |
Obtain collateral information from family and significant others if feasible and appropriate |
Detailed assessment of suicide risk |
Follow-up assessments |
Substance use since last visit by self-report |
Consider UDS, CDT, GGT |
Medication adherence |
Detailed assessment of suicide risk |
BD: bipolar disorder; CDT: carbohydrate-deficient transferrin; GGT: gamma-glutamyltransferase; HIV: human immunodeficiency virus; SUDs: substance use disorders; UDS: urine drug screen |
Table 3
Pharmacotherapy for bipolar disorder and co-occurring SUDs
Study | Diagnoses/N* | Medications | Outcome |
---|---|---|---|
Salloum et al, 200524 | BD I, alcohol dependence. N=59 [20] | Divalproex + lithium vs lithium, 24 weeks | Decreased number of heavy drinking days, fewer drinks per heavy drinking day |
Geller et al, 199825 | BD I, BP II, MDD (adolescents); alcohol, cannabis abuse. N=25 [21] | Lithium, 6 weeks | Decreased cannabis-positive urine drug screen (lithium > placebo) |
Brady et al, 200226 | BD I, BD II, cyclothymia; cocaine dependence. N=57 | Carbamazepine, 12 weeks | Trend toward longer time to cocaine use |
Brown et al, 200827 | BD I, BD II; N=102 | Quetiapine, 12 weeks | Decreased HAM-D scores (quetiapine > placebo) |
Kemp et al, 200928 | BD I, BD II, rapid cycling; alcohol, cannabis, cocaine abuse or dependence. N=31 [8] | Divalproex + lithium vs lithium, 6 months | No group differences |
Brown et al, 200929 | BD I, BD II; alcohol dependence. N=50 [26] | Naltrexone, 12 weeks | Trend toward increased probability of no drinking days |
* N=number of subjects randomized to double-blind treatment. Numbers in brackets indicate the number of subjects who completed all study visits (when reported) | |||
BD: bipolar disorder; HAM-D: 17-item Hamilton Rating Scale for Depression; MDD: major depressive disorder; SUDs: substance use disorders |
Related Resources
- International Society for Bipolar Disorders. www.isbd.org.
- National Institute on Alcohol Abuse and Alcoholism. www.niaaa.nih.gov.
- National Institute on Drug Abuse. www.nida.nih.gov.
Drug Brand Names
- Carbamazepine • Tegretol
- Divalproex/valproic acid • Depakote
- Lamotrigine • Lamictal
- Lithium • Lithobid
- Naltrexone • ReVia
- Quetiapine • Seroquel
Disclosure
Dr. Tolliver receives research grant funding from Forest Laboratories and the National Institute on Alcohol Abuse and Alcoholism. Neither source influenced the content or submission of this manuscript.
A recent analysis of data from >65,000 veterans found bipolar patients with comorbid substance use disorders (SUDs) were 7 times more likely to have hepatitis C virus (HCV) than patients with no serious mental illness.a Matthews and colleagues found that 29.6% of persons diagnosed with bipolar disorder (BD) and SUDs tested positive for HCV—roughly 5 times the relative risk of patients without either diagnosis.b The high prevalence of HCV in patients with comorbid BD and SUDs may be the result of injection drug use, increased risky sexual behavior while manic or intoxicated, or both.
HCV has multiple treatment implications for these patients. Alcohol abuse and dependence are the most common SUDs that co-occur with BD,c-e and patients with HCV who drink alcohol excessively have more severe hepatic fibrosis, accelerated disease progression, and higher rates of cirrhosis and hepatocellular carcinoma than HCV patients who do not drink.f Medications commonly used to treat BD or alcohol dependence may have adverse effects on the liver and require more careful monitoring in the presence of HCV infection. For example, valproic acid has been reported to improve drinking outcomes in alcohol-dependent patients with BDg but has been associated with higher rates of marked hepatic transaminase elevation in patients with HCV infection compared with those without HCV.h Marked elevation of hepatic transaminases may be observed in HCV-infected individuals treated with other medications such as lithium or antidepressants,h and valproic acid use is not an absolute contraindication in HCV patients. Nevertheless, the effects of valproic acid in HCV-infected BD patients who drink alcohol are unknown and therefore cautious and frequent monitoring of hepatic enzymes are warranted in this population.
Finally, both SUDs and BD complicate HCV treatment. In a database review of >113,000 veterans with HCV infection, Butt and colleagues found that individuals with BD accounted for 10.4% of the HCV-infected sample but only 5% of those who received HCV treatment.i Similarly, patients with alcohol abuse or dependence made up 44.3% of the HCV-infected sample but only 28.9% of those who received HCV treatment.
Because the rate of liver biopsy in untreated patients was low, the decision not to treat appeared to be based more often on other criteria. This is not surprising; pegylated interferon-alfa—the most effective treatment for chronic HCV—has been associated with multiple neuropsychiatric symptoms observed in BD, including depression, mania, psychosis, and suicidal ideation.j Emergence of severe psychiatric complications usually results in permanent discontinuation of interferon treatment. Likewise, the presence of alcohol abuse or other SUDs is a strong negative predictor of interferon treatment response and retention and generally has been considered a relative contraindication for interferon initiation.f
References
a. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
b. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
c. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
d. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
e. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
f. Bhattacharya R, Shuhart M. Hepatitis C and alcohol. J Clin Gastroenterol. 2003;36:242-252.
g. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
h. Felker BL, Sloan KL, Dominitz JA, et al. The safety of valproic acid use for patients with hepatitis C infection. Am J Psychiatry. 2003;160:174-178.
i. Butt AA, Justice AC, Skanderson M, et al. Rate and predictors of treatment prescription for hepatitis C. Gut. 2006;56:385-389.
j. Onyike CU, Bonner JO, Lyketsos CG, et al. Mania during treatment of chronic hepatitis C with pegylated interferon and ribavirin. Am J Psychiatry. 2004;161:429-435.
Long recognized to be more prevalent in forensic populations, bipolar disorder (BD) is especially overrepresented among those with repeat arrests and incarcerations. In a recent study of >79,000 inmates incarcerated in Texas in 2006 and 2007, those with BD were 3.3 times more likely to have had ≥4 previous incarcerations.k
Comorbid substance use disorder (SUD) is a significant risk factor for criminal arrest. For example, in a Los Angeles County (CA) sample of inmates with BD, 75.8% had co-occurring SUDs, compared with 18.5% in a comparison group of hospitalized BD patients.l The association of SUDs with arrest is especially high in females with BD. In the Los Angeles sample mentioned above, incarcerated bipolar women were >38 times more likely to have a SUD than a comparison group of female patients treated in the community.m
References
k. Baillargeon J, Binswanger IA, Penn JV, et al. Psychiatric disorders and repeat incarcerations: the revolving prison door. Am J Psychiatry. 2009;166:103-109.
l. Quanbeck CD, Stone DC, Scott CL, et al. Clinical and legal correlates of inmates with bipolar disorder at time of criminal arrest. J Clin Psychiatry. 2004;65:198-203.
m. McDermott BE, Quanbeck CD, Frye MA. Comorbid substance use disorder in women with bipolar disorder is associated with criminal arrest. Bipolar Disord. 2007;9(5):536-540.
1. Levin FR, Hennessey G. Bipolar disorder and substance abuse. Biol Psychiatry. 2004;56:738-748.
2. Frye MA, Salloum IM. Bipolar disorder and comorbid alcoholism: prevalence rate and treatment considerations. Bipolar Disord. 2006;8:677-685.
3. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
4. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
5. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
6. Sublette EM, Carballo JJ, Moreno C, et al. Substance use disorders and suicide attempts in bipolar subtypes. J Psychiatric Res. 2009;43:230-238.
7. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship to historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry. 2001;158:420-426.
8. Frye MA, Altshuler LL, McElroy SL, et al. Gender differences in prevalence, risk, and clinical correlates of alcoholism comorbidity in bipolar disorder. Am J Psychiatry. 2003;160:883-889.
9. Simon NM, Otto MW, Wisniewski SR, et al. Anxiety disorder comorbidity in bipolar disorder patients: data from the first 500 participants in the systematic treatment enhancement program for bipolar disorder (STEP-BD). Am J Psychiatry. 2004;161:2222-2229.
10. Perron BE, Howard MO, Nienhuis JK, et al. Prevalence and burden of general medical conditions among adults with bipolar I disorder: results from the National Epidemiologic Survey on Alcohol and Related Conditions. J Clin Psychiatry. 2009;70:1407-1415.
11. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
12. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
13. Singh JB, Zarate CA. Pharmacological treatment of psychiatric comorbidity in bipolar disorder: a review of controlled trials. Bipolar Disord. 2006;8:696-709.
14. Vornik LA, Brown ES. Management of comorbid bipolar disorder and substance abuse. J Clin Psychiatry. 2006;67(suppl 7):24-30.
15. Strakowski SM, DelBello MP. The co-occurrence of bipolar and substance use disorders. Clin Psych Rev. 2000;20:191-206.
16. Berk M, Dodd S, Callaly P, et al. History of illness prior to a diagnosis of bipolar disorder or schizoaffective disorder. J Affect Disord. 2007;103:181-186.
17. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
18. McGuffin P, Rijsdijk F, Andrew M, et al. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch Gen Psychiatry. 2003;60:497-502.
19. Kendler KS, Prescott CA, Myers J, et al. The structure of genetic and environmental risk factors for common psychiatric and substance abuse disorders in men and women. Arch Gen Psychiatry. 2003;60:929-937.
20. Weiss RD, Greenfield SF, Najavits LM, et al. Medication compliance among patients with bipolar disorder and substance use disorder. J Clin Psychiatry. 1998;59:172-174.
21. Goldberg JF, Garno JL, Callahan AM, et al. Overdiagnosis of bipolar disorder among substance use disorder inpatients with mood instability. J Clin Psychiatry. 2008;69:1751-1757.
22. Winokur G, Coryell W, Akiskal HS, et al. Alcoholism in manic-depressive (bipolar) illness: familial illness, course of illness, and the primary-secondary distinction. Am J Psychiatry. 1995;152:365-372.
23. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing of adults, adolescents, and pregnant women in health care settings. MMWR. 2006;55(RR14):1-17.
24. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
25. Geller B, Cooper TB, Sun K, et al. Double-blind and placebo-controlled study of lithium for adolescent bipolar disorders with secondary substance dependency. J Am Acad Child Adolesc Psychiatry. 1998;37(2):171-178.
26. Brady KT, Sonne SC, Malcolm RJ, et al. Carbamazepine in the treatment of cocaine dependence: subtyping by affective disorder. Exp Clin Psychopharmacol. 2002;10:276-285.
27. Brown ES, Garza M, Carmody TJ. A randomized, double-blind, placebo-controlled add-on trial of quetiapine in outpatients with bipolar disorder and alcohol use disorders. J Clin Psychiatry. 2008;69:701-705.
28. Kemp DE, Gao K, Ganocy S, et al. A 6-month, double-blind, maintenance trial of lithium monotherapy versus the combination of lithium and divalproex for rapid-cycling bipolar disorder and co-occurring substance abuse or dependence. J Clin Psychiatry. 2009;70:113-121.
29. Brown ES, Carmody TJ, Schmitz JM, et al. A randomized, double-blind, placebo-controlled pilot study of naltrexone in outpatients with bipolar disorder and alcohol dependence. Alcohol Clin Exp Res. 2009;3:1863-1869.
30. Lamictal [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2009.
31. Weiss RD, Griffin ML, Kolodziej MR, et al. A randomized trial of integrated group therapy versus group drug counseling for patients with bipolar disorder and substance dependence. Am J Psychiatry. 2007;164:100-107.
After testing positive for cocaine on a recent court-mandated urine drug screen, Mr. M, age 49, is referred by his parole officer for psychiatric and substance abuse treatment. Mr. M has bipolar I disorder and alcohol, cocaine, and opioid dependence. He says he has been hospitalized or incarcerated at least once each year for the past 22 years. Mr. M has seen numerous psychiatrists as an outpatient, but rarely for more than 2 to 3 months and has not taken any psychotropics for more than 5 months.
Approximately 1 week before his recent urine drug screen, Mr. M became euphoric, stopped sleeping for several days, and spent $2,000 on cocaine and “escorts.” He reports that each day he smokes 30 cigarettes and drinks 1 pint of liquor and prefers to use cocaine and opiates by IV injection. Several years ago Mr. M was diagnosed with hepatitis C virus (HCV) but received no further workup or treatment. Although he denies manic or psychotic symptoms, Mr. M is observed speaking to unseen others in the waiting room and has difficulty remaining still during his interview. His chief concern is insomnia, stating, “Doc, I just need something to help me sleep.”
The high prevalence of substance use disorders (SUDs) in persons with bipolar disorder (BD) is well documented.1,2 Up to 60% of bipolar patients develop an SUD at some point in their lives.3-5 Alcohol use disorders are particularly common among BD patients, with a lifetime prevalence of roughly 50%.2-5 Recent epidemiologic data indicate that 38% of persons with bipolar I disorder and 19% of those with bipolar II disorder meet criteria for alcohol dependence.5 Comorbid SUDs in patients with BD are associated with:
- poor treatment compliance
- longer and more frequent mood episodes
- more mixed episodes
- more hospitalizations
- more frequent suicide attempts.1,2
The impact of co-occurring SUDs on suicidality is particularly high among those with bipolar I disorder.6
Frequently referred to as “dual diagnosis” conditions, co-occurring BD and SUDs may be more accurately envisioned as multi-morbid, rather than comorbid, illnesses. Data from the Stanley Foundation Bipolar Network suggest that 42% of BD patients have a lifetime history of ≥2 comorbid axis I disorders.7 Rates of generalized anxiety disorder, panic disorder, and posttraumatic stress disorder are particularly high in BD patients with co-occurring SUDs.8,9 In addition, the presence of 1 SUD may mark the presence of other SUDs; for example, alcohol dependence is strongly associated with polysubstance abuse, especially in females with BD.8 Furthermore, medical comorbidities that impact treatment decisions also are highly prevalent in BD patients with comorbid SUDs.10 In particular, HCV rates are higher in persons with BD compared with the general population,11 and are >5 times as likely in bipolar patients with co-occurring SUDs.12
Unfortunately, limited treatment research guides clinical management of comorbid BD and SUDs.2,13,14 Clinical trials of medications for BD traditionally have excluded patients with SUDs, and persons with serious mental illness usually are ineligible for SUD treatment studies.13 Furthermore, the few randomized controlled trials (RCTs) conducted in persons with both illnesses have been constrained by relatively small sample sizes and low retention rates. In the absence of a definitive consensus for optimal treatment, this article outlines general clinical considerations and an integrated approach to assessing and managing this complex patient population.
Birds of a feather
Multiple hypotheses try to account for the high rate of SUDs in patients with BD (Table 1), but none fully explain the complex interaction observed clinically.14,15 Substance dependence and BD are chronic remitting/relapsing disorders with heterogeneous presentations and highly variable natural histories. As with SUDs, BD may go undiagnosed and untreated for years, coming to clinical attention only after substantial disease progression.16
The fluctuating illness course in BD and SUDs makes diagnosis and treatment difficult. Symptomatic periods often are interrupted by spontaneous remissions and longer—although usually temporary—periods of perceived control. Both BD and SUDs may be associated with profound mood instability, increased impulsivity, altered responsiveness to reward, and impaired executive function.17 Finally, the high degree of heritability in BD18 and many SUDs19 may make treatment engagement more difficult if either disorder is present in multiple family members because of:
- potential for greater clinical severity
- reduced psychosocial resources
- altered familial behavioral norms that may impede the patient’s recognition of illness.
Denial of illness is a critical symptom that may fluctuate with disease course in both disorders. Persons with BD or SUDs may be least likely to recognize that they are ill during periods of highest symptom severity. Accordingly, treatment adherence in patients with either disorder may be limited at baseline and decline further when the 2 illnesses co-occur.20
Involvement in the criminal justice system and medical comorbidities, particularly HCV, also complicate diagnosis and treatment of BD patients with SUDs. For more information about these topics, see Box1 and Box2.
Table 1
Why is substance abuse so prevalent among bipolar patients?
Proposed hypothesis | Selected limitations of this hypothesis |
---|---|
Self-medication: substance abuse occurs as an attempt to regulate mood | High rates of substance use during euthymia; high prevalence of alcohol/depressant use during depressive phase, stimulant use during manic phase |
Common neurobiologic or genetic risk factors | Specific evidence from linkage/association studies currently is lacking |
Substance use occurs as a symptom of bipolar disorder | High percentage of patients with bipolar disorder do not have SUDs; there is a poor correlation of onset, course of bipolar, and SUD symptoms |
Substance use unmasks bipolar disorder or a bipolar diathesis | Emergence of mania before SUD is common and predictive of more severe course of bipolar disorder |
High comorbidity rates are an artifact of misdiagnosis based on overlapping symptoms and poor diagnostic boundaries | Very high prevalence of SUDs also is observed in longitudinal studies of patients initially hospitalized for mania |
SUDs: substance use disorders | |
Source: References 14,15 |
Integrated clinical management
Assessment. Although not intended to be comprehensive, suggestions for routine assessment of patients with suspected SUDs and/or BD are listed in Table 2. Because clinicians may encounter dual diagnosis patients in general psychiatric clinics or specialty (addiction or mood disorder) clinics, it is useful to obtain a thorough substance use history in all patients with known or suspected BD as well as a thorough history of hypomania/mania and depression in all patients with addictive disorders. BD diagnoses by self-report or chart history in patients with SUDs should be considered cautiously because BD often is overdiagnosed in persons engaged in active substance abuse or experiencing withdrawal.21 If past or present mood symptoms and substance use have co-occurred, further focused assessment of mood symptoms before alcohol and drug use or during extended periods of abstinence are necessary to make the diagnosis of bipolar disorder with confidence. Family history of SUDs and/or BD are neither necessary nor sufficient for either diagnosis; however, collateral information from family or significant others could help make the diagnosis and may identify aids and obstacles to treatment planning and engagement.
When a patient’s clinical history strongly supports the diagnoses of BD and co-occurring SUDs, more detailed inquiry is warranted. Determining the patient’s age at onset of each disorder may have prognostic value because onset of mania before SUDs developed, especially in adolescence, may predict a more severe course of both illnesses.22 A complete alcohol use history should include routine questioning about past withdrawal. Previous withdrawal seizures in an actively drinking BD patient may tip the balance toward adding an anticonvulsant for mood stabilization. A thorough SUD history should elicit information about polysubstance abuse or dependence and include screening for injection drug use and other risk factors for HCV and human immunodeficiency virus (HIV), such as hypersexuality during manic or hypomanic episodes. Document the date of the last screening for HCV/ HIV in BD patients at high risk of infection. The U.S. Centers for Disease Control and Prevention recommends that all patients at high risk for HIV consider voluntary screening at least annually.23
Assess your patient’s historical and ongoing alcohol and other drug abuse at the initial visit, and continue to monitor substance use at all subsequent visits, especially in patients with HCV. When feasible, order urine drug screening and laboratory testing for alcohol use biomarkers such as carbohydrate-deficient transferrin and gamma-glutamyltransferase to supplement self-report data, especially in patients with poor insight or low motivation. Assess suicidal ideation and any changes in suicide risk factors at every visit.
Treatment. No biologic therapies have been FDA-approved for treating patients with co-occurring BD and SUDs. Comorbid SUDs in BD patients—as well as rapid cycling and mixed mood episodes, both of which are more common in patients with comorbid SUDs—predict poor response to lithium.17 However, the evidence base for optimal pharmacotherapy remains extremely limited. Published double-blind, placebo-controlled RCTs in persons with BD and co-morbid SUDs are limited to only 1 trial each of lithium, carbamazepine, quetiapine, and naltrexone, and 2 comparisons of lithium plus divalproex vs lithium alone (Table 3).24-29
Salloum et al24 reported that bipolar I disorder patients with alcohol dependence who received divalproex plus lithium as maintenance treatment had fewer heavy drinking days and fewer drinks per heavy drinking day than those receiving lithium plus placebo. However, the addition of divalproex did not improve manic or depressive symptoms, and depression remission rates remained relatively low in both groups. A recent 6-month study comparing lithium vs lithium plus divalproex in patients with SUDs and rapid-cycling BD found no additional benefit of divalproex over lithium monotherapy in retention, mood, or substance use outcomes.28 However, modest evidence that anticonvulsants such as valproic acid and carbamazepine may help treat acute alcohol withdrawal2 could support their preferential use as mood stabilizers over lithium in actively drinking BD patients.
Research underscores the difficulty in keeping dual diagnosis patients in treatment. Salloum et al24 reported that only one-third of randomized subjects completed the 24-week study. In the Kemp study,28 79% of 149 recruited subjects failed to complete the lead-in stabilization phase. Of the 31 remaining subjects who were randomized to lithium or lithium/divalproex combination, only 8 (26% of those randomized, 5% of those recruited) completed the 6-month trial.
Substance abuse is associated with significantly decreased treatment adherence in persons with BD20 and may affect medication choice. For example, caution may be warranted in the use of lamotrigine in substance-abusing patients with poor adherence because re-titration from the starting dose is recommended if the medicine has been missed for a consecutive period exceeding 5 half-lives of the drug.30
Notable progress has been made in developing psychosocial treatments for comorbid SUDs and BD. Integrated group therapy has been designed to address the 2 disorders simultaneously by emphasizing the relationship between the disorders and highlighting similarities in cognitive and behavioral change that promote recovery in both.31 In a recent well-controlled RCT, this approach reduced alcohol and other drug use to approximately one-half the levels observed in those who received only group drug counseling.31
Research suggests that an integrated approach that encompasses psychiatric, medical, psychosocial, and legal dimensions simultaneously may be most effective. For patients such as Mr. M, this would include aggressively treating mood symptoms while employing motivational interviewing techniques to improve engagement in substance dependence treatment. If possible, involving family members, parole officials, housing agencies, and other public assistance workers in the treatment plan may increase treatment adherence and reduce loss of contact during illness exacerbations. Stabilization of substance use and psychiatric morbidity should be accompanied by timely evaluation of HCV and other medical comorbidities in order to improve long-term prognosis.
Table 2
Assessing patients you suspect have comorbid BD and SUDs
Initial assessment |
---|
Thorough substance use history in all patients with known or suspected bipolar disorder:
|
Thorough evaluation of any history of hypomania/mania and depression in all patients with known or suspected SUDs:
|
Assess risk factors, screening status for hepatitis C, HIV |
Obtain collateral information from family and significant others if feasible and appropriate |
Detailed assessment of suicide risk |
Follow-up assessments |
Substance use since last visit by self-report |
Consider UDS, CDT, GGT |
Medication adherence |
Detailed assessment of suicide risk |
BD: bipolar disorder; CDT: carbohydrate-deficient transferrin; GGT: gamma-glutamyltransferase; HIV: human immunodeficiency virus; SUDs: substance use disorders; UDS: urine drug screen |
Table 3
Pharmacotherapy for bipolar disorder and co-occurring SUDs
Study | Diagnoses/N* | Medications | Outcome |
---|---|---|---|
Salloum et al, 200524 | BD I, alcohol dependence. N=59 [20] | Divalproex + lithium vs lithium, 24 weeks | Decreased number of heavy drinking days, fewer drinks per heavy drinking day |
Geller et al, 199825 | BD I, BP II, MDD (adolescents); alcohol, cannabis abuse. N=25 [21] | Lithium, 6 weeks | Decreased cannabis-positive urine drug screen (lithium > placebo) |
Brady et al, 200226 | BD I, BD II, cyclothymia; cocaine dependence. N=57 | Carbamazepine, 12 weeks | Trend toward longer time to cocaine use |
Brown et al, 200827 | BD I, BD II; N=102 | Quetiapine, 12 weeks | Decreased HAM-D scores (quetiapine > placebo) |
Kemp et al, 200928 | BD I, BD II, rapid cycling; alcohol, cannabis, cocaine abuse or dependence. N=31 [8] | Divalproex + lithium vs lithium, 6 months | No group differences |
Brown et al, 200929 | BD I, BD II; alcohol dependence. N=50 [26] | Naltrexone, 12 weeks | Trend toward increased probability of no drinking days |
* N=number of subjects randomized to double-blind treatment. Numbers in brackets indicate the number of subjects who completed all study visits (when reported) | |||
BD: bipolar disorder; HAM-D: 17-item Hamilton Rating Scale for Depression; MDD: major depressive disorder; SUDs: substance use disorders |
Related Resources
- International Society for Bipolar Disorders. www.isbd.org.
- National Institute on Alcohol Abuse and Alcoholism. www.niaaa.nih.gov.
- National Institute on Drug Abuse. www.nida.nih.gov.
Drug Brand Names
- Carbamazepine • Tegretol
- Divalproex/valproic acid • Depakote
- Lamotrigine • Lamictal
- Lithium • Lithobid
- Naltrexone • ReVia
- Quetiapine • Seroquel
Disclosure
Dr. Tolliver receives research grant funding from Forest Laboratories and the National Institute on Alcohol Abuse and Alcoholism. Neither source influenced the content or submission of this manuscript.
A recent analysis of data from >65,000 veterans found bipolar patients with comorbid substance use disorders (SUDs) were 7 times more likely to have hepatitis C virus (HCV) than patients with no serious mental illness.a Matthews and colleagues found that 29.6% of persons diagnosed with bipolar disorder (BD) and SUDs tested positive for HCV—roughly 5 times the relative risk of patients without either diagnosis.b The high prevalence of HCV in patients with comorbid BD and SUDs may be the result of injection drug use, increased risky sexual behavior while manic or intoxicated, or both.
HCV has multiple treatment implications for these patients. Alcohol abuse and dependence are the most common SUDs that co-occur with BD,c-e and patients with HCV who drink alcohol excessively have more severe hepatic fibrosis, accelerated disease progression, and higher rates of cirrhosis and hepatocellular carcinoma than HCV patients who do not drink.f Medications commonly used to treat BD or alcohol dependence may have adverse effects on the liver and require more careful monitoring in the presence of HCV infection. For example, valproic acid has been reported to improve drinking outcomes in alcohol-dependent patients with BDg but has been associated with higher rates of marked hepatic transaminase elevation in patients with HCV infection compared with those without HCV.h Marked elevation of hepatic transaminases may be observed in HCV-infected individuals treated with other medications such as lithium or antidepressants,h and valproic acid use is not an absolute contraindication in HCV patients. Nevertheless, the effects of valproic acid in HCV-infected BD patients who drink alcohol are unknown and therefore cautious and frequent monitoring of hepatic enzymes are warranted in this population.
Finally, both SUDs and BD complicate HCV treatment. In a database review of >113,000 veterans with HCV infection, Butt and colleagues found that individuals with BD accounted for 10.4% of the HCV-infected sample but only 5% of those who received HCV treatment.i Similarly, patients with alcohol abuse or dependence made up 44.3% of the HCV-infected sample but only 28.9% of those who received HCV treatment.
Because the rate of liver biopsy in untreated patients was low, the decision not to treat appeared to be based more often on other criteria. This is not surprising; pegylated interferon-alfa—the most effective treatment for chronic HCV—has been associated with multiple neuropsychiatric symptoms observed in BD, including depression, mania, psychosis, and suicidal ideation.j Emergence of severe psychiatric complications usually results in permanent discontinuation of interferon treatment. Likewise, the presence of alcohol abuse or other SUDs is a strong negative predictor of interferon treatment response and retention and generally has been considered a relative contraindication for interferon initiation.f
References
a. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
b. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
c. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
d. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
e. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
f. Bhattacharya R, Shuhart M. Hepatitis C and alcohol. J Clin Gastroenterol. 2003;36:242-252.
g. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
h. Felker BL, Sloan KL, Dominitz JA, et al. The safety of valproic acid use for patients with hepatitis C infection. Am J Psychiatry. 2003;160:174-178.
i. Butt AA, Justice AC, Skanderson M, et al. Rate and predictors of treatment prescription for hepatitis C. Gut. 2006;56:385-389.
j. Onyike CU, Bonner JO, Lyketsos CG, et al. Mania during treatment of chronic hepatitis C with pegylated interferon and ribavirin. Am J Psychiatry. 2004;161:429-435.
Long recognized to be more prevalent in forensic populations, bipolar disorder (BD) is especially overrepresented among those with repeat arrests and incarcerations. In a recent study of >79,000 inmates incarcerated in Texas in 2006 and 2007, those with BD were 3.3 times more likely to have had ≥4 previous incarcerations.k
Comorbid substance use disorder (SUD) is a significant risk factor for criminal arrest. For example, in a Los Angeles County (CA) sample of inmates with BD, 75.8% had co-occurring SUDs, compared with 18.5% in a comparison group of hospitalized BD patients.l The association of SUDs with arrest is especially high in females with BD. In the Los Angeles sample mentioned above, incarcerated bipolar women were >38 times more likely to have a SUD than a comparison group of female patients treated in the community.m
References
k. Baillargeon J, Binswanger IA, Penn JV, et al. Psychiatric disorders and repeat incarcerations: the revolving prison door. Am J Psychiatry. 2009;166:103-109.
l. Quanbeck CD, Stone DC, Scott CL, et al. Clinical and legal correlates of inmates with bipolar disorder at time of criminal arrest. J Clin Psychiatry. 2004;65:198-203.
m. McDermott BE, Quanbeck CD, Frye MA. Comorbid substance use disorder in women with bipolar disorder is associated with criminal arrest. Bipolar Disord. 2007;9(5):536-540.
After testing positive for cocaine on a recent court-mandated urine drug screen, Mr. M, age 49, is referred by his parole officer for psychiatric and substance abuse treatment. Mr. M has bipolar I disorder and alcohol, cocaine, and opioid dependence. He says he has been hospitalized or incarcerated at least once each year for the past 22 years. Mr. M has seen numerous psychiatrists as an outpatient, but rarely for more than 2 to 3 months and has not taken any psychotropics for more than 5 months.
Approximately 1 week before his recent urine drug screen, Mr. M became euphoric, stopped sleeping for several days, and spent $2,000 on cocaine and “escorts.” He reports that each day he smokes 30 cigarettes and drinks 1 pint of liquor and prefers to use cocaine and opiates by IV injection. Several years ago Mr. M was diagnosed with hepatitis C virus (HCV) but received no further workup or treatment. Although he denies manic or psychotic symptoms, Mr. M is observed speaking to unseen others in the waiting room and has difficulty remaining still during his interview. His chief concern is insomnia, stating, “Doc, I just need something to help me sleep.”
The high prevalence of substance use disorders (SUDs) in persons with bipolar disorder (BD) is well documented.1,2 Up to 60% of bipolar patients develop an SUD at some point in their lives.3-5 Alcohol use disorders are particularly common among BD patients, with a lifetime prevalence of roughly 50%.2-5 Recent epidemiologic data indicate that 38% of persons with bipolar I disorder and 19% of those with bipolar II disorder meet criteria for alcohol dependence.5 Comorbid SUDs in patients with BD are associated with:
- poor treatment compliance
- longer and more frequent mood episodes
- more mixed episodes
- more hospitalizations
- more frequent suicide attempts.1,2
The impact of co-occurring SUDs on suicidality is particularly high among those with bipolar I disorder.6
Frequently referred to as “dual diagnosis” conditions, co-occurring BD and SUDs may be more accurately envisioned as multi-morbid, rather than comorbid, illnesses. Data from the Stanley Foundation Bipolar Network suggest that 42% of BD patients have a lifetime history of ≥2 comorbid axis I disorders.7 Rates of generalized anxiety disorder, panic disorder, and posttraumatic stress disorder are particularly high in BD patients with co-occurring SUDs.8,9 In addition, the presence of 1 SUD may mark the presence of other SUDs; for example, alcohol dependence is strongly associated with polysubstance abuse, especially in females with BD.8 Furthermore, medical comorbidities that impact treatment decisions also are highly prevalent in BD patients with comorbid SUDs.10 In particular, HCV rates are higher in persons with BD compared with the general population,11 and are >5 times as likely in bipolar patients with co-occurring SUDs.12
Unfortunately, limited treatment research guides clinical management of comorbid BD and SUDs.2,13,14 Clinical trials of medications for BD traditionally have excluded patients with SUDs, and persons with serious mental illness usually are ineligible for SUD treatment studies.13 Furthermore, the few randomized controlled trials (RCTs) conducted in persons with both illnesses have been constrained by relatively small sample sizes and low retention rates. In the absence of a definitive consensus for optimal treatment, this article outlines general clinical considerations and an integrated approach to assessing and managing this complex patient population.
Birds of a feather
Multiple hypotheses try to account for the high rate of SUDs in patients with BD (Table 1), but none fully explain the complex interaction observed clinically.14,15 Substance dependence and BD are chronic remitting/relapsing disorders with heterogeneous presentations and highly variable natural histories. As with SUDs, BD may go undiagnosed and untreated for years, coming to clinical attention only after substantial disease progression.16
The fluctuating illness course in BD and SUDs makes diagnosis and treatment difficult. Symptomatic periods often are interrupted by spontaneous remissions and longer—although usually temporary—periods of perceived control. Both BD and SUDs may be associated with profound mood instability, increased impulsivity, altered responsiveness to reward, and impaired executive function.17 Finally, the high degree of heritability in BD18 and many SUDs19 may make treatment engagement more difficult if either disorder is present in multiple family members because of:
- potential for greater clinical severity
- reduced psychosocial resources
- altered familial behavioral norms that may impede the patient’s recognition of illness.
Denial of illness is a critical symptom that may fluctuate with disease course in both disorders. Persons with BD or SUDs may be least likely to recognize that they are ill during periods of highest symptom severity. Accordingly, treatment adherence in patients with either disorder may be limited at baseline and decline further when the 2 illnesses co-occur.20
Involvement in the criminal justice system and medical comorbidities, particularly HCV, also complicate diagnosis and treatment of BD patients with SUDs. For more information about these topics, see Box1 and Box2.
Table 1
Why is substance abuse so prevalent among bipolar patients?
Proposed hypothesis | Selected limitations of this hypothesis |
---|---|
Self-medication: substance abuse occurs as an attempt to regulate mood | High rates of substance use during euthymia; high prevalence of alcohol/depressant use during depressive phase, stimulant use during manic phase |
Common neurobiologic or genetic risk factors | Specific evidence from linkage/association studies currently is lacking |
Substance use occurs as a symptom of bipolar disorder | High percentage of patients with bipolar disorder do not have SUDs; there is a poor correlation of onset, course of bipolar, and SUD symptoms |
Substance use unmasks bipolar disorder or a bipolar diathesis | Emergence of mania before SUD is common and predictive of more severe course of bipolar disorder |
High comorbidity rates are an artifact of misdiagnosis based on overlapping symptoms and poor diagnostic boundaries | Very high prevalence of SUDs also is observed in longitudinal studies of patients initially hospitalized for mania |
SUDs: substance use disorders | |
Source: References 14,15 |
Integrated clinical management
Assessment. Although not intended to be comprehensive, suggestions for routine assessment of patients with suspected SUDs and/or BD are listed in Table 2. Because clinicians may encounter dual diagnosis patients in general psychiatric clinics or specialty (addiction or mood disorder) clinics, it is useful to obtain a thorough substance use history in all patients with known or suspected BD as well as a thorough history of hypomania/mania and depression in all patients with addictive disorders. BD diagnoses by self-report or chart history in patients with SUDs should be considered cautiously because BD often is overdiagnosed in persons engaged in active substance abuse or experiencing withdrawal.21 If past or present mood symptoms and substance use have co-occurred, further focused assessment of mood symptoms before alcohol and drug use or during extended periods of abstinence are necessary to make the diagnosis of bipolar disorder with confidence. Family history of SUDs and/or BD are neither necessary nor sufficient for either diagnosis; however, collateral information from family or significant others could help make the diagnosis and may identify aids and obstacles to treatment planning and engagement.
When a patient’s clinical history strongly supports the diagnoses of BD and co-occurring SUDs, more detailed inquiry is warranted. Determining the patient’s age at onset of each disorder may have prognostic value because onset of mania before SUDs developed, especially in adolescence, may predict a more severe course of both illnesses.22 A complete alcohol use history should include routine questioning about past withdrawal. Previous withdrawal seizures in an actively drinking BD patient may tip the balance toward adding an anticonvulsant for mood stabilization. A thorough SUD history should elicit information about polysubstance abuse or dependence and include screening for injection drug use and other risk factors for HCV and human immunodeficiency virus (HIV), such as hypersexuality during manic or hypomanic episodes. Document the date of the last screening for HCV/ HIV in BD patients at high risk of infection. The U.S. Centers for Disease Control and Prevention recommends that all patients at high risk for HIV consider voluntary screening at least annually.23
Assess your patient’s historical and ongoing alcohol and other drug abuse at the initial visit, and continue to monitor substance use at all subsequent visits, especially in patients with HCV. When feasible, order urine drug screening and laboratory testing for alcohol use biomarkers such as carbohydrate-deficient transferrin and gamma-glutamyltransferase to supplement self-report data, especially in patients with poor insight or low motivation. Assess suicidal ideation and any changes in suicide risk factors at every visit.
Treatment. No biologic therapies have been FDA-approved for treating patients with co-occurring BD and SUDs. Comorbid SUDs in BD patients—as well as rapid cycling and mixed mood episodes, both of which are more common in patients with comorbid SUDs—predict poor response to lithium.17 However, the evidence base for optimal pharmacotherapy remains extremely limited. Published double-blind, placebo-controlled RCTs in persons with BD and co-morbid SUDs are limited to only 1 trial each of lithium, carbamazepine, quetiapine, and naltrexone, and 2 comparisons of lithium plus divalproex vs lithium alone (Table 3).24-29
Salloum et al24 reported that bipolar I disorder patients with alcohol dependence who received divalproex plus lithium as maintenance treatment had fewer heavy drinking days and fewer drinks per heavy drinking day than those receiving lithium plus placebo. However, the addition of divalproex did not improve manic or depressive symptoms, and depression remission rates remained relatively low in both groups. A recent 6-month study comparing lithium vs lithium plus divalproex in patients with SUDs and rapid-cycling BD found no additional benefit of divalproex over lithium monotherapy in retention, mood, or substance use outcomes.28 However, modest evidence that anticonvulsants such as valproic acid and carbamazepine may help treat acute alcohol withdrawal2 could support their preferential use as mood stabilizers over lithium in actively drinking BD patients.
Research underscores the difficulty in keeping dual diagnosis patients in treatment. Salloum et al24 reported that only one-third of randomized subjects completed the 24-week study. In the Kemp study,28 79% of 149 recruited subjects failed to complete the lead-in stabilization phase. Of the 31 remaining subjects who were randomized to lithium or lithium/divalproex combination, only 8 (26% of those randomized, 5% of those recruited) completed the 6-month trial.
Substance abuse is associated with significantly decreased treatment adherence in persons with BD20 and may affect medication choice. For example, caution may be warranted in the use of lamotrigine in substance-abusing patients with poor adherence because re-titration from the starting dose is recommended if the medicine has been missed for a consecutive period exceeding 5 half-lives of the drug.30
Notable progress has been made in developing psychosocial treatments for comorbid SUDs and BD. Integrated group therapy has been designed to address the 2 disorders simultaneously by emphasizing the relationship between the disorders and highlighting similarities in cognitive and behavioral change that promote recovery in both.31 In a recent well-controlled RCT, this approach reduced alcohol and other drug use to approximately one-half the levels observed in those who received only group drug counseling.31
Research suggests that an integrated approach that encompasses psychiatric, medical, psychosocial, and legal dimensions simultaneously may be most effective. For patients such as Mr. M, this would include aggressively treating mood symptoms while employing motivational interviewing techniques to improve engagement in substance dependence treatment. If possible, involving family members, parole officials, housing agencies, and other public assistance workers in the treatment plan may increase treatment adherence and reduce loss of contact during illness exacerbations. Stabilization of substance use and psychiatric morbidity should be accompanied by timely evaluation of HCV and other medical comorbidities in order to improve long-term prognosis.
Table 2
Assessing patients you suspect have comorbid BD and SUDs
Initial assessment |
---|
Thorough substance use history in all patients with known or suspected bipolar disorder:
|
Thorough evaluation of any history of hypomania/mania and depression in all patients with known or suspected SUDs:
|
Assess risk factors, screening status for hepatitis C, HIV |
Obtain collateral information from family and significant others if feasible and appropriate |
Detailed assessment of suicide risk |
Follow-up assessments |
Substance use since last visit by self-report |
Consider UDS, CDT, GGT |
Medication adherence |
Detailed assessment of suicide risk |
BD: bipolar disorder; CDT: carbohydrate-deficient transferrin; GGT: gamma-glutamyltransferase; HIV: human immunodeficiency virus; SUDs: substance use disorders; UDS: urine drug screen |
Table 3
Pharmacotherapy for bipolar disorder and co-occurring SUDs
Study | Diagnoses/N* | Medications | Outcome |
---|---|---|---|
Salloum et al, 200524 | BD I, alcohol dependence. N=59 [20] | Divalproex + lithium vs lithium, 24 weeks | Decreased number of heavy drinking days, fewer drinks per heavy drinking day |
Geller et al, 199825 | BD I, BP II, MDD (adolescents); alcohol, cannabis abuse. N=25 [21] | Lithium, 6 weeks | Decreased cannabis-positive urine drug screen (lithium > placebo) |
Brady et al, 200226 | BD I, BD II, cyclothymia; cocaine dependence. N=57 | Carbamazepine, 12 weeks | Trend toward longer time to cocaine use |
Brown et al, 200827 | BD I, BD II; N=102 | Quetiapine, 12 weeks | Decreased HAM-D scores (quetiapine > placebo) |
Kemp et al, 200928 | BD I, BD II, rapid cycling; alcohol, cannabis, cocaine abuse or dependence. N=31 [8] | Divalproex + lithium vs lithium, 6 months | No group differences |
Brown et al, 200929 | BD I, BD II; alcohol dependence. N=50 [26] | Naltrexone, 12 weeks | Trend toward increased probability of no drinking days |
* N=number of subjects randomized to double-blind treatment. Numbers in brackets indicate the number of subjects who completed all study visits (when reported) | |||
BD: bipolar disorder; HAM-D: 17-item Hamilton Rating Scale for Depression; MDD: major depressive disorder; SUDs: substance use disorders |
Related Resources
- International Society for Bipolar Disorders. www.isbd.org.
- National Institute on Alcohol Abuse and Alcoholism. www.niaaa.nih.gov.
- National Institute on Drug Abuse. www.nida.nih.gov.
Drug Brand Names
- Carbamazepine • Tegretol
- Divalproex/valproic acid • Depakote
- Lamotrigine • Lamictal
- Lithium • Lithobid
- Naltrexone • ReVia
- Quetiapine • Seroquel
Disclosure
Dr. Tolliver receives research grant funding from Forest Laboratories and the National Institute on Alcohol Abuse and Alcoholism. Neither source influenced the content or submission of this manuscript.
A recent analysis of data from >65,000 veterans found bipolar patients with comorbid substance use disorders (SUDs) were 7 times more likely to have hepatitis C virus (HCV) than patients with no serious mental illness.a Matthews and colleagues found that 29.6% of persons diagnosed with bipolar disorder (BD) and SUDs tested positive for HCV—roughly 5 times the relative risk of patients without either diagnosis.b The high prevalence of HCV in patients with comorbid BD and SUDs may be the result of injection drug use, increased risky sexual behavior while manic or intoxicated, or both.
HCV has multiple treatment implications for these patients. Alcohol abuse and dependence are the most common SUDs that co-occur with BD,c-e and patients with HCV who drink alcohol excessively have more severe hepatic fibrosis, accelerated disease progression, and higher rates of cirrhosis and hepatocellular carcinoma than HCV patients who do not drink.f Medications commonly used to treat BD or alcohol dependence may have adverse effects on the liver and require more careful monitoring in the presence of HCV infection. For example, valproic acid has been reported to improve drinking outcomes in alcohol-dependent patients with BDg but has been associated with higher rates of marked hepatic transaminase elevation in patients with HCV infection compared with those without HCV.h Marked elevation of hepatic transaminases may be observed in HCV-infected individuals treated with other medications such as lithium or antidepressants,h and valproic acid use is not an absolute contraindication in HCV patients. Nevertheless, the effects of valproic acid in HCV-infected BD patients who drink alcohol are unknown and therefore cautious and frequent monitoring of hepatic enzymes are warranted in this population.
Finally, both SUDs and BD complicate HCV treatment. In a database review of >113,000 veterans with HCV infection, Butt and colleagues found that individuals with BD accounted for 10.4% of the HCV-infected sample but only 5% of those who received HCV treatment.i Similarly, patients with alcohol abuse or dependence made up 44.3% of the HCV-infected sample but only 28.9% of those who received HCV treatment.
Because the rate of liver biopsy in untreated patients was low, the decision not to treat appeared to be based more often on other criteria. This is not surprising; pegylated interferon-alfa—the most effective treatment for chronic HCV—has been associated with multiple neuropsychiatric symptoms observed in BD, including depression, mania, psychosis, and suicidal ideation.j Emergence of severe psychiatric complications usually results in permanent discontinuation of interferon treatment. Likewise, the presence of alcohol abuse or other SUDs is a strong negative predictor of interferon treatment response and retention and generally has been considered a relative contraindication for interferon initiation.f
References
a. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
b. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
c. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
d. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
e. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
f. Bhattacharya R, Shuhart M. Hepatitis C and alcohol. J Clin Gastroenterol. 2003;36:242-252.
g. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
h. Felker BL, Sloan KL, Dominitz JA, et al. The safety of valproic acid use for patients with hepatitis C infection. Am J Psychiatry. 2003;160:174-178.
i. Butt AA, Justice AC, Skanderson M, et al. Rate and predictors of treatment prescription for hepatitis C. Gut. 2006;56:385-389.
j. Onyike CU, Bonner JO, Lyketsos CG, et al. Mania during treatment of chronic hepatitis C with pegylated interferon and ribavirin. Am J Psychiatry. 2004;161:429-435.
Long recognized to be more prevalent in forensic populations, bipolar disorder (BD) is especially overrepresented among those with repeat arrests and incarcerations. In a recent study of >79,000 inmates incarcerated in Texas in 2006 and 2007, those with BD were 3.3 times more likely to have had ≥4 previous incarcerations.k
Comorbid substance use disorder (SUD) is a significant risk factor for criminal arrest. For example, in a Los Angeles County (CA) sample of inmates with BD, 75.8% had co-occurring SUDs, compared with 18.5% in a comparison group of hospitalized BD patients.l The association of SUDs with arrest is especially high in females with BD. In the Los Angeles sample mentioned above, incarcerated bipolar women were >38 times more likely to have a SUD than a comparison group of female patients treated in the community.m
References
k. Baillargeon J, Binswanger IA, Penn JV, et al. Psychiatric disorders and repeat incarcerations: the revolving prison door. Am J Psychiatry. 2009;166:103-109.
l. Quanbeck CD, Stone DC, Scott CL, et al. Clinical and legal correlates of inmates with bipolar disorder at time of criminal arrest. J Clin Psychiatry. 2004;65:198-203.
m. McDermott BE, Quanbeck CD, Frye MA. Comorbid substance use disorder in women with bipolar disorder is associated with criminal arrest. Bipolar Disord. 2007;9(5):536-540.
1. Levin FR, Hennessey G. Bipolar disorder and substance abuse. Biol Psychiatry. 2004;56:738-748.
2. Frye MA, Salloum IM. Bipolar disorder and comorbid alcoholism: prevalence rate and treatment considerations. Bipolar Disord. 2006;8:677-685.
3. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
4. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
5. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
6. Sublette EM, Carballo JJ, Moreno C, et al. Substance use disorders and suicide attempts in bipolar subtypes. J Psychiatric Res. 2009;43:230-238.
7. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship to historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry. 2001;158:420-426.
8. Frye MA, Altshuler LL, McElroy SL, et al. Gender differences in prevalence, risk, and clinical correlates of alcoholism comorbidity in bipolar disorder. Am J Psychiatry. 2003;160:883-889.
9. Simon NM, Otto MW, Wisniewski SR, et al. Anxiety disorder comorbidity in bipolar disorder patients: data from the first 500 participants in the systematic treatment enhancement program for bipolar disorder (STEP-BD). Am J Psychiatry. 2004;161:2222-2229.
10. Perron BE, Howard MO, Nienhuis JK, et al. Prevalence and burden of general medical conditions among adults with bipolar I disorder: results from the National Epidemiologic Survey on Alcohol and Related Conditions. J Clin Psychiatry. 2009;70:1407-1415.
11. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
12. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
13. Singh JB, Zarate CA. Pharmacological treatment of psychiatric comorbidity in bipolar disorder: a review of controlled trials. Bipolar Disord. 2006;8:696-709.
14. Vornik LA, Brown ES. Management of comorbid bipolar disorder and substance abuse. J Clin Psychiatry. 2006;67(suppl 7):24-30.
15. Strakowski SM, DelBello MP. The co-occurrence of bipolar and substance use disorders. Clin Psych Rev. 2000;20:191-206.
16. Berk M, Dodd S, Callaly P, et al. History of illness prior to a diagnosis of bipolar disorder or schizoaffective disorder. J Affect Disord. 2007;103:181-186.
17. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
18. McGuffin P, Rijsdijk F, Andrew M, et al. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch Gen Psychiatry. 2003;60:497-502.
19. Kendler KS, Prescott CA, Myers J, et al. The structure of genetic and environmental risk factors for common psychiatric and substance abuse disorders in men and women. Arch Gen Psychiatry. 2003;60:929-937.
20. Weiss RD, Greenfield SF, Najavits LM, et al. Medication compliance among patients with bipolar disorder and substance use disorder. J Clin Psychiatry. 1998;59:172-174.
21. Goldberg JF, Garno JL, Callahan AM, et al. Overdiagnosis of bipolar disorder among substance use disorder inpatients with mood instability. J Clin Psychiatry. 2008;69:1751-1757.
22. Winokur G, Coryell W, Akiskal HS, et al. Alcoholism in manic-depressive (bipolar) illness: familial illness, course of illness, and the primary-secondary distinction. Am J Psychiatry. 1995;152:365-372.
23. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing of adults, adolescents, and pregnant women in health care settings. MMWR. 2006;55(RR14):1-17.
24. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
25. Geller B, Cooper TB, Sun K, et al. Double-blind and placebo-controlled study of lithium for adolescent bipolar disorders with secondary substance dependency. J Am Acad Child Adolesc Psychiatry. 1998;37(2):171-178.
26. Brady KT, Sonne SC, Malcolm RJ, et al. Carbamazepine in the treatment of cocaine dependence: subtyping by affective disorder. Exp Clin Psychopharmacol. 2002;10:276-285.
27. Brown ES, Garza M, Carmody TJ. A randomized, double-blind, placebo-controlled add-on trial of quetiapine in outpatients with bipolar disorder and alcohol use disorders. J Clin Psychiatry. 2008;69:701-705.
28. Kemp DE, Gao K, Ganocy S, et al. A 6-month, double-blind, maintenance trial of lithium monotherapy versus the combination of lithium and divalproex for rapid-cycling bipolar disorder and co-occurring substance abuse or dependence. J Clin Psychiatry. 2009;70:113-121.
29. Brown ES, Carmody TJ, Schmitz JM, et al. A randomized, double-blind, placebo-controlled pilot study of naltrexone in outpatients with bipolar disorder and alcohol dependence. Alcohol Clin Exp Res. 2009;3:1863-1869.
30. Lamictal [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2009.
31. Weiss RD, Griffin ML, Kolodziej MR, et al. A randomized trial of integrated group therapy versus group drug counseling for patients with bipolar disorder and substance dependence. Am J Psychiatry. 2007;164:100-107.
1. Levin FR, Hennessey G. Bipolar disorder and substance abuse. Biol Psychiatry. 2004;56:738-748.
2. Frye MA, Salloum IM. Bipolar disorder and comorbid alcoholism: prevalence rate and treatment considerations. Bipolar Disord. 2006;8:677-685.
3. Grant BF, Stinson FS, Dawson DA, et al. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:807-816.
4. Hasin DS, Stinson FS, Ogburn E, et al. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64:830-842.
5. Merikangas KR, Akiskal HS, Angst J, et al. Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2007;64:543-552.
6. Sublette EM, Carballo JJ, Moreno C, et al. Substance use disorders and suicide attempts in bipolar subtypes. J Psychiatric Res. 2009;43:230-238.
7. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship to historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry. 2001;158:420-426.
8. Frye MA, Altshuler LL, McElroy SL, et al. Gender differences in prevalence, risk, and clinical correlates of alcoholism comorbidity in bipolar disorder. Am J Psychiatry. 2003;160:883-889.
9. Simon NM, Otto MW, Wisniewski SR, et al. Anxiety disorder comorbidity in bipolar disorder patients: data from the first 500 participants in the systematic treatment enhancement program for bipolar disorder (STEP-BD). Am J Psychiatry. 2004;161:2222-2229.
10. Perron BE, Howard MO, Nienhuis JK, et al. Prevalence and burden of general medical conditions among adults with bipolar I disorder: results from the National Epidemiologic Survey on Alcohol and Related Conditions. J Clin Psychiatry. 2009;70:1407-1415.
11. Himelhoch S, McCarthy JF, Ganoczy D, et al. Understanding associations between serious mental illness and hepatitis C virus among veterans: a national multivariate analysis. Psychosomatics. 2009;50:30-37.
12. Matthews AM, Huckans MS, Blackwell AD, et al. Hepatitis C testing and infection rates in bipolar patients with and without comorbid substance use disorders. Bipolar Disord. 2008;10:266-270.
13. Singh JB, Zarate CA. Pharmacological treatment of psychiatric comorbidity in bipolar disorder: a review of controlled trials. Bipolar Disord. 2006;8:696-709.
14. Vornik LA, Brown ES. Management of comorbid bipolar disorder and substance abuse. J Clin Psychiatry. 2006;67(suppl 7):24-30.
15. Strakowski SM, DelBello MP. The co-occurrence of bipolar and substance use disorders. Clin Psych Rev. 2000;20:191-206.
16. Berk M, Dodd S, Callaly P, et al. History of illness prior to a diagnosis of bipolar disorder or schizoaffective disorder. J Affect Disord. 2007;103:181-186.
17. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
18. McGuffin P, Rijsdijk F, Andrew M, et al. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch Gen Psychiatry. 2003;60:497-502.
19. Kendler KS, Prescott CA, Myers J, et al. The structure of genetic and environmental risk factors for common psychiatric and substance abuse disorders in men and women. Arch Gen Psychiatry. 2003;60:929-937.
20. Weiss RD, Greenfield SF, Najavits LM, et al. Medication compliance among patients with bipolar disorder and substance use disorder. J Clin Psychiatry. 1998;59:172-174.
21. Goldberg JF, Garno JL, Callahan AM, et al. Overdiagnosis of bipolar disorder among substance use disorder inpatients with mood instability. J Clin Psychiatry. 2008;69:1751-1757.
22. Winokur G, Coryell W, Akiskal HS, et al. Alcoholism in manic-depressive (bipolar) illness: familial illness, course of illness, and the primary-secondary distinction. Am J Psychiatry. 1995;152:365-372.
23. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing of adults, adolescents, and pregnant women in health care settings. MMWR. 2006;55(RR14):1-17.
24. Salloum IM, Cornelius JR, Daley DC, et al. Efficacy of valproate maintenance in patients with bipolar disorder and alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62:37-45.
25. Geller B, Cooper TB, Sun K, et al. Double-blind and placebo-controlled study of lithium for adolescent bipolar disorders with secondary substance dependency. J Am Acad Child Adolesc Psychiatry. 1998;37(2):171-178.
26. Brady KT, Sonne SC, Malcolm RJ, et al. Carbamazepine in the treatment of cocaine dependence: subtyping by affective disorder. Exp Clin Psychopharmacol. 2002;10:276-285.
27. Brown ES, Garza M, Carmody TJ. A randomized, double-blind, placebo-controlled add-on trial of quetiapine in outpatients with bipolar disorder and alcohol use disorders. J Clin Psychiatry. 2008;69:701-705.
28. Kemp DE, Gao K, Ganocy S, et al. A 6-month, double-blind, maintenance trial of lithium monotherapy versus the combination of lithium and divalproex for rapid-cycling bipolar disorder and co-occurring substance abuse or dependence. J Clin Psychiatry. 2009;70:113-121.
29. Brown ES, Carmody TJ, Schmitz JM, et al. A randomized, double-blind, placebo-controlled pilot study of naltrexone in outpatients with bipolar disorder and alcohol dependence. Alcohol Clin Exp Res. 2009;3:1863-1869.
30. Lamictal [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2009.
31. Weiss RD, Griffin ML, Kolodziej MR, et al. A randomized trial of integrated group therapy versus group drug counseling for patients with bipolar disorder and substance dependence. Am J Psychiatry. 2007;164:100-107.