Welcome to Current Psychiatry, a leading source of information, online and in print, for practitioners of psychiatry and its related subspecialties, including addiction psychiatry, child and adolescent psychiatry, and geriatric psychiatry. This Web site contains evidence-based reviews of the prevention, diagnosis, and treatment of mental illness and psychological disorders; case reports; updates on psychopharmacology; news about the specialty of psychiatry; pearls for practice; and other topics of interest and use to this audience.

Theme
medstat_cp
Top Sections
Evidence-Based Reviews
Cases That Test Your Skills
cp

Dear Drupal User: You're seeing this because you're logged in to Drupal, and not redirected to MDedge.com/psychiatry. 

Main menu
CP Main Menu
Explore menu
CP Explore Menu
Proclivity ID
18803001
Unpublish
Negative Keywords
Bipolar depression
Depression
adolescent depression
adolescent major depressive disorder
adolescent schizophrenia
adolescent with major depressive disorder
animals
autism
baby
brexpiprazole
child
child bipolar
child depression
child schizophrenia
children with bipolar disorder
children with depression
children with major depressive disorder
compulsive behaviors
cure
elderly bipolar
elderly depression
elderly major depressive disorder
elderly schizophrenia
elderly with dementia
first break
first episode
gambling
gaming
geriatric depression
geriatric major depressive disorder
geriatric schizophrenia
infant
kid
major depressive disorder
major depressive disorder in adolescents
major depressive disorder in children
parenting
pediatric
pediatric bipolar
pediatric depression
pediatric major depressive disorder
pediatric schizophrenia
pregnancy
pregnant
rexulti
skin care
teen
wine
Negative Keywords Excluded Elements
header[@id='header']
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-current-psychiatry')]
div[contains(@class, 'pane-pub-home-current-psychiatry')]
div[contains(@class, 'pane-pub-topic-current-psychiatry')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Altmetric
DSM Affiliated
Display in offset block
QuickLearn Excluded Topics/Sections
Best Practices
CME
CME Supplements
Education Center
Medical Education Library
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Clinical
Slot System
Top 25
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Publication LayerRX Default ID
798,799
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC

Prevent drug-drug interactions with cholinesterase inhibitors

Article Type
Changed
Tue, 12/11/2018 - 15:07
Display Headline
Prevent drug-drug interactions with cholinesterase inhibitors

Mr. B, age 78, has a long history of well-controlled bipolar disorder and was diagnosed with Alzheimer’s dementia 6 months ago. He is living at home and has been taking donepezil, 10 mg/d, and lamotrigine, 100 mg bid.

This morning Mr. B’s wife calls and reports that he is experiencing sudden difficulty walking, dizziness, and “feeling drunk.” When you ask about Mr. B’s medications, his wife says that her husband’s internist had prescribed itraconazole, 200 mg/d, for onychomycosis, and Mr. B has taken 1 dose. You promptly discontinue the itraconazole, and Mr. B’s symptoms resolve.

Drug-drug interactions (DDIs) in Alzheimer’s disease (AD) patients such as Mr. B can be serious and even life-threatening. On average, persons age ≥65 use 4.5 prescription agents and 2 over-the-counter preparations per day,1 and the number of concurrently used medications is a significant predictor of adverse drug reactions.2

Cognitive enhancers, including acetylcholinesterase inhibitors (AChEIs) and memantine, are the most widely prescribed agents for AD patients. The FDA has approved galantamine and rivastigmine for mild to moderate dementia, memantine for moderate to severe dementia, and donepezil for mild to severe dementia (Table 1).3-5

To help you minimize adverse DDIs in AD patients, this article describes:

  • pharmacokinetic and pharmacodynamic effects of cognitive enhancers used in AD management
  • DDIs with medications commonly prescribed to AD patients
  • how to avoid adverse events related to antipsychotics, antidepressants, and benzodiazepines.

Table 1

Pharmacokinetic features of cognitive enhancers

AgentProtein bindingCYP-450 activityOther features
AChEIs
  Donepezil96%CYP 2D6, 3A4 substrateOnce-daily dosing
  Rivastigmine40%NoneMetabolized by cholinesterases
  Galantamine18%CYP 2D6, 3A4 substrateNicotinic cholinergic receptor modulation
NMDA receptor antagonist
  Memantine45%NoneNo hepatic metabolism
CYP-450: cytochrome P-450; AChEIs: acetylcholinesterase inhibitors; NMDA: N-methyl-D-aspartate
Source: References 3-5

Pharmacologic changes with aging

Pharmacokinetics is the study of the time course of drugs and their metabolites through the body. Pharmacokinetic interactions involve alterations in the plasma concentration of a drug by a second agent.3

Absorption of medications is decreased in the elderly because of reduced intestinal blood flow and motility. Absorption further decreases if patients concomitantly take antacids, high-fiber supplements, or anticholinergic medications.

Distribution. With aging, lean body mass typically decreases and adipose tissue increases. Because most psychotropics are lipid-soluble, their volume of distribution increases with age. This leads to drug accumulation and longer half-lives. On the other hand, water-soluble medications such as lithium distribute in a smaller volume and pose a higher risk of toxicity.

In plasma, drugs circulate freely or bound to proteins—mainly albumin and α1-acid glycoprotein. Aging can cause decreased plasma albumin and increased α1-acid glycoprotein.6 Additionally, malnutrition, diabetes mellitus, and hepatic and renal disease—all more common with advancing age—may cause hypoalbuminemia, which increases the free fraction of drugs bound to albumin.6Table 1 includes information about cognitive enhancers’ protein binding.

When 2 or more highly protein-bound drugs are coadministered, mutual displacement occurs and the free fraction of each drug increases. A recent case report described valproate toxicity with dizziness, ataxia, and falling in a 76-year-old man after aspirin was added to his regimen.7 The mechanism appeared to be mutual displacement from albumin combined with metabolism of valproate inhibited by aspirin.7

Metabolism. Liver size and hepatic blood flow decrease with aging.6 Cytochrome P-450 3A4 pathway activity slows, but the 2D6 pathway is not affected.4 Oxidative metabolism through CYP pathways is slower, but conjugation reactions are not.6Table 23,5,7,8 lists major substrates and inhibitors of CYP enzymes.

Azole antifungals are potent inhibitors of CYP 3A4,4 of which both donepezil and lamotrigine are substrates (Table 2). In Mr. B’s case, lamotrigine and donepezil levels increased because of this pharmacokinetic interaction. Because donepezil also is metabolized by the CYP 2D6 pathway, the increase in concentration is unlikely to modify the drug effect. Mr. B experienced symptoms consistent with lamotrigine toxicity.

Excretion. The age-associated decline in renal clearance related to a diminished glomerular filtration rate leads to decreased excretion of active metabolites and lithium, making older patients more susceptible to lithium toxicity. The magnitude of the decline in renal clearance varies among patients and is exacerbated by concomitant conditions—such as diabetes and hypertension—and medications—such as nonsteroidal anti-inflammatory drugs (NSAIDs).4 Thiazide diuretics, angiotensin-converting enzyme inhibitors, and cyclooxygenase-2 (COX-2) inhibitors such as celecoxib may elevate lithium levels.3

Pharmacokinetics of AChEIs. AChEIs have relatively few pharmacokinetic interactions, although donepezil and galantamine are metabolized through the liver’s CYP 2D6 and 3A4 pathways.

Because rivastigmine does not undergo hepatic metabolism, it is least likely of the cognitive enhancers to have pharmacokinetic interactions with other medications. Rivastigmine did not lead to increased adverse events when administered concomitantly with 22 different classes of medications—including antidiabetics, cardiovascular drugs, gastrointestinal agents, and NSAIDs.9

 

 

Table 2

DDIs in AD patients: CYP-450 substrates and inhibitors*

 CYP 2D6CYP 3A4
Substrates (substances metabolized by enzyme)Second-generation antipsychotics
Citalopram
Donepezil
Duloxetine
Galantamine
Haloperidol
Tricyclic antidepressants
Trazodone
Venlafaxine
Second-generation antipsychotics
Benzodiazepines
Buspirone
Carbamazepine
Donepezil
Galantamine
Haloperidol
Lamotrigine
Mirtazapine
Nefazodone
Sertraline
Tricyclic antidepressants
Trazodone
Zolpidem
InhibitorsBupropion
Cimetidine
Duloxetine
Fluoxetine
Paroxetine
Sertraline
Erythromycin
Fluconazole
Fluvoxamine
Grapefruit juice
Itraconazole
Nefazodone
*All cytochrome P (CYP) 450 enzymes are induced by barbiturates, phenytoin, carbamazepine, and rifampicin. Smoking also induces CYP 1A2.
DDIs: drug-drug interactions; AD: Alzheimer’s disease
Source: References 3,5,7,8
Pharmacodynamics is the study of the time course and intensity of drugs’ pharmacologic effects. Pharmacodynamic interactions involve changes in a drug’s action at a receptor or biologically active site.3 Pharmacodynamic interactions may result from an antagonistic or synergistic mechanism (Table 3).3,5,10 Dopamine neurons degenerate with aging, particularly after age 70, and the number of cholinergic receptors decreases in AD patients. As a result, these patients may become more sensitive to antipsychotics, selective serotonin reuptake inhibitors (SSRIs)—which indirectly reduce dopamine outflow—and medications with anticholinergic effects.4

Memantine, an amantadine derivative and N-methyl-D-aspartate (NMDA) receptor antagonist, is a weak dopaminergic agonist with atropinic effects.11 Because memantine is not metabolized by the CYP-450 pathway, it lacks pharmacokinetic DDIs.12 However, combining memantine with other NMDA antagonists—such as amantadine or dextromethorphan—could cause hallucinations, dizziness, headache, fatigue, and confusion.11 Concurrent use with drugs that lower seizure threshold, such as tricyclic antidepressants, may increase the risk of seizures.

Table 3

Potential drug-drug interactions in AD patients taking cognitive enhancers

InteractionMechanismPotential sequela(e)
AChEIs + anticholinergics↓ Acetylcholine in CNSCognitive worsening, delirium
AChEIs + beta blockersVagal stimulation and sympathetic blockadeBradycardia, syncope
AChEIs + cholinergics↑ Acetylcholine in PNSCholinergic crisis: hypersalivation, abdominal pain, diarrhea
AChEIs + antipsychotics (rare)↑ Acetylcholine/↓ dopamine in striatumParkinsonian syndrome, rigidity
Ginkgo biloba + warfarinAntiplatelet aggregation and anticoagulationGastrointestinal bleeding, hematuria, subcutaneous ecchymosis
AChEIs: acetylcholinesterase inhibitors; PNS: peripheral nervous system
Source: References 3,5,10

DDIs with cognitive enhancers

Anticholinergics. Because anticholinergic drugs can worsen cognitive impairment and cause delirium they are contraindicated in older patients—especially those with AD. Antihistamines, histamine H2 blockers, low-potency first-generation antipsychotics (FGAs), and tricyclic antidepressants are common medications with anticholinergic effects (Table 4).5,13,14

Anticholinergics can counteract AChEIs’ beneficial effect. Concurrent use of anticholinergics and AChEIs is fairly common in clinical practice but is rarely appropriate because of pharmacologic antagonism. In a retrospective study of 836 community-living older adults (age ≥65) with probable dementia, Roe et al13 compared anticholinergic use in 418 who were taking donepezil with 418 matched controls who were not taking donepezil. They found:

  • 33% of those taking donepezil also were receiving anticholinergics, compared with 23% of controls
  • 26% of all patients in the study used multiple anticholinergic medications.
Similarly, a study of pharmacy claims for AChEIs among 557 Medicaid beneficiaries aged ≥50 found that 35% of patients taking AChEIs also received at least 1 anticholinergic drug.14

Antiparkinsonian agents. Interaction of antiparkinsonian medications with AChEIs could limit the efficacy of either drug when treating comorbid AD and Parkinson’s disease (PD),5 although in practice, clinical deterioration of parkinsonism has not been reported.15 In one study, 25 PD patients stabilized on levodopa/carbidopa were given donepezil, 5 mg/d, or placebo for two 2-week courses separated by a washout of at least 2 weeks. At steady state, pharmacokinetic parameters were unchanged and no clinically significant DDIs were observed.16

Cardiovascular agents. Concurrent use of AChEIs and beta blockers, calcium channel inhibitors, or digoxin could worsen bradycardia and cause syncope. The risk is higher in patients:

  • with sick sinus syndrome or other bradyarrhythmias
  • taking antipsychotics that could induce torsades de pointes,11 such as ziprasidone or haloperidol.
In patients taking these cardiovascular drugs, make sure that heart rate is >60 bpm before AChEI treatment, and monitor regularly.

Other agents. AChEIs inhibit the metabolism of succinylcholine and therefore augment and prolong this drug’s neuromuscular blockade. Discontinue AChEIs before administering succinylcholine for anesthesia, such as for electroconvulsive treatment.

AChEIs may lead to toxicity when added to cholinergic agents such as bethanechol.11 Similarly, AChEIs may precipitate a cholinergic crisis—with increasing weakness, hypersalivation, abdominal pains, and diarrhea—when used in conjunction with peripheral acetylcholinesterase inhibitors such as the myasthenia gravis agents pyridostigmine and neostigmine. The mechanism is increased acetylcholine available at the neuromuscular junction.

Table 4

Medications with moderate to strong anticholinergic activity

ClassExamples
AntiarrhythmicsDisopyramide
AntiemeticsMeclizine
AntiparkinsoniansBenztropine, biperiden, trihexyphenidyl
AntipsychoticsChlorpromazine, clozapine, olanzapine, pimozide, thioridazine
AntihistaminesChlorpheniramine, cyproheptadine, diphenhydramine, hydroxyzine, Promethazine
Gastrointestinal/urinary antispasmodicsAtropine, belladonna alkaloids, dicyclomine, hyoscyamine, oxybutynin, scopolamine, tolterodine
H2 histamineCimetidine, ranitidine
Muscle relaxantsCyclobenzaprine
Tricyclic antidepressantsAmitriptyline, amoxapine, clomipramine, doxepin, imipramine, protriptyline
Source: References 5,13,14

DDIs with other psychotropics

 

 

Antipsychotics. Nearly one-half of AD patients experience delusions, often in the middle stage of the disease, and many are prescribed second-generation antipsychotics (SGAs) to control delusions, hallucinations, sundowning, agitation, or aggression. Concomitant use of AChEIs and antipsychotics may increase the risk of extrapyramidal symptoms by disrupting the acetylcholine/dopamine balance in the striatum.5

In AD patients taking donepezil and risperidone, case reports describe parkinsonian syndrome and rigidity with immobility, which resolved after the antipsychotic was discontinued.5,11 When rivastigmine and risperidone were coadministered, however, no clinically relevant adverse interactions were noted in a 20-week, open-label trial of 65 patients with AD, 10 with vascular dementia, and 10 with both.17

The FDA has warned of increased risk of death when SGAs are used to treat behavioral disturbances in dementia patients. In a recent meta-analysis of 15 placebo-controlled trials, cognitive tests scores worsened when AD patients took aripiprazole, olanzapine, quetiapine, or risperidone. A significant risk for cerebrovascular events was seen, especially with risperidone, although no clear causal relationship was established.18 Falls, injury, and syncope were not increased, and patients with less severe dementia, outpatients, and those selected for psychosis were less affected. Thus, provide careful follow-up and avoid long-term unwarranted antipsychotic use in AD patients.

Highly anticholinergic FGAs such as chlorpromazine are not recommended for AD patients (Table 4).

Antidepressants. Up to 30% of AD patients experience major depression.19 SSRIs are the antidepressants most often used to treat depression and anxiety in AD patients.

Citalopram, escitalopram, or venlafaxine are good choices for patients with AD because of minimal CYP inhibitory activity.4 Fluvoxamine, fluoxetine, and paroxetine inhibit CYP 2C9, through which warfarin and some other drugs with a narrow therapeutic index are metabolized.6

Benzodiazepines are contraindicated in elderly patients (especially those with AD) because of the high risk of delirium, worsened cognitive function, paradoxical disinhibition, and falls.20 If benzodiazepines are necessary to control anxiety, use intermediate-duration agents that do not undergo oxidative metabolism and have no active metabolites, such as lorazepam, oxazepam, or temazepam.19 See Table 2 for more information on benzodiazepine DDIs.

Herbal supplements. Ginkgo biloba and huperzine A (Chinese club moss) are the herbal supplements used most commonly by dementia patients. Ginkgo inhibits platelet aggregation and can cause bleeding complications, with or without concomitant antiplatelet or anticoagulant therapy such as aspirin, warfarin, and NSAIDs. Enzyme induction of CYP 2C19 by ginkgo, leading to subtherapeutic levels of anticonvulsants, has been implicated in a report of fatal seizures. Huperzine A is a natural cholinesterase inhibitor and should not be combined with AChEIs because of the risk of additive adverse effects.10

Related resources

  • Jacobson SA, Pies RW, Greenblatt DJ. Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002.
  • Sandson, NB. Drug-drug interaction primer. Washington, DC: American Psychiatric Publishing; 2007.
Drug brand names

  • Amantadine • Symmetrel
  • Amitriptyline • Elavil
  • Amoxapine • Asendin
  • Aripiprazole • Abilify
  • Atropine • Sal-Tropine
  • Benztropine • Cogentin
  • Bethanechol • Urecholine
  • Biperiden • Akineton
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Tegretol
  • Celecoxib • Celebrex
  • Chlorpheniramine • Chlor-Trimeton
  • Chlorpromazine • Thorazine
  • Cimetidine • Tagamet
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Cyclobenzaprine • Flexeril
  • Cyproheptadine • Periactin
  • Dextromethorphan • Benylin, Delsym, others
  • Dicyclomine • Bentyl
  • Digoxin • Lanoxin
  • Diphenhydramine • Benadryl
  • Disopyramide • Norpace
  • Donepezil • Aricept
  • Doxepin • Adapin, Sinequan
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Erythromycin • E-Mycin
  • Fluconazole • Diflucan
  • Fluvoxamine • Luvox
  • Fluoxetine • Prozac
  • Galantamine • Reminyl, Razadyne
  • Haloperidol • Haldol
  • Hydroxyzine • Vistaril
  • Hyoscyamine • Anaspaz, Levbid, Levsin
  • Imipramine • Tofranil
  • Itraconazole • Sporanox
  • Lamotrigine • Lamictal
  • Levodopa/carbidopa • Sinemet
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Meclizine • Antivert
  • Memantine • Namenda
  • Mirtazapine • Remeron
  • Nefazodone • Serzone
  • Neostigmine • Prostigmin
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Oxybutynin • Ditropan
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Promethazine • Phenergan
  • Protriptyline • Vivactil
  • Pyridostigmine • Mestinon
  • Quetiapine • Seroquel
  • Ranitidine • Zantac
  • Risperidone • Risperdal
  • Rivastigmine • Exelon
  • Scopolamine • Scopace
  • Sertraline • Zoloft
  • Succinylcholine • Anectine
  • Temazepam • Restoril
  • Thioridazine • Mellaril
  • Tolterodine • Detrol
  • Trazodone • Desyrel
  • Trihexyphenidyl • Artane
  • Valproate • Depakote
  • Venlafaxine • Effexor
  • Warfarin • Coumadin
  • Ziprasidone • Geodon
  • Zolpidem • Ambien
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Prescription drugs and the elderly: many still receive potentially harmful drugs despite recent improvements. Washington, DC: United States General Accounting Office; 1996. Publication HEHS 95-152.

2. Atkin PA, Veitch PC, Veitch EM, Ogle SJ. The epidemiology of serious adverse drug reactions among the elderly. Drugs Aging 1999;14:141-52.

3. Marangell LB, Martinez JM, Silver JM, Yudofsky SC, eds. Concise guide to psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:4-7, 129,173, 171-80.

4. Roose SP, Pollock BG, Devanand DD. Treatment during late life. In: Schatzberg AF, Nemeroff CB, eds. Textbook of psychopharmacology. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:1083-5.

5. Bentue-Ferrer D, Tribut O, Polard E, Allain H. Clinically significant drug interactions with cholinesterase inhibitors: a guide for neurologists. CNS Drugs 2003;17:947-63.

6. Mulsant BH, Pollock BG. Psychopharmacology. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:387-411.

7. Sandson NB, Marcucci C, Bourke DL, Smith-Lamacchia R. An interaction between aspirin and valproate: the relevance of plasma protein displacement drug-drug interaction. Am J Psychiatry 2006;163:1891-6.

8. Spina E, Scordo MG, D’Arrigo C. Metabolic drug interactions with new psychotropic agents. Fundam Clin Pharmacol 2003;17:517-38.

9. Grossberg GT, Stahelin HB, Messina JC, et al. Lack of adverse pharmacodynamic drug interactions with rivastigmine and twenty-two classes of medications. Int J Geriatr Psychiatry 2000;15:242-7.

10. Beier MT. Harmless herbs? Think again: merits of a complete medication history. J Am Med Dir Assoc 2006;7:446-7.

11. [No authors listed]. Alzheimer’s disease: beware of interactions with cholinesterase inhibitors. Prescrire Int 2006;15:103-6.

12. Grossberg GT, Edwards KR, Zhao Q. Rationale for combining therapy with galantamine and memantine in Alzheimer’s disease. J Clin Pharmacol 2006;46(suppl 1):S17-S26.

13. Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medications by older adults with dementia. J Am Geriatr Soc 2002;50:836-42.

14. Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The concurrent use of anticholinergics and cholinesterase inhibitors: rare event or common practice? J Am Geriatr Soc 2004;52:2082-7.

15. Schrag A. Psychiatric aspects of Parkinson’s disease. J Neurol 2004;251:795-804.

16. Okereke CS, Kirby L, Kumar D, et al. Concurrent administration of donepezil HCl and levodopa/carbidopa in patients with Parkinson’s disease: assessment of pharmacokinetic changes and safety following multiple oral doses. Br J Clin Pharmacol 2004;58(suppl 1):41-9.

17. Weiser M, Rotmensch HH, Korczyn AD, et al. A pilot, randomized, open-label trial assessing safety and pharmacokinetic parameters of co-administration of rivastigmine with risperidone in dementia patients with behavioral disturbances. Int J Geriatr Psychiatry 2002;17:343-6.

18. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse events of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry 2006;14(3):191-210.

19. Koenig HG, Blazer DG. Mood disorders. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:254.

20. Jacobson SA, Pies RW, Greenblatt DJ. Anxiolytic and sedative-hypnotic medications. In: Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:249-312.

Article PDF
Author and Disclosure Information

Andreea L. Seritan, MD
Assistant clinical professor, Department of psychiatry and behavioral sciences, University of California, Davis

Issue
Current Psychiatry - 07(02)
Publications
Page Number
57-66
Legacy Keywords
Andreea L. Seritan MD; drug-drug interactions; cholinesterase inhibitors; Alzheimer’s dementia; Alzheimer’s disease; acetylcholinesterase inhibitors; AChEIs; memantine; galantamine; rivastigmine; donepezil; dementia
Sections
Author and Disclosure Information

Andreea L. Seritan, MD
Assistant clinical professor, Department of psychiatry and behavioral sciences, University of California, Davis

Author and Disclosure Information

Andreea L. Seritan, MD
Assistant clinical professor, Department of psychiatry and behavioral sciences, University of California, Davis

Article PDF
Article PDF

Mr. B, age 78, has a long history of well-controlled bipolar disorder and was diagnosed with Alzheimer’s dementia 6 months ago. He is living at home and has been taking donepezil, 10 mg/d, and lamotrigine, 100 mg bid.

This morning Mr. B’s wife calls and reports that he is experiencing sudden difficulty walking, dizziness, and “feeling drunk.” When you ask about Mr. B’s medications, his wife says that her husband’s internist had prescribed itraconazole, 200 mg/d, for onychomycosis, and Mr. B has taken 1 dose. You promptly discontinue the itraconazole, and Mr. B’s symptoms resolve.

Drug-drug interactions (DDIs) in Alzheimer’s disease (AD) patients such as Mr. B can be serious and even life-threatening. On average, persons age ≥65 use 4.5 prescription agents and 2 over-the-counter preparations per day,1 and the number of concurrently used medications is a significant predictor of adverse drug reactions.2

Cognitive enhancers, including acetylcholinesterase inhibitors (AChEIs) and memantine, are the most widely prescribed agents for AD patients. The FDA has approved galantamine and rivastigmine for mild to moderate dementia, memantine for moderate to severe dementia, and donepezil for mild to severe dementia (Table 1).3-5

To help you minimize adverse DDIs in AD patients, this article describes:

  • pharmacokinetic and pharmacodynamic effects of cognitive enhancers used in AD management
  • DDIs with medications commonly prescribed to AD patients
  • how to avoid adverse events related to antipsychotics, antidepressants, and benzodiazepines.

Table 1

Pharmacokinetic features of cognitive enhancers

AgentProtein bindingCYP-450 activityOther features
AChEIs
  Donepezil96%CYP 2D6, 3A4 substrateOnce-daily dosing
  Rivastigmine40%NoneMetabolized by cholinesterases
  Galantamine18%CYP 2D6, 3A4 substrateNicotinic cholinergic receptor modulation
NMDA receptor antagonist
  Memantine45%NoneNo hepatic metabolism
CYP-450: cytochrome P-450; AChEIs: acetylcholinesterase inhibitors; NMDA: N-methyl-D-aspartate
Source: References 3-5

Pharmacologic changes with aging

Pharmacokinetics is the study of the time course of drugs and their metabolites through the body. Pharmacokinetic interactions involve alterations in the plasma concentration of a drug by a second agent.3

Absorption of medications is decreased in the elderly because of reduced intestinal blood flow and motility. Absorption further decreases if patients concomitantly take antacids, high-fiber supplements, or anticholinergic medications.

Distribution. With aging, lean body mass typically decreases and adipose tissue increases. Because most psychotropics are lipid-soluble, their volume of distribution increases with age. This leads to drug accumulation and longer half-lives. On the other hand, water-soluble medications such as lithium distribute in a smaller volume and pose a higher risk of toxicity.

In plasma, drugs circulate freely or bound to proteins—mainly albumin and α1-acid glycoprotein. Aging can cause decreased plasma albumin and increased α1-acid glycoprotein.6 Additionally, malnutrition, diabetes mellitus, and hepatic and renal disease—all more common with advancing age—may cause hypoalbuminemia, which increases the free fraction of drugs bound to albumin.6Table 1 includes information about cognitive enhancers’ protein binding.

When 2 or more highly protein-bound drugs are coadministered, mutual displacement occurs and the free fraction of each drug increases. A recent case report described valproate toxicity with dizziness, ataxia, and falling in a 76-year-old man after aspirin was added to his regimen.7 The mechanism appeared to be mutual displacement from albumin combined with metabolism of valproate inhibited by aspirin.7

Metabolism. Liver size and hepatic blood flow decrease with aging.6 Cytochrome P-450 3A4 pathway activity slows, but the 2D6 pathway is not affected.4 Oxidative metabolism through CYP pathways is slower, but conjugation reactions are not.6Table 23,5,7,8 lists major substrates and inhibitors of CYP enzymes.

Azole antifungals are potent inhibitors of CYP 3A4,4 of which both donepezil and lamotrigine are substrates (Table 2). In Mr. B’s case, lamotrigine and donepezil levels increased because of this pharmacokinetic interaction. Because donepezil also is metabolized by the CYP 2D6 pathway, the increase in concentration is unlikely to modify the drug effect. Mr. B experienced symptoms consistent with lamotrigine toxicity.

Excretion. The age-associated decline in renal clearance related to a diminished glomerular filtration rate leads to decreased excretion of active metabolites and lithium, making older patients more susceptible to lithium toxicity. The magnitude of the decline in renal clearance varies among patients and is exacerbated by concomitant conditions—such as diabetes and hypertension—and medications—such as nonsteroidal anti-inflammatory drugs (NSAIDs).4 Thiazide diuretics, angiotensin-converting enzyme inhibitors, and cyclooxygenase-2 (COX-2) inhibitors such as celecoxib may elevate lithium levels.3

Pharmacokinetics of AChEIs. AChEIs have relatively few pharmacokinetic interactions, although donepezil and galantamine are metabolized through the liver’s CYP 2D6 and 3A4 pathways.

Because rivastigmine does not undergo hepatic metabolism, it is least likely of the cognitive enhancers to have pharmacokinetic interactions with other medications. Rivastigmine did not lead to increased adverse events when administered concomitantly with 22 different classes of medications—including antidiabetics, cardiovascular drugs, gastrointestinal agents, and NSAIDs.9

 

 

Table 2

DDIs in AD patients: CYP-450 substrates and inhibitors*

 CYP 2D6CYP 3A4
Substrates (substances metabolized by enzyme)Second-generation antipsychotics
Citalopram
Donepezil
Duloxetine
Galantamine
Haloperidol
Tricyclic antidepressants
Trazodone
Venlafaxine
Second-generation antipsychotics
Benzodiazepines
Buspirone
Carbamazepine
Donepezil
Galantamine
Haloperidol
Lamotrigine
Mirtazapine
Nefazodone
Sertraline
Tricyclic antidepressants
Trazodone
Zolpidem
InhibitorsBupropion
Cimetidine
Duloxetine
Fluoxetine
Paroxetine
Sertraline
Erythromycin
Fluconazole
Fluvoxamine
Grapefruit juice
Itraconazole
Nefazodone
*All cytochrome P (CYP) 450 enzymes are induced by barbiturates, phenytoin, carbamazepine, and rifampicin. Smoking also induces CYP 1A2.
DDIs: drug-drug interactions; AD: Alzheimer’s disease
Source: References 3,5,7,8
Pharmacodynamics is the study of the time course and intensity of drugs’ pharmacologic effects. Pharmacodynamic interactions involve changes in a drug’s action at a receptor or biologically active site.3 Pharmacodynamic interactions may result from an antagonistic or synergistic mechanism (Table 3).3,5,10 Dopamine neurons degenerate with aging, particularly after age 70, and the number of cholinergic receptors decreases in AD patients. As a result, these patients may become more sensitive to antipsychotics, selective serotonin reuptake inhibitors (SSRIs)—which indirectly reduce dopamine outflow—and medications with anticholinergic effects.4

Memantine, an amantadine derivative and N-methyl-D-aspartate (NMDA) receptor antagonist, is a weak dopaminergic agonist with atropinic effects.11 Because memantine is not metabolized by the CYP-450 pathway, it lacks pharmacokinetic DDIs.12 However, combining memantine with other NMDA antagonists—such as amantadine or dextromethorphan—could cause hallucinations, dizziness, headache, fatigue, and confusion.11 Concurrent use with drugs that lower seizure threshold, such as tricyclic antidepressants, may increase the risk of seizures.

Table 3

Potential drug-drug interactions in AD patients taking cognitive enhancers

InteractionMechanismPotential sequela(e)
AChEIs + anticholinergics↓ Acetylcholine in CNSCognitive worsening, delirium
AChEIs + beta blockersVagal stimulation and sympathetic blockadeBradycardia, syncope
AChEIs + cholinergics↑ Acetylcholine in PNSCholinergic crisis: hypersalivation, abdominal pain, diarrhea
AChEIs + antipsychotics (rare)↑ Acetylcholine/↓ dopamine in striatumParkinsonian syndrome, rigidity
Ginkgo biloba + warfarinAntiplatelet aggregation and anticoagulationGastrointestinal bleeding, hematuria, subcutaneous ecchymosis
AChEIs: acetylcholinesterase inhibitors; PNS: peripheral nervous system
Source: References 3,5,10

DDIs with cognitive enhancers

Anticholinergics. Because anticholinergic drugs can worsen cognitive impairment and cause delirium they are contraindicated in older patients—especially those with AD. Antihistamines, histamine H2 blockers, low-potency first-generation antipsychotics (FGAs), and tricyclic antidepressants are common medications with anticholinergic effects (Table 4).5,13,14

Anticholinergics can counteract AChEIs’ beneficial effect. Concurrent use of anticholinergics and AChEIs is fairly common in clinical practice but is rarely appropriate because of pharmacologic antagonism. In a retrospective study of 836 community-living older adults (age ≥65) with probable dementia, Roe et al13 compared anticholinergic use in 418 who were taking donepezil with 418 matched controls who were not taking donepezil. They found:

  • 33% of those taking donepezil also were receiving anticholinergics, compared with 23% of controls
  • 26% of all patients in the study used multiple anticholinergic medications.
Similarly, a study of pharmacy claims for AChEIs among 557 Medicaid beneficiaries aged ≥50 found that 35% of patients taking AChEIs also received at least 1 anticholinergic drug.14

Antiparkinsonian agents. Interaction of antiparkinsonian medications with AChEIs could limit the efficacy of either drug when treating comorbid AD and Parkinson’s disease (PD),5 although in practice, clinical deterioration of parkinsonism has not been reported.15 In one study, 25 PD patients stabilized on levodopa/carbidopa were given donepezil, 5 mg/d, or placebo for two 2-week courses separated by a washout of at least 2 weeks. At steady state, pharmacokinetic parameters were unchanged and no clinically significant DDIs were observed.16

Cardiovascular agents. Concurrent use of AChEIs and beta blockers, calcium channel inhibitors, or digoxin could worsen bradycardia and cause syncope. The risk is higher in patients:

  • with sick sinus syndrome or other bradyarrhythmias
  • taking antipsychotics that could induce torsades de pointes,11 such as ziprasidone or haloperidol.
In patients taking these cardiovascular drugs, make sure that heart rate is >60 bpm before AChEI treatment, and monitor regularly.

Other agents. AChEIs inhibit the metabolism of succinylcholine and therefore augment and prolong this drug’s neuromuscular blockade. Discontinue AChEIs before administering succinylcholine for anesthesia, such as for electroconvulsive treatment.

AChEIs may lead to toxicity when added to cholinergic agents such as bethanechol.11 Similarly, AChEIs may precipitate a cholinergic crisis—with increasing weakness, hypersalivation, abdominal pains, and diarrhea—when used in conjunction with peripheral acetylcholinesterase inhibitors such as the myasthenia gravis agents pyridostigmine and neostigmine. The mechanism is increased acetylcholine available at the neuromuscular junction.

Table 4

Medications with moderate to strong anticholinergic activity

ClassExamples
AntiarrhythmicsDisopyramide
AntiemeticsMeclizine
AntiparkinsoniansBenztropine, biperiden, trihexyphenidyl
AntipsychoticsChlorpromazine, clozapine, olanzapine, pimozide, thioridazine
AntihistaminesChlorpheniramine, cyproheptadine, diphenhydramine, hydroxyzine, Promethazine
Gastrointestinal/urinary antispasmodicsAtropine, belladonna alkaloids, dicyclomine, hyoscyamine, oxybutynin, scopolamine, tolterodine
H2 histamineCimetidine, ranitidine
Muscle relaxantsCyclobenzaprine
Tricyclic antidepressantsAmitriptyline, amoxapine, clomipramine, doxepin, imipramine, protriptyline
Source: References 5,13,14

DDIs with other psychotropics

 

 

Antipsychotics. Nearly one-half of AD patients experience delusions, often in the middle stage of the disease, and many are prescribed second-generation antipsychotics (SGAs) to control delusions, hallucinations, sundowning, agitation, or aggression. Concomitant use of AChEIs and antipsychotics may increase the risk of extrapyramidal symptoms by disrupting the acetylcholine/dopamine balance in the striatum.5

In AD patients taking donepezil and risperidone, case reports describe parkinsonian syndrome and rigidity with immobility, which resolved after the antipsychotic was discontinued.5,11 When rivastigmine and risperidone were coadministered, however, no clinically relevant adverse interactions were noted in a 20-week, open-label trial of 65 patients with AD, 10 with vascular dementia, and 10 with both.17

The FDA has warned of increased risk of death when SGAs are used to treat behavioral disturbances in dementia patients. In a recent meta-analysis of 15 placebo-controlled trials, cognitive tests scores worsened when AD patients took aripiprazole, olanzapine, quetiapine, or risperidone. A significant risk for cerebrovascular events was seen, especially with risperidone, although no clear causal relationship was established.18 Falls, injury, and syncope were not increased, and patients with less severe dementia, outpatients, and those selected for psychosis were less affected. Thus, provide careful follow-up and avoid long-term unwarranted antipsychotic use in AD patients.

Highly anticholinergic FGAs such as chlorpromazine are not recommended for AD patients (Table 4).

Antidepressants. Up to 30% of AD patients experience major depression.19 SSRIs are the antidepressants most often used to treat depression and anxiety in AD patients.

Citalopram, escitalopram, or venlafaxine are good choices for patients with AD because of minimal CYP inhibitory activity.4 Fluvoxamine, fluoxetine, and paroxetine inhibit CYP 2C9, through which warfarin and some other drugs with a narrow therapeutic index are metabolized.6

Benzodiazepines are contraindicated in elderly patients (especially those with AD) because of the high risk of delirium, worsened cognitive function, paradoxical disinhibition, and falls.20 If benzodiazepines are necessary to control anxiety, use intermediate-duration agents that do not undergo oxidative metabolism and have no active metabolites, such as lorazepam, oxazepam, or temazepam.19 See Table 2 for more information on benzodiazepine DDIs.

Herbal supplements. Ginkgo biloba and huperzine A (Chinese club moss) are the herbal supplements used most commonly by dementia patients. Ginkgo inhibits platelet aggregation and can cause bleeding complications, with or without concomitant antiplatelet or anticoagulant therapy such as aspirin, warfarin, and NSAIDs. Enzyme induction of CYP 2C19 by ginkgo, leading to subtherapeutic levels of anticonvulsants, has been implicated in a report of fatal seizures. Huperzine A is a natural cholinesterase inhibitor and should not be combined with AChEIs because of the risk of additive adverse effects.10

Related resources

  • Jacobson SA, Pies RW, Greenblatt DJ. Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002.
  • Sandson, NB. Drug-drug interaction primer. Washington, DC: American Psychiatric Publishing; 2007.
Drug brand names

  • Amantadine • Symmetrel
  • Amitriptyline • Elavil
  • Amoxapine • Asendin
  • Aripiprazole • Abilify
  • Atropine • Sal-Tropine
  • Benztropine • Cogentin
  • Bethanechol • Urecholine
  • Biperiden • Akineton
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Tegretol
  • Celecoxib • Celebrex
  • Chlorpheniramine • Chlor-Trimeton
  • Chlorpromazine • Thorazine
  • Cimetidine • Tagamet
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Cyclobenzaprine • Flexeril
  • Cyproheptadine • Periactin
  • Dextromethorphan • Benylin, Delsym, others
  • Dicyclomine • Bentyl
  • Digoxin • Lanoxin
  • Diphenhydramine • Benadryl
  • Disopyramide • Norpace
  • Donepezil • Aricept
  • Doxepin • Adapin, Sinequan
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Erythromycin • E-Mycin
  • Fluconazole • Diflucan
  • Fluvoxamine • Luvox
  • Fluoxetine • Prozac
  • Galantamine • Reminyl, Razadyne
  • Haloperidol • Haldol
  • Hydroxyzine • Vistaril
  • Hyoscyamine • Anaspaz, Levbid, Levsin
  • Imipramine • Tofranil
  • Itraconazole • Sporanox
  • Lamotrigine • Lamictal
  • Levodopa/carbidopa • Sinemet
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Meclizine • Antivert
  • Memantine • Namenda
  • Mirtazapine • Remeron
  • Nefazodone • Serzone
  • Neostigmine • Prostigmin
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Oxybutynin • Ditropan
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Promethazine • Phenergan
  • Protriptyline • Vivactil
  • Pyridostigmine • Mestinon
  • Quetiapine • Seroquel
  • Ranitidine • Zantac
  • Risperidone • Risperdal
  • Rivastigmine • Exelon
  • Scopolamine • Scopace
  • Sertraline • Zoloft
  • Succinylcholine • Anectine
  • Temazepam • Restoril
  • Thioridazine • Mellaril
  • Tolterodine • Detrol
  • Trazodone • Desyrel
  • Trihexyphenidyl • Artane
  • Valproate • Depakote
  • Venlafaxine • Effexor
  • Warfarin • Coumadin
  • Ziprasidone • Geodon
  • Zolpidem • Ambien
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Mr. B, age 78, has a long history of well-controlled bipolar disorder and was diagnosed with Alzheimer’s dementia 6 months ago. He is living at home and has been taking donepezil, 10 mg/d, and lamotrigine, 100 mg bid.

This morning Mr. B’s wife calls and reports that he is experiencing sudden difficulty walking, dizziness, and “feeling drunk.” When you ask about Mr. B’s medications, his wife says that her husband’s internist had prescribed itraconazole, 200 mg/d, for onychomycosis, and Mr. B has taken 1 dose. You promptly discontinue the itraconazole, and Mr. B’s symptoms resolve.

Drug-drug interactions (DDIs) in Alzheimer’s disease (AD) patients such as Mr. B can be serious and even life-threatening. On average, persons age ≥65 use 4.5 prescription agents and 2 over-the-counter preparations per day,1 and the number of concurrently used medications is a significant predictor of adverse drug reactions.2

Cognitive enhancers, including acetylcholinesterase inhibitors (AChEIs) and memantine, are the most widely prescribed agents for AD patients. The FDA has approved galantamine and rivastigmine for mild to moderate dementia, memantine for moderate to severe dementia, and donepezil for mild to severe dementia (Table 1).3-5

To help you minimize adverse DDIs in AD patients, this article describes:

  • pharmacokinetic and pharmacodynamic effects of cognitive enhancers used in AD management
  • DDIs with medications commonly prescribed to AD patients
  • how to avoid adverse events related to antipsychotics, antidepressants, and benzodiazepines.

Table 1

Pharmacokinetic features of cognitive enhancers

AgentProtein bindingCYP-450 activityOther features
AChEIs
  Donepezil96%CYP 2D6, 3A4 substrateOnce-daily dosing
  Rivastigmine40%NoneMetabolized by cholinesterases
  Galantamine18%CYP 2D6, 3A4 substrateNicotinic cholinergic receptor modulation
NMDA receptor antagonist
  Memantine45%NoneNo hepatic metabolism
CYP-450: cytochrome P-450; AChEIs: acetylcholinesterase inhibitors; NMDA: N-methyl-D-aspartate
Source: References 3-5

Pharmacologic changes with aging

Pharmacokinetics is the study of the time course of drugs and their metabolites through the body. Pharmacokinetic interactions involve alterations in the plasma concentration of a drug by a second agent.3

Absorption of medications is decreased in the elderly because of reduced intestinal blood flow and motility. Absorption further decreases if patients concomitantly take antacids, high-fiber supplements, or anticholinergic medications.

Distribution. With aging, lean body mass typically decreases and adipose tissue increases. Because most psychotropics are lipid-soluble, their volume of distribution increases with age. This leads to drug accumulation and longer half-lives. On the other hand, water-soluble medications such as lithium distribute in a smaller volume and pose a higher risk of toxicity.

In plasma, drugs circulate freely or bound to proteins—mainly albumin and α1-acid glycoprotein. Aging can cause decreased plasma albumin and increased α1-acid glycoprotein.6 Additionally, malnutrition, diabetes mellitus, and hepatic and renal disease—all more common with advancing age—may cause hypoalbuminemia, which increases the free fraction of drugs bound to albumin.6Table 1 includes information about cognitive enhancers’ protein binding.

When 2 or more highly protein-bound drugs are coadministered, mutual displacement occurs and the free fraction of each drug increases. A recent case report described valproate toxicity with dizziness, ataxia, and falling in a 76-year-old man after aspirin was added to his regimen.7 The mechanism appeared to be mutual displacement from albumin combined with metabolism of valproate inhibited by aspirin.7

Metabolism. Liver size and hepatic blood flow decrease with aging.6 Cytochrome P-450 3A4 pathway activity slows, but the 2D6 pathway is not affected.4 Oxidative metabolism through CYP pathways is slower, but conjugation reactions are not.6Table 23,5,7,8 lists major substrates and inhibitors of CYP enzymes.

Azole antifungals are potent inhibitors of CYP 3A4,4 of which both donepezil and lamotrigine are substrates (Table 2). In Mr. B’s case, lamotrigine and donepezil levels increased because of this pharmacokinetic interaction. Because donepezil also is metabolized by the CYP 2D6 pathway, the increase in concentration is unlikely to modify the drug effect. Mr. B experienced symptoms consistent with lamotrigine toxicity.

Excretion. The age-associated decline in renal clearance related to a diminished glomerular filtration rate leads to decreased excretion of active metabolites and lithium, making older patients more susceptible to lithium toxicity. The magnitude of the decline in renal clearance varies among patients and is exacerbated by concomitant conditions—such as diabetes and hypertension—and medications—such as nonsteroidal anti-inflammatory drugs (NSAIDs).4 Thiazide diuretics, angiotensin-converting enzyme inhibitors, and cyclooxygenase-2 (COX-2) inhibitors such as celecoxib may elevate lithium levels.3

Pharmacokinetics of AChEIs. AChEIs have relatively few pharmacokinetic interactions, although donepezil and galantamine are metabolized through the liver’s CYP 2D6 and 3A4 pathways.

Because rivastigmine does not undergo hepatic metabolism, it is least likely of the cognitive enhancers to have pharmacokinetic interactions with other medications. Rivastigmine did not lead to increased adverse events when administered concomitantly with 22 different classes of medications—including antidiabetics, cardiovascular drugs, gastrointestinal agents, and NSAIDs.9

 

 

Table 2

DDIs in AD patients: CYP-450 substrates and inhibitors*

 CYP 2D6CYP 3A4
Substrates (substances metabolized by enzyme)Second-generation antipsychotics
Citalopram
Donepezil
Duloxetine
Galantamine
Haloperidol
Tricyclic antidepressants
Trazodone
Venlafaxine
Second-generation antipsychotics
Benzodiazepines
Buspirone
Carbamazepine
Donepezil
Galantamine
Haloperidol
Lamotrigine
Mirtazapine
Nefazodone
Sertraline
Tricyclic antidepressants
Trazodone
Zolpidem
InhibitorsBupropion
Cimetidine
Duloxetine
Fluoxetine
Paroxetine
Sertraline
Erythromycin
Fluconazole
Fluvoxamine
Grapefruit juice
Itraconazole
Nefazodone
*All cytochrome P (CYP) 450 enzymes are induced by barbiturates, phenytoin, carbamazepine, and rifampicin. Smoking also induces CYP 1A2.
DDIs: drug-drug interactions; AD: Alzheimer’s disease
Source: References 3,5,7,8
Pharmacodynamics is the study of the time course and intensity of drugs’ pharmacologic effects. Pharmacodynamic interactions involve changes in a drug’s action at a receptor or biologically active site.3 Pharmacodynamic interactions may result from an antagonistic or synergistic mechanism (Table 3).3,5,10 Dopamine neurons degenerate with aging, particularly after age 70, and the number of cholinergic receptors decreases in AD patients. As a result, these patients may become more sensitive to antipsychotics, selective serotonin reuptake inhibitors (SSRIs)—which indirectly reduce dopamine outflow—and medications with anticholinergic effects.4

Memantine, an amantadine derivative and N-methyl-D-aspartate (NMDA) receptor antagonist, is a weak dopaminergic agonist with atropinic effects.11 Because memantine is not metabolized by the CYP-450 pathway, it lacks pharmacokinetic DDIs.12 However, combining memantine with other NMDA antagonists—such as amantadine or dextromethorphan—could cause hallucinations, dizziness, headache, fatigue, and confusion.11 Concurrent use with drugs that lower seizure threshold, such as tricyclic antidepressants, may increase the risk of seizures.

Table 3

Potential drug-drug interactions in AD patients taking cognitive enhancers

InteractionMechanismPotential sequela(e)
AChEIs + anticholinergics↓ Acetylcholine in CNSCognitive worsening, delirium
AChEIs + beta blockersVagal stimulation and sympathetic blockadeBradycardia, syncope
AChEIs + cholinergics↑ Acetylcholine in PNSCholinergic crisis: hypersalivation, abdominal pain, diarrhea
AChEIs + antipsychotics (rare)↑ Acetylcholine/↓ dopamine in striatumParkinsonian syndrome, rigidity
Ginkgo biloba + warfarinAntiplatelet aggregation and anticoagulationGastrointestinal bleeding, hematuria, subcutaneous ecchymosis
AChEIs: acetylcholinesterase inhibitors; PNS: peripheral nervous system
Source: References 3,5,10

DDIs with cognitive enhancers

Anticholinergics. Because anticholinergic drugs can worsen cognitive impairment and cause delirium they are contraindicated in older patients—especially those with AD. Antihistamines, histamine H2 blockers, low-potency first-generation antipsychotics (FGAs), and tricyclic antidepressants are common medications with anticholinergic effects (Table 4).5,13,14

Anticholinergics can counteract AChEIs’ beneficial effect. Concurrent use of anticholinergics and AChEIs is fairly common in clinical practice but is rarely appropriate because of pharmacologic antagonism. In a retrospective study of 836 community-living older adults (age ≥65) with probable dementia, Roe et al13 compared anticholinergic use in 418 who were taking donepezil with 418 matched controls who were not taking donepezil. They found:

  • 33% of those taking donepezil also were receiving anticholinergics, compared with 23% of controls
  • 26% of all patients in the study used multiple anticholinergic medications.
Similarly, a study of pharmacy claims for AChEIs among 557 Medicaid beneficiaries aged ≥50 found that 35% of patients taking AChEIs also received at least 1 anticholinergic drug.14

Antiparkinsonian agents. Interaction of antiparkinsonian medications with AChEIs could limit the efficacy of either drug when treating comorbid AD and Parkinson’s disease (PD),5 although in practice, clinical deterioration of parkinsonism has not been reported.15 In one study, 25 PD patients stabilized on levodopa/carbidopa were given donepezil, 5 mg/d, or placebo for two 2-week courses separated by a washout of at least 2 weeks. At steady state, pharmacokinetic parameters were unchanged and no clinically significant DDIs were observed.16

Cardiovascular agents. Concurrent use of AChEIs and beta blockers, calcium channel inhibitors, or digoxin could worsen bradycardia and cause syncope. The risk is higher in patients:

  • with sick sinus syndrome or other bradyarrhythmias
  • taking antipsychotics that could induce torsades de pointes,11 such as ziprasidone or haloperidol.
In patients taking these cardiovascular drugs, make sure that heart rate is >60 bpm before AChEI treatment, and monitor regularly.

Other agents. AChEIs inhibit the metabolism of succinylcholine and therefore augment and prolong this drug’s neuromuscular blockade. Discontinue AChEIs before administering succinylcholine for anesthesia, such as for electroconvulsive treatment.

AChEIs may lead to toxicity when added to cholinergic agents such as bethanechol.11 Similarly, AChEIs may precipitate a cholinergic crisis—with increasing weakness, hypersalivation, abdominal pains, and diarrhea—when used in conjunction with peripheral acetylcholinesterase inhibitors such as the myasthenia gravis agents pyridostigmine and neostigmine. The mechanism is increased acetylcholine available at the neuromuscular junction.

Table 4

Medications with moderate to strong anticholinergic activity

ClassExamples
AntiarrhythmicsDisopyramide
AntiemeticsMeclizine
AntiparkinsoniansBenztropine, biperiden, trihexyphenidyl
AntipsychoticsChlorpromazine, clozapine, olanzapine, pimozide, thioridazine
AntihistaminesChlorpheniramine, cyproheptadine, diphenhydramine, hydroxyzine, Promethazine
Gastrointestinal/urinary antispasmodicsAtropine, belladonna alkaloids, dicyclomine, hyoscyamine, oxybutynin, scopolamine, tolterodine
H2 histamineCimetidine, ranitidine
Muscle relaxantsCyclobenzaprine
Tricyclic antidepressantsAmitriptyline, amoxapine, clomipramine, doxepin, imipramine, protriptyline
Source: References 5,13,14

DDIs with other psychotropics

 

 

Antipsychotics. Nearly one-half of AD patients experience delusions, often in the middle stage of the disease, and many are prescribed second-generation antipsychotics (SGAs) to control delusions, hallucinations, sundowning, agitation, or aggression. Concomitant use of AChEIs and antipsychotics may increase the risk of extrapyramidal symptoms by disrupting the acetylcholine/dopamine balance in the striatum.5

In AD patients taking donepezil and risperidone, case reports describe parkinsonian syndrome and rigidity with immobility, which resolved after the antipsychotic was discontinued.5,11 When rivastigmine and risperidone were coadministered, however, no clinically relevant adverse interactions were noted in a 20-week, open-label trial of 65 patients with AD, 10 with vascular dementia, and 10 with both.17

The FDA has warned of increased risk of death when SGAs are used to treat behavioral disturbances in dementia patients. In a recent meta-analysis of 15 placebo-controlled trials, cognitive tests scores worsened when AD patients took aripiprazole, olanzapine, quetiapine, or risperidone. A significant risk for cerebrovascular events was seen, especially with risperidone, although no clear causal relationship was established.18 Falls, injury, and syncope were not increased, and patients with less severe dementia, outpatients, and those selected for psychosis were less affected. Thus, provide careful follow-up and avoid long-term unwarranted antipsychotic use in AD patients.

Highly anticholinergic FGAs such as chlorpromazine are not recommended for AD patients (Table 4).

Antidepressants. Up to 30% of AD patients experience major depression.19 SSRIs are the antidepressants most often used to treat depression and anxiety in AD patients.

Citalopram, escitalopram, or venlafaxine are good choices for patients with AD because of minimal CYP inhibitory activity.4 Fluvoxamine, fluoxetine, and paroxetine inhibit CYP 2C9, through which warfarin and some other drugs with a narrow therapeutic index are metabolized.6

Benzodiazepines are contraindicated in elderly patients (especially those with AD) because of the high risk of delirium, worsened cognitive function, paradoxical disinhibition, and falls.20 If benzodiazepines are necessary to control anxiety, use intermediate-duration agents that do not undergo oxidative metabolism and have no active metabolites, such as lorazepam, oxazepam, or temazepam.19 See Table 2 for more information on benzodiazepine DDIs.

Herbal supplements. Ginkgo biloba and huperzine A (Chinese club moss) are the herbal supplements used most commonly by dementia patients. Ginkgo inhibits platelet aggregation and can cause bleeding complications, with or without concomitant antiplatelet or anticoagulant therapy such as aspirin, warfarin, and NSAIDs. Enzyme induction of CYP 2C19 by ginkgo, leading to subtherapeutic levels of anticonvulsants, has been implicated in a report of fatal seizures. Huperzine A is a natural cholinesterase inhibitor and should not be combined with AChEIs because of the risk of additive adverse effects.10

Related resources

  • Jacobson SA, Pies RW, Greenblatt DJ. Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002.
  • Sandson, NB. Drug-drug interaction primer. Washington, DC: American Psychiatric Publishing; 2007.
Drug brand names

  • Amantadine • Symmetrel
  • Amitriptyline • Elavil
  • Amoxapine • Asendin
  • Aripiprazole • Abilify
  • Atropine • Sal-Tropine
  • Benztropine • Cogentin
  • Bethanechol • Urecholine
  • Biperiden • Akineton
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Tegretol
  • Celecoxib • Celebrex
  • Chlorpheniramine • Chlor-Trimeton
  • Chlorpromazine • Thorazine
  • Cimetidine • Tagamet
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Cyclobenzaprine • Flexeril
  • Cyproheptadine • Periactin
  • Dextromethorphan • Benylin, Delsym, others
  • Dicyclomine • Bentyl
  • Digoxin • Lanoxin
  • Diphenhydramine • Benadryl
  • Disopyramide • Norpace
  • Donepezil • Aricept
  • Doxepin • Adapin, Sinequan
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Erythromycin • E-Mycin
  • Fluconazole • Diflucan
  • Fluvoxamine • Luvox
  • Fluoxetine • Prozac
  • Galantamine • Reminyl, Razadyne
  • Haloperidol • Haldol
  • Hydroxyzine • Vistaril
  • Hyoscyamine • Anaspaz, Levbid, Levsin
  • Imipramine • Tofranil
  • Itraconazole • Sporanox
  • Lamotrigine • Lamictal
  • Levodopa/carbidopa • Sinemet
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Meclizine • Antivert
  • Memantine • Namenda
  • Mirtazapine • Remeron
  • Nefazodone • Serzone
  • Neostigmine • Prostigmin
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Oxybutynin • Ditropan
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Promethazine • Phenergan
  • Protriptyline • Vivactil
  • Pyridostigmine • Mestinon
  • Quetiapine • Seroquel
  • Ranitidine • Zantac
  • Risperidone • Risperdal
  • Rivastigmine • Exelon
  • Scopolamine • Scopace
  • Sertraline • Zoloft
  • Succinylcholine • Anectine
  • Temazepam • Restoril
  • Thioridazine • Mellaril
  • Tolterodine • Detrol
  • Trazodone • Desyrel
  • Trihexyphenidyl • Artane
  • Valproate • Depakote
  • Venlafaxine • Effexor
  • Warfarin • Coumadin
  • Ziprasidone • Geodon
  • Zolpidem • Ambien
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Prescription drugs and the elderly: many still receive potentially harmful drugs despite recent improvements. Washington, DC: United States General Accounting Office; 1996. Publication HEHS 95-152.

2. Atkin PA, Veitch PC, Veitch EM, Ogle SJ. The epidemiology of serious adverse drug reactions among the elderly. Drugs Aging 1999;14:141-52.

3. Marangell LB, Martinez JM, Silver JM, Yudofsky SC, eds. Concise guide to psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:4-7, 129,173, 171-80.

4. Roose SP, Pollock BG, Devanand DD. Treatment during late life. In: Schatzberg AF, Nemeroff CB, eds. Textbook of psychopharmacology. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:1083-5.

5. Bentue-Ferrer D, Tribut O, Polard E, Allain H. Clinically significant drug interactions with cholinesterase inhibitors: a guide for neurologists. CNS Drugs 2003;17:947-63.

6. Mulsant BH, Pollock BG. Psychopharmacology. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:387-411.

7. Sandson NB, Marcucci C, Bourke DL, Smith-Lamacchia R. An interaction between aspirin and valproate: the relevance of plasma protein displacement drug-drug interaction. Am J Psychiatry 2006;163:1891-6.

8. Spina E, Scordo MG, D’Arrigo C. Metabolic drug interactions with new psychotropic agents. Fundam Clin Pharmacol 2003;17:517-38.

9. Grossberg GT, Stahelin HB, Messina JC, et al. Lack of adverse pharmacodynamic drug interactions with rivastigmine and twenty-two classes of medications. Int J Geriatr Psychiatry 2000;15:242-7.

10. Beier MT. Harmless herbs? Think again: merits of a complete medication history. J Am Med Dir Assoc 2006;7:446-7.

11. [No authors listed]. Alzheimer’s disease: beware of interactions with cholinesterase inhibitors. Prescrire Int 2006;15:103-6.

12. Grossberg GT, Edwards KR, Zhao Q. Rationale for combining therapy with galantamine and memantine in Alzheimer’s disease. J Clin Pharmacol 2006;46(suppl 1):S17-S26.

13. Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medications by older adults with dementia. J Am Geriatr Soc 2002;50:836-42.

14. Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The concurrent use of anticholinergics and cholinesterase inhibitors: rare event or common practice? J Am Geriatr Soc 2004;52:2082-7.

15. Schrag A. Psychiatric aspects of Parkinson’s disease. J Neurol 2004;251:795-804.

16. Okereke CS, Kirby L, Kumar D, et al. Concurrent administration of donepezil HCl and levodopa/carbidopa in patients with Parkinson’s disease: assessment of pharmacokinetic changes and safety following multiple oral doses. Br J Clin Pharmacol 2004;58(suppl 1):41-9.

17. Weiser M, Rotmensch HH, Korczyn AD, et al. A pilot, randomized, open-label trial assessing safety and pharmacokinetic parameters of co-administration of rivastigmine with risperidone in dementia patients with behavioral disturbances. Int J Geriatr Psychiatry 2002;17:343-6.

18. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse events of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry 2006;14(3):191-210.

19. Koenig HG, Blazer DG. Mood disorders. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:254.

20. Jacobson SA, Pies RW, Greenblatt DJ. Anxiolytic and sedative-hypnotic medications. In: Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:249-312.

References

1. Prescription drugs and the elderly: many still receive potentially harmful drugs despite recent improvements. Washington, DC: United States General Accounting Office; 1996. Publication HEHS 95-152.

2. Atkin PA, Veitch PC, Veitch EM, Ogle SJ. The epidemiology of serious adverse drug reactions among the elderly. Drugs Aging 1999;14:141-52.

3. Marangell LB, Martinez JM, Silver JM, Yudofsky SC, eds. Concise guide to psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:4-7, 129,173, 171-80.

4. Roose SP, Pollock BG, Devanand DD. Treatment during late life. In: Schatzberg AF, Nemeroff CB, eds. Textbook of psychopharmacology. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:1083-5.

5. Bentue-Ferrer D, Tribut O, Polard E, Allain H. Clinically significant drug interactions with cholinesterase inhibitors: a guide for neurologists. CNS Drugs 2003;17:947-63.

6. Mulsant BH, Pollock BG. Psychopharmacology. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry. 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:387-411.

7. Sandson NB, Marcucci C, Bourke DL, Smith-Lamacchia R. An interaction between aspirin and valproate: the relevance of plasma protein displacement drug-drug interaction. Am J Psychiatry 2006;163:1891-6.

8. Spina E, Scordo MG, D’Arrigo C. Metabolic drug interactions with new psychotropic agents. Fundam Clin Pharmacol 2003;17:517-38.

9. Grossberg GT, Stahelin HB, Messina JC, et al. Lack of adverse pharmacodynamic drug interactions with rivastigmine and twenty-two classes of medications. Int J Geriatr Psychiatry 2000;15:242-7.

10. Beier MT. Harmless herbs? Think again: merits of a complete medication history. J Am Med Dir Assoc 2006;7:446-7.

11. [No authors listed]. Alzheimer’s disease: beware of interactions with cholinesterase inhibitors. Prescrire Int 2006;15:103-6.

12. Grossberg GT, Edwards KR, Zhao Q. Rationale for combining therapy with galantamine and memantine in Alzheimer’s disease. J Clin Pharmacol 2006;46(suppl 1):S17-S26.

13. Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medications by older adults with dementia. J Am Geriatr Soc 2002;50:836-42.

14. Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The concurrent use of anticholinergics and cholinesterase inhibitors: rare event or common practice? J Am Geriatr Soc 2004;52:2082-7.

15. Schrag A. Psychiatric aspects of Parkinson’s disease. J Neurol 2004;251:795-804.

16. Okereke CS, Kirby L, Kumar D, et al. Concurrent administration of donepezil HCl and levodopa/carbidopa in patients with Parkinson’s disease: assessment of pharmacokinetic changes and safety following multiple oral doses. Br J Clin Pharmacol 2004;58(suppl 1):41-9.

17. Weiser M, Rotmensch HH, Korczyn AD, et al. A pilot, randomized, open-label trial assessing safety and pharmacokinetic parameters of co-administration of rivastigmine with risperidone in dementia patients with behavioral disturbances. Int J Geriatr Psychiatry 2002;17:343-6.

18. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse events of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry 2006;14(3):191-210.

19. Koenig HG, Blazer DG. Mood disorders. In: Blazer DG, Steffens DC, Busse EW, eds. Textbook of geriatric psychiatry 3rd ed. Washington, DC: American Psychiatric Publishing; 2004:254.

20. Jacobson SA, Pies RW, Greenblatt DJ. Anxiolytic and sedative-hypnotic medications. In: Handbook of geriatric psychopharmacology. Washington, DC: American Psychiatric Publishing; 2002:249-312.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
57-66
Page Number
57-66
Publications
Publications
Article Type
Display Headline
Prevent drug-drug interactions with cholinesterase inhibitors
Display Headline
Prevent drug-drug interactions with cholinesterase inhibitors
Legacy Keywords
Andreea L. Seritan MD; drug-drug interactions; cholinesterase inhibitors; Alzheimer’s dementia; Alzheimer’s disease; acetylcholinesterase inhibitors; AChEIs; memantine; galantamine; rivastigmine; donepezil; dementia
Legacy Keywords
Andreea L. Seritan MD; drug-drug interactions; cholinesterase inhibitors; Alzheimer’s dementia; Alzheimer’s disease; acetylcholinesterase inhibitors; AChEIs; memantine; galantamine; rivastigmine; donepezil; dementia
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Was the patient still suicidal?

Article Type
Changed
Mon, 04/16/2018 - 14:21
Display Headline
Was the patient still suicidal?

THE PATIENT. A 30-year-old police officer reports thoughts of suicide. He was under investigation for illegal work-related activities and feared he would have to report his coworkers’ involvement in these activities and lose his job.

CASE FACTS. The patient was voluntarily hospitalized for 4 days and received medication and inpatient psychotherapy. When he was discharged, a psychiatrist prescribed follow-up outpatient psychotherapy and antidepressant and antipsychotic medications. The next day, the officer fatally shot himself.

THE PATIENT’S FAMILY’S CLAIM. The psychiatrist did not adequately weigh the patient’s depression and stressors, including possibly losing his job, and did not properly assess suicidal ideation. Also, the patient’s mother claims she attended the discharge meeting with the psychiatrist and that her son expressed suicidal intentions at that time.

THE DOCTOR’S DEFENSE. The patient believed he could get another job if necessary and was no longer contemplating suicide. Also, he was a voluntary patient and could not be hospitalized any longer without consent.

Submit your verdict and find out how the court ruled at CurrentPsychiatry.com. Click on “Have more to say about this topic?” to comment.

References

Cases are selected by Current Psychiatry from Medical Malpractice Verdicts, Settlements & Experts, with permission of its editor, Lewis Laska of Nashville, TN (www.verdictslaska.com). Information may be incomplete in some instances, but these cases represent clinical situations that typically result in litigation.

Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 07(02)
Publications
Page Number
86-86
Legacy Keywords
malpractice; suicide; depression
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

THE PATIENT. A 30-year-old police officer reports thoughts of suicide. He was under investigation for illegal work-related activities and feared he would have to report his coworkers’ involvement in these activities and lose his job.

CASE FACTS. The patient was voluntarily hospitalized for 4 days and received medication and inpatient psychotherapy. When he was discharged, a psychiatrist prescribed follow-up outpatient psychotherapy and antidepressant and antipsychotic medications. The next day, the officer fatally shot himself.

THE PATIENT’S FAMILY’S CLAIM. The psychiatrist did not adequately weigh the patient’s depression and stressors, including possibly losing his job, and did not properly assess suicidal ideation. Also, the patient’s mother claims she attended the discharge meeting with the psychiatrist and that her son expressed suicidal intentions at that time.

THE DOCTOR’S DEFENSE. The patient believed he could get another job if necessary and was no longer contemplating suicide. Also, he was a voluntary patient and could not be hospitalized any longer without consent.

Submit your verdict and find out how the court ruled at CurrentPsychiatry.com. Click on “Have more to say about this topic?” to comment.

THE PATIENT. A 30-year-old police officer reports thoughts of suicide. He was under investigation for illegal work-related activities and feared he would have to report his coworkers’ involvement in these activities and lose his job.

CASE FACTS. The patient was voluntarily hospitalized for 4 days and received medication and inpatient psychotherapy. When he was discharged, a psychiatrist prescribed follow-up outpatient psychotherapy and antidepressant and antipsychotic medications. The next day, the officer fatally shot himself.

THE PATIENT’S FAMILY’S CLAIM. The psychiatrist did not adequately weigh the patient’s depression and stressors, including possibly losing his job, and did not properly assess suicidal ideation. Also, the patient’s mother claims she attended the discharge meeting with the psychiatrist and that her son expressed suicidal intentions at that time.

THE DOCTOR’S DEFENSE. The patient believed he could get another job if necessary and was no longer contemplating suicide. Also, he was a voluntary patient and could not be hospitalized any longer without consent.

Submit your verdict and find out how the court ruled at CurrentPsychiatry.com. Click on “Have more to say about this topic?” to comment.

References

Cases are selected by Current Psychiatry from Medical Malpractice Verdicts, Settlements & Experts, with permission of its editor, Lewis Laska of Nashville, TN (www.verdictslaska.com). Information may be incomplete in some instances, but these cases represent clinical situations that typically result in litigation.

References

Cases are selected by Current Psychiatry from Medical Malpractice Verdicts, Settlements & Experts, with permission of its editor, Lewis Laska of Nashville, TN (www.verdictslaska.com). Information may be incomplete in some instances, but these cases represent clinical situations that typically result in litigation.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
86-86
Page Number
86-86
Publications
Publications
Article Type
Display Headline
Was the patient still suicidal?
Display Headline
Was the patient still suicidal?
Legacy Keywords
malpractice; suicide; depression
Legacy Keywords
malpractice; suicide; depression
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Tips to make documentation easier, faster, and more satisfying

Article Type
Changed
Mon, 04/16/2018 - 14:21
Display Headline
Tips to make documentation easier, faster, and more satisfying

Current Psychiatry’s malpractice column is evolving. Previously, “Malpractice Verdicts,” used case decisions to initiate discussions of clinical situations that can generate lawsuits. The verdicts remain as “Malpractice Minute”, but Current Psychiatry has invited me to contribute a new column, “Malpractice Rx,” that will solicit questions and address practicing clinicians’ concerns about malpractice risk.

To start this dialogue, I’ll begin with a question that often comes up in discussions with colleagues, and especially when I teach psychiatry residents: “What should I document?” In this article, we will review why proper documentation is essential. We’ll also look at some ideas that might make documentation easier, more efficient, and more satisfying.

Do you have a question about possible liability?

  • If so, please submit your malpractice-related questions to Dr. Mossman at [email protected].
  • Include your name, address, and practice location. If your question is chosen for publication, your name can be withheld by request.
  • All readers who submit questions will be included in quarterly drawings for a $50 gift certificate for Professional Risk Management Services, Inc’s online marketplace of risk management publications and resources (www.prms.com).
Purposes of documentation

When I was in medical school, my professors said the primary reason for accurate charting was to communicate with the rest of the treatment team. This is still true. But in these sadder-but-wiser days, when I ask psychiatry residents “What is the purpose of documentation?” they always answer, “to create a legal record.”

Documentation plays many roles (Table 1). From the standpoint of preventing a malpractice judgment, the clinical record can accomplish 3 important things:

Lawsuit deterrence. Records are a key source—and often the only source—of information an attorney uses when deciding whether to file a lawsuit. An attorney won’t risk time and money on a malpractice case if the clinical record suggests that a psychiatrist was conscientious and met the standard of care.1

Impression management. The patient’s chart is what plaintiffs’ and defendants’ experts use when forming their initial opinions about the quality of care delivered.

Credibility. Clinical records are the most believable source of information about what you observed, what you thought, what you did, why you did it, and when you did it. The adage “if it wasn’t written, it didn’t happen” is not always applicable,2 but if an adverse event occurs, a defendant doctor’s verbal testimony about delivering good care will be more convincing when backed up by documentation created before the event.

Table 1

Purposes of medical record documentation

  • Communicate clinical information to current and future caregivers
  • Remind you of what happened and what you did
  • Justify care to third-party payers
  • Inform professional standards review organizations
  • Satisfy accrediting agencies
  • Create a basis for defense in a malpractice action

Improving documentation

Because it is impossible to describe everything you see, hear, say, do, and think during clinical encounters with patients, you must make choices about what to include in the record. The components of good documentation depend on the clinical context, but the following general principles may avert some malpractice actions.

1 More is better. Psychiatric practice often requires you to be discreet about patients’ personal information. Within appropriate bounds, however, the more information the record contains about objective findings, patients’ statements, clinical judgments, and your decision making, the better the portrayal of competent care.

2 Record the time and date. When attorneys and experts try to reconstruct what happened before an adverse occurrence, knowing the exact time you saw the patient, recorded findings, wrote orders, followed up on lab tests, or discussed problems with others—including family and treatment team members—can make a big difference.

3 Sooner is better. The most credible charting is done during or just after a service is rendered. Charting completed after an adverse event is vulnerable to accusations of fabrication.

4 Describe your thinking. Most aspects of clinical medicine are far from certain. Documenting the reasoning behind your diagnosis and treatment selection—what you’ve ruled out, what still seems tentative, and what risks and benefits you’ve weighed—helps emphasize this reality.3 After something bad happens, people retrospectively regard the event as more probable than it really was.4 Documenting your uncertainty and ways of addressing it may help counter this “hindsight bias.” It also shows that you were thoughtful and took therapeutic steps prudently.

5 Collaborate with the patient. In some circumstances, it is appropriate to draft documents in a patient’s presence. Examples might include information sent to third-party payers or referrals to other clinicians. Noting that you’ve done this demonstrates the patient’s awareness and implicit concurrence. Also, collaborative documentation reinforces the “working together” aspects of a doctor-patient relationship and can be therapeutic.
 

 

5

6 Clarify capacity. Jurors may believe that all psychiatric patients are incompetent, and plaintiff’s attorneys sometimes try to create the impression that patients are completely controlled by weird whims and aberrant thoughts. To counter this, when appropriate indicate in the chart that the patient can handle responsibilities such as reporting side effects, seeking emergency attention, or notifying you about changes in thought or mood.3,5

7 Manage appearance and content. Under Health Insurance Portability and Accountability Act (HIPAA) regulations, patients have the right to review their medical records.6 If a lawsuit occurs, the records might be read out loud in court. Documentation will make a better impression if it is clear, legible, and free of gratuitous comments.

8 Include quotations. Documenting verbatim statements from a patient, such as “I’ve never considered suicide,” can quickly convey key information that you considered when making a therapeutic decision.

Technical approaches

Table 27,8 lists several techniques and technologies that might improve documentation. For example, computer users can create templates or customize software to quickly produce thorough documentation for frequently encountered procedures or clinical events. Whether these approaches are useful and appropriate will depend on your work setting, but all aim to improve the speed and quality of clinical documentation.

Think creatively about improving documentation. Even if you’re never sued, better documentation helps you and your patients. For example, several years ago a colleague9 designed an emergency room form that allowed clinicians to complete in a few seconds a Brief Psychiatric Rating Scale on every patient we evaluated. This innovation shortened the time needed to document a systematic, comprehensive assessment and increased the quantity, quality, and reliability of information in patients’ records.

Table 2

Purposes of medical record documentation

IdeaComment
Use speech recognitionYou speak faster than you write. Transcription software
Softwareaccuracy has improved in the last few years.
Use handouts andPatients often do not remember or understand much of what
medication instructionsdoctors tell them,7,8 so handouts may be more useful than verbal instructions. Good handouts about medications are available on the Internet. Note in the chart that you gave the patient the document.
Seek anonymousDocumenting consultations shows you are prudent and
consultations with colleaguesa colleague agreed with your treatment.
Ask patients to rate theirThis practice may improve your information gathering
own symptoms and progressand help document what the patient told you.
Use standard rating scalesRating scales can help you record more information in a scientifically validated format.
Use macros and templatesMacros can reduce time needed for documentation. Your memory isn’t perfect, but templates can help you include everything you need to cover.
References

1. Simpson S, Stacy M. Avoiding the malpractice snare: documenting suicide risk assessment. J Psychiatr Pract 2004;10:185-9.

2. Zurad EG. Don’t be the target of a malpractice suit. Fam Pract Manag 2006;13(6):57-64.

3. Gutheil TG. Fundamentals of medical record documentation. Psychiatry 2004;1:26-8.

4. Fischhoff B, Beyth R. “I knew it would happen” remembered probabilities of once-future things. Organ Behav Hum Perform 1975;13:1-16.

5. Appelbaum PS, Gutheil TG. Clinical handbook of psychiatry and the law 4th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2007.

6. 45 CFR § 164.524(a)(1).

7. Rogers AE, Addington-Hall JM, Abery AJ, et al. Knowledge and communication difficulties for patients with chronic heart failure: qualitative study. BMJ 2000;321:605-7.

8. Chesanow N. Are you getting through? Med Econ 2006;83(13):41,45-6.

9. Somoza E, Somoza JR. A neural-network approach to predicting admission decisions in a psychiatric emergency room. Med Decis Making 1993;13:273-80.

Article PDF
Author and Disclosure Information

Douglas Mossman, MD
Dr. Mossman is professor and director, division of forensic psychiatry, Wright State University Boonshoft School of Medicine, Dayton, OH, and administrative director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law.

Issue
Current Psychiatry - 07(02)
Publications
Page Number
80-86
Legacy Keywords
Douglas Mossman MD; documentation; malpractice
Sections
Author and Disclosure Information

Douglas Mossman, MD
Dr. Mossman is professor and director, division of forensic psychiatry, Wright State University Boonshoft School of Medicine, Dayton, OH, and administrative director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law.

Author and Disclosure Information

Douglas Mossman, MD
Dr. Mossman is professor and director, division of forensic psychiatry, Wright State University Boonshoft School of Medicine, Dayton, OH, and administrative director, Glenn M. Weaver Institute of Law and Psychiatry, University of Cincinnati College of Law.

Article PDF
Article PDF

Current Psychiatry’s malpractice column is evolving. Previously, “Malpractice Verdicts,” used case decisions to initiate discussions of clinical situations that can generate lawsuits. The verdicts remain as “Malpractice Minute”, but Current Psychiatry has invited me to contribute a new column, “Malpractice Rx,” that will solicit questions and address practicing clinicians’ concerns about malpractice risk.

To start this dialogue, I’ll begin with a question that often comes up in discussions with colleagues, and especially when I teach psychiatry residents: “What should I document?” In this article, we will review why proper documentation is essential. We’ll also look at some ideas that might make documentation easier, more efficient, and more satisfying.

Do you have a question about possible liability?

  • If so, please submit your malpractice-related questions to Dr. Mossman at [email protected].
  • Include your name, address, and practice location. If your question is chosen for publication, your name can be withheld by request.
  • All readers who submit questions will be included in quarterly drawings for a $50 gift certificate for Professional Risk Management Services, Inc’s online marketplace of risk management publications and resources (www.prms.com).
Purposes of documentation

When I was in medical school, my professors said the primary reason for accurate charting was to communicate with the rest of the treatment team. This is still true. But in these sadder-but-wiser days, when I ask psychiatry residents “What is the purpose of documentation?” they always answer, “to create a legal record.”

Documentation plays many roles (Table 1). From the standpoint of preventing a malpractice judgment, the clinical record can accomplish 3 important things:

Lawsuit deterrence. Records are a key source—and often the only source—of information an attorney uses when deciding whether to file a lawsuit. An attorney won’t risk time and money on a malpractice case if the clinical record suggests that a psychiatrist was conscientious and met the standard of care.1

Impression management. The patient’s chart is what plaintiffs’ and defendants’ experts use when forming their initial opinions about the quality of care delivered.

Credibility. Clinical records are the most believable source of information about what you observed, what you thought, what you did, why you did it, and when you did it. The adage “if it wasn’t written, it didn’t happen” is not always applicable,2 but if an adverse event occurs, a defendant doctor’s verbal testimony about delivering good care will be more convincing when backed up by documentation created before the event.

Table 1

Purposes of medical record documentation

  • Communicate clinical information to current and future caregivers
  • Remind you of what happened and what you did
  • Justify care to third-party payers
  • Inform professional standards review organizations
  • Satisfy accrediting agencies
  • Create a basis for defense in a malpractice action

Improving documentation

Because it is impossible to describe everything you see, hear, say, do, and think during clinical encounters with patients, you must make choices about what to include in the record. The components of good documentation depend on the clinical context, but the following general principles may avert some malpractice actions.

1 More is better. Psychiatric practice often requires you to be discreet about patients’ personal information. Within appropriate bounds, however, the more information the record contains about objective findings, patients’ statements, clinical judgments, and your decision making, the better the portrayal of competent care.

2 Record the time and date. When attorneys and experts try to reconstruct what happened before an adverse occurrence, knowing the exact time you saw the patient, recorded findings, wrote orders, followed up on lab tests, or discussed problems with others—including family and treatment team members—can make a big difference.

3 Sooner is better. The most credible charting is done during or just after a service is rendered. Charting completed after an adverse event is vulnerable to accusations of fabrication.

4 Describe your thinking. Most aspects of clinical medicine are far from certain. Documenting the reasoning behind your diagnosis and treatment selection—what you’ve ruled out, what still seems tentative, and what risks and benefits you’ve weighed—helps emphasize this reality.3 After something bad happens, people retrospectively regard the event as more probable than it really was.4 Documenting your uncertainty and ways of addressing it may help counter this “hindsight bias.” It also shows that you were thoughtful and took therapeutic steps prudently.

5 Collaborate with the patient. In some circumstances, it is appropriate to draft documents in a patient’s presence. Examples might include information sent to third-party payers or referrals to other clinicians. Noting that you’ve done this demonstrates the patient’s awareness and implicit concurrence. Also, collaborative documentation reinforces the “working together” aspects of a doctor-patient relationship and can be therapeutic.
 

 

5

6 Clarify capacity. Jurors may believe that all psychiatric patients are incompetent, and plaintiff’s attorneys sometimes try to create the impression that patients are completely controlled by weird whims and aberrant thoughts. To counter this, when appropriate indicate in the chart that the patient can handle responsibilities such as reporting side effects, seeking emergency attention, or notifying you about changes in thought or mood.3,5

7 Manage appearance and content. Under Health Insurance Portability and Accountability Act (HIPAA) regulations, patients have the right to review their medical records.6 If a lawsuit occurs, the records might be read out loud in court. Documentation will make a better impression if it is clear, legible, and free of gratuitous comments.

8 Include quotations. Documenting verbatim statements from a patient, such as “I’ve never considered suicide,” can quickly convey key information that you considered when making a therapeutic decision.

Technical approaches

Table 27,8 lists several techniques and technologies that might improve documentation. For example, computer users can create templates or customize software to quickly produce thorough documentation for frequently encountered procedures or clinical events. Whether these approaches are useful and appropriate will depend on your work setting, but all aim to improve the speed and quality of clinical documentation.

Think creatively about improving documentation. Even if you’re never sued, better documentation helps you and your patients. For example, several years ago a colleague9 designed an emergency room form that allowed clinicians to complete in a few seconds a Brief Psychiatric Rating Scale on every patient we evaluated. This innovation shortened the time needed to document a systematic, comprehensive assessment and increased the quantity, quality, and reliability of information in patients’ records.

Table 2

Purposes of medical record documentation

IdeaComment
Use speech recognitionYou speak faster than you write. Transcription software
Softwareaccuracy has improved in the last few years.
Use handouts andPatients often do not remember or understand much of what
medication instructionsdoctors tell them,7,8 so handouts may be more useful than verbal instructions. Good handouts about medications are available on the Internet. Note in the chart that you gave the patient the document.
Seek anonymousDocumenting consultations shows you are prudent and
consultations with colleaguesa colleague agreed with your treatment.
Ask patients to rate theirThis practice may improve your information gathering
own symptoms and progressand help document what the patient told you.
Use standard rating scalesRating scales can help you record more information in a scientifically validated format.
Use macros and templatesMacros can reduce time needed for documentation. Your memory isn’t perfect, but templates can help you include everything you need to cover.

Current Psychiatry’s malpractice column is evolving. Previously, “Malpractice Verdicts,” used case decisions to initiate discussions of clinical situations that can generate lawsuits. The verdicts remain as “Malpractice Minute”, but Current Psychiatry has invited me to contribute a new column, “Malpractice Rx,” that will solicit questions and address practicing clinicians’ concerns about malpractice risk.

To start this dialogue, I’ll begin with a question that often comes up in discussions with colleagues, and especially when I teach psychiatry residents: “What should I document?” In this article, we will review why proper documentation is essential. We’ll also look at some ideas that might make documentation easier, more efficient, and more satisfying.

Do you have a question about possible liability?

  • If so, please submit your malpractice-related questions to Dr. Mossman at [email protected].
  • Include your name, address, and practice location. If your question is chosen for publication, your name can be withheld by request.
  • All readers who submit questions will be included in quarterly drawings for a $50 gift certificate for Professional Risk Management Services, Inc’s online marketplace of risk management publications and resources (www.prms.com).
Purposes of documentation

When I was in medical school, my professors said the primary reason for accurate charting was to communicate with the rest of the treatment team. This is still true. But in these sadder-but-wiser days, when I ask psychiatry residents “What is the purpose of documentation?” they always answer, “to create a legal record.”

Documentation plays many roles (Table 1). From the standpoint of preventing a malpractice judgment, the clinical record can accomplish 3 important things:

Lawsuit deterrence. Records are a key source—and often the only source—of information an attorney uses when deciding whether to file a lawsuit. An attorney won’t risk time and money on a malpractice case if the clinical record suggests that a psychiatrist was conscientious and met the standard of care.1

Impression management. The patient’s chart is what plaintiffs’ and defendants’ experts use when forming their initial opinions about the quality of care delivered.

Credibility. Clinical records are the most believable source of information about what you observed, what you thought, what you did, why you did it, and when you did it. The adage “if it wasn’t written, it didn’t happen” is not always applicable,2 but if an adverse event occurs, a defendant doctor’s verbal testimony about delivering good care will be more convincing when backed up by documentation created before the event.

Table 1

Purposes of medical record documentation

  • Communicate clinical information to current and future caregivers
  • Remind you of what happened and what you did
  • Justify care to third-party payers
  • Inform professional standards review organizations
  • Satisfy accrediting agencies
  • Create a basis for defense in a malpractice action

Improving documentation

Because it is impossible to describe everything you see, hear, say, do, and think during clinical encounters with patients, you must make choices about what to include in the record. The components of good documentation depend on the clinical context, but the following general principles may avert some malpractice actions.

1 More is better. Psychiatric practice often requires you to be discreet about patients’ personal information. Within appropriate bounds, however, the more information the record contains about objective findings, patients’ statements, clinical judgments, and your decision making, the better the portrayal of competent care.

2 Record the time and date. When attorneys and experts try to reconstruct what happened before an adverse occurrence, knowing the exact time you saw the patient, recorded findings, wrote orders, followed up on lab tests, or discussed problems with others—including family and treatment team members—can make a big difference.

3 Sooner is better. The most credible charting is done during or just after a service is rendered. Charting completed after an adverse event is vulnerable to accusations of fabrication.

4 Describe your thinking. Most aspects of clinical medicine are far from certain. Documenting the reasoning behind your diagnosis and treatment selection—what you’ve ruled out, what still seems tentative, and what risks and benefits you’ve weighed—helps emphasize this reality.3 After something bad happens, people retrospectively regard the event as more probable than it really was.4 Documenting your uncertainty and ways of addressing it may help counter this “hindsight bias.” It also shows that you were thoughtful and took therapeutic steps prudently.

5 Collaborate with the patient. In some circumstances, it is appropriate to draft documents in a patient’s presence. Examples might include information sent to third-party payers or referrals to other clinicians. Noting that you’ve done this demonstrates the patient’s awareness and implicit concurrence. Also, collaborative documentation reinforces the “working together” aspects of a doctor-patient relationship and can be therapeutic.
 

 

5

6 Clarify capacity. Jurors may believe that all psychiatric patients are incompetent, and plaintiff’s attorneys sometimes try to create the impression that patients are completely controlled by weird whims and aberrant thoughts. To counter this, when appropriate indicate in the chart that the patient can handle responsibilities such as reporting side effects, seeking emergency attention, or notifying you about changes in thought or mood.3,5

7 Manage appearance and content. Under Health Insurance Portability and Accountability Act (HIPAA) regulations, patients have the right to review their medical records.6 If a lawsuit occurs, the records might be read out loud in court. Documentation will make a better impression if it is clear, legible, and free of gratuitous comments.

8 Include quotations. Documenting verbatim statements from a patient, such as “I’ve never considered suicide,” can quickly convey key information that you considered when making a therapeutic decision.

Technical approaches

Table 27,8 lists several techniques and technologies that might improve documentation. For example, computer users can create templates or customize software to quickly produce thorough documentation for frequently encountered procedures or clinical events. Whether these approaches are useful and appropriate will depend on your work setting, but all aim to improve the speed and quality of clinical documentation.

Think creatively about improving documentation. Even if you’re never sued, better documentation helps you and your patients. For example, several years ago a colleague9 designed an emergency room form that allowed clinicians to complete in a few seconds a Brief Psychiatric Rating Scale on every patient we evaluated. This innovation shortened the time needed to document a systematic, comprehensive assessment and increased the quantity, quality, and reliability of information in patients’ records.

Table 2

Purposes of medical record documentation

IdeaComment
Use speech recognitionYou speak faster than you write. Transcription software
Softwareaccuracy has improved in the last few years.
Use handouts andPatients often do not remember or understand much of what
medication instructionsdoctors tell them,7,8 so handouts may be more useful than verbal instructions. Good handouts about medications are available on the Internet. Note in the chart that you gave the patient the document.
Seek anonymousDocumenting consultations shows you are prudent and
consultations with colleaguesa colleague agreed with your treatment.
Ask patients to rate theirThis practice may improve your information gathering
own symptoms and progressand help document what the patient told you.
Use standard rating scalesRating scales can help you record more information in a scientifically validated format.
Use macros and templatesMacros can reduce time needed for documentation. Your memory isn’t perfect, but templates can help you include everything you need to cover.
References

1. Simpson S, Stacy M. Avoiding the malpractice snare: documenting suicide risk assessment. J Psychiatr Pract 2004;10:185-9.

2. Zurad EG. Don’t be the target of a malpractice suit. Fam Pract Manag 2006;13(6):57-64.

3. Gutheil TG. Fundamentals of medical record documentation. Psychiatry 2004;1:26-8.

4. Fischhoff B, Beyth R. “I knew it would happen” remembered probabilities of once-future things. Organ Behav Hum Perform 1975;13:1-16.

5. Appelbaum PS, Gutheil TG. Clinical handbook of psychiatry and the law 4th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2007.

6. 45 CFR § 164.524(a)(1).

7. Rogers AE, Addington-Hall JM, Abery AJ, et al. Knowledge and communication difficulties for patients with chronic heart failure: qualitative study. BMJ 2000;321:605-7.

8. Chesanow N. Are you getting through? Med Econ 2006;83(13):41,45-6.

9. Somoza E, Somoza JR. A neural-network approach to predicting admission decisions in a psychiatric emergency room. Med Decis Making 1993;13:273-80.

References

1. Simpson S, Stacy M. Avoiding the malpractice snare: documenting suicide risk assessment. J Psychiatr Pract 2004;10:185-9.

2. Zurad EG. Don’t be the target of a malpractice suit. Fam Pract Manag 2006;13(6):57-64.

3. Gutheil TG. Fundamentals of medical record documentation. Psychiatry 2004;1:26-8.

4. Fischhoff B, Beyth R. “I knew it would happen” remembered probabilities of once-future things. Organ Behav Hum Perform 1975;13:1-16.

5. Appelbaum PS, Gutheil TG. Clinical handbook of psychiatry and the law 4th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2007.

6. 45 CFR § 164.524(a)(1).

7. Rogers AE, Addington-Hall JM, Abery AJ, et al. Knowledge and communication difficulties for patients with chronic heart failure: qualitative study. BMJ 2000;321:605-7.

8. Chesanow N. Are you getting through? Med Econ 2006;83(13):41,45-6.

9. Somoza E, Somoza JR. A neural-network approach to predicting admission decisions in a psychiatric emergency room. Med Decis Making 1993;13:273-80.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
80-86
Page Number
80-86
Publications
Publications
Article Type
Display Headline
Tips to make documentation easier, faster, and more satisfying
Display Headline
Tips to make documentation easier, faster, and more satisfying
Legacy Keywords
Douglas Mossman MD; documentation; malpractice
Legacy Keywords
Douglas Mossman MD; documentation; malpractice
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Pregnant and mentally ill: A labor-intensive clinical challenge

Article Type
Changed
Mon, 04/16/2018 - 14:21
Display Headline
Pregnant and mentally ill: A labor-intensive clinical challenge

Life in the womb is fraught with hazards. Any deviation from a healthy pregnancy can damage a major organ system (first trimester), impair brain structural/functional development (second trimester), or cause prematurity and low birth weight (third trimester).

So many things can go wrong in the intrauterine environment that it is miraculous most babies are born with no apparent physical malformations. But behavioral teratogenesis is more subtle than physical defects and might not manifest until years later. Numerous studies have linked prenatal and obstetric complications to serious psychiatric disorders later in childhood or adulthood.

No wonder, then, that the pharmacologic management of mentally ill pregnant women is a high-stakes challenge. Consider the article in this issue by Louann Brizendine et al on treating anxiety during pregnancy with selective serotonin reuptake inhibitors vs benzodiazepines (page 38).

Risk vs benefit. Psychotropics can have unpredictable, serious effects on fetal growth and development, but fetal repercussions may be equally devastating if we do not stabilize the mentally ill mother and guard her against self-neglect, nonadherence to prenatal care, suicide, or infanticide. Thus, the benefit: risk ratio is sufficiently high to justify pharmacologic intervention during pregnancy—with the requisite caution this treatment deserves.

The greatest risk to the fetus is teratogenicity, which 1 review article1 defined as “the dysgenesis of fetal organs as evidenced either structurally or functionally (eg, brain functions). The typical manifestations of teratogenesis are restricted growth or death of the fetus, carcinogenesis, and malformations, defined as defects in organ structure or function. These abnormalities vary in severity (eg, hypospadias that is mild and may be missed, or is severe, necessitating several corrective operations). Major malformations may be life-threatening and require major surgery or may have serious cosmetic or functional effects.”

Because of teratogenicity concerns, pregnant women are excluded from clinical trials of investigational drugs. Thus, new drugs are not approved for use in these patients, and FDA rankings of drugs’ teratogenic potential (Table) are guided by nonblinded, noncontrolled, naturalistic, after-the-fact observations—the lowest tier of evidence-based medicine.

Table

FDA classification of medications’ teratogenic potential

 

CategoryExamples
A: Controlled studies in pregnant women demonstrate no fetal riskFolic acid, levothyroxine
B: Controlled animal studies have not shown a fetal risk, but there are no studies done on women OR controlled studies in animals have shown a fetal risk that was not reproduced in controlled human studiesAmoxicillin, ceftriaxone
C: Controlled animal studies have demonstrated adverse fetal effects and there are no human studies OR there are no controlled studies in humans or animalsNifedipine, omeprazole
D: Controlled studies in humans demonstrate adverse fetal effects but the benefits of using the drug may be greater than the risksPropylthiouracil
X: Controlled studies in animals and humans have demonstrated adverse fetal effects OR there is evidence of fetal risk based on human experience. The risk of using these drugs outweighs any possible benefit. The drug is absolutely contraindicated in pregnancyMisoprostol, warfarin, isotretinoin
Source: Adapted from Food and Drug Administration. Current categories for drug use in pregnancy (www.fda.gov/fdac/features/2001/301_preg.html)

Proceed with caution. Against this background, I follow these principles when treating pregnant patients:

 

  • Counsel all mentally ill women about the potential risks of conceiving while receiving a psychotropic before they consider pregnancy. Counseling should include all prescription and nonprescription drugs.
  • Obtain a family history of psychiatric disorders from all pregnant patients.
  • Make an accurate psychiatric diagnosis in pregnant patients, and assess the risks of providing vs withholding needed pharmacotherapy.
  • Use nondrug treatments (if evidence-based) before medications. Options include behavioral therapies, interpersonal therapy, supportive therapy, and somatic treatments such as electroconvulsive therapy, repetitive transcranial magnetic stimulation, and light therapy.
  • When using psychotropics, select the lowest-risk agents (Category A) first, and use the lowest efficacious dose.
  • Collaborate with the patient’s obstetrician. I coined the term “psychiatric dystocia” to describe the complicating potential of mental illness on pregnancy.
  • Completely avoid drugs with established teratogenicity, and educate the patient not to take these drugs if another physician prescribes them to her.
  • Prescribe high-dose folate (4 to 5 mg/d) for psychotic, bipolar, or depressed pregnant patients to protect against neural tube defects and enhance fetal CNS development.
  • Regularly check the patient’s nutrition, sleep hygiene, substance use (smoking, alcohol, coffee, illicit drugs), and use of over-the-counter supplements.
  • Use stress-reduction techniques to reduce potential deleterious effects of stress-induced hypercortisolemia on the fetus, and involve the patient’s partner.
  • See the mentally ill pregnant patient frequently for check-ups on response and/or side effects.
  • Arrange for a child psychiatrist to examine the infant of a seriously mentally ill patient shortly after birth. A newborn’s irritability, crankiness, or insomnia may be perceived as withdrawal symptoms or behavioral teratogenesis, whereas it could very well be a genetically inherited temperament instability from a mother suffering from anxiety, depression, or psychosis.
 

 

Helping the mother without harming the child is like walking a tightrope: it calls upon all our skills, experience, and sound judgment.

P.S. To help you manage potential medico-legal issues such as prescribing during pregnancy, Current Psychiatry welcomes Douglas Mossman, MD, as editor of Malpractice Rx. This month, Dr. Mossman discusses documentation and invites you to submit questions about liability.

References

 

1. Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Engl J Med 1998;338:1128-37.

Article PDF
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, contact Dr. Nasrallah at [email protected] or visit currentpsychiatry.com and click on the “Contact Us” link.

Issue
Current Psychiatry - 07(02)
Publications
Page Number
15-16
Legacy Keywords
Henry A. Nasrallah MD; Louann Brizendine; behavioral teratogenesis; mentally ill pregnant women; selective serotonin reuptake inhibitors; benzodiazepines; teratogenicity
Sections
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, contact Dr. Nasrallah at [email protected] or visit currentpsychiatry.com and click on the “Contact Us” link.

Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, contact Dr. Nasrallah at [email protected] or visit currentpsychiatry.com and click on the “Contact Us” link.

Article PDF
Article PDF

Life in the womb is fraught with hazards. Any deviation from a healthy pregnancy can damage a major organ system (first trimester), impair brain structural/functional development (second trimester), or cause prematurity and low birth weight (third trimester).

So many things can go wrong in the intrauterine environment that it is miraculous most babies are born with no apparent physical malformations. But behavioral teratogenesis is more subtle than physical defects and might not manifest until years later. Numerous studies have linked prenatal and obstetric complications to serious psychiatric disorders later in childhood or adulthood.

No wonder, then, that the pharmacologic management of mentally ill pregnant women is a high-stakes challenge. Consider the article in this issue by Louann Brizendine et al on treating anxiety during pregnancy with selective serotonin reuptake inhibitors vs benzodiazepines (page 38).

Risk vs benefit. Psychotropics can have unpredictable, serious effects on fetal growth and development, but fetal repercussions may be equally devastating if we do not stabilize the mentally ill mother and guard her against self-neglect, nonadherence to prenatal care, suicide, or infanticide. Thus, the benefit: risk ratio is sufficiently high to justify pharmacologic intervention during pregnancy—with the requisite caution this treatment deserves.

The greatest risk to the fetus is teratogenicity, which 1 review article1 defined as “the dysgenesis of fetal organs as evidenced either structurally or functionally (eg, brain functions). The typical manifestations of teratogenesis are restricted growth or death of the fetus, carcinogenesis, and malformations, defined as defects in organ structure or function. These abnormalities vary in severity (eg, hypospadias that is mild and may be missed, or is severe, necessitating several corrective operations). Major malformations may be life-threatening and require major surgery or may have serious cosmetic or functional effects.”

Because of teratogenicity concerns, pregnant women are excluded from clinical trials of investigational drugs. Thus, new drugs are not approved for use in these patients, and FDA rankings of drugs’ teratogenic potential (Table) are guided by nonblinded, noncontrolled, naturalistic, after-the-fact observations—the lowest tier of evidence-based medicine.

Table

FDA classification of medications’ teratogenic potential

 

CategoryExamples
A: Controlled studies in pregnant women demonstrate no fetal riskFolic acid, levothyroxine
B: Controlled animal studies have not shown a fetal risk, but there are no studies done on women OR controlled studies in animals have shown a fetal risk that was not reproduced in controlled human studiesAmoxicillin, ceftriaxone
C: Controlled animal studies have demonstrated adverse fetal effects and there are no human studies OR there are no controlled studies in humans or animalsNifedipine, omeprazole
D: Controlled studies in humans demonstrate adverse fetal effects but the benefits of using the drug may be greater than the risksPropylthiouracil
X: Controlled studies in animals and humans have demonstrated adverse fetal effects OR there is evidence of fetal risk based on human experience. The risk of using these drugs outweighs any possible benefit. The drug is absolutely contraindicated in pregnancyMisoprostol, warfarin, isotretinoin
Source: Adapted from Food and Drug Administration. Current categories for drug use in pregnancy (www.fda.gov/fdac/features/2001/301_preg.html)

Proceed with caution. Against this background, I follow these principles when treating pregnant patients:

 

  • Counsel all mentally ill women about the potential risks of conceiving while receiving a psychotropic before they consider pregnancy. Counseling should include all prescription and nonprescription drugs.
  • Obtain a family history of psychiatric disorders from all pregnant patients.
  • Make an accurate psychiatric diagnosis in pregnant patients, and assess the risks of providing vs withholding needed pharmacotherapy.
  • Use nondrug treatments (if evidence-based) before medications. Options include behavioral therapies, interpersonal therapy, supportive therapy, and somatic treatments such as electroconvulsive therapy, repetitive transcranial magnetic stimulation, and light therapy.
  • When using psychotropics, select the lowest-risk agents (Category A) first, and use the lowest efficacious dose.
  • Collaborate with the patient’s obstetrician. I coined the term “psychiatric dystocia” to describe the complicating potential of mental illness on pregnancy.
  • Completely avoid drugs with established teratogenicity, and educate the patient not to take these drugs if another physician prescribes them to her.
  • Prescribe high-dose folate (4 to 5 mg/d) for psychotic, bipolar, or depressed pregnant patients to protect against neural tube defects and enhance fetal CNS development.
  • Regularly check the patient’s nutrition, sleep hygiene, substance use (smoking, alcohol, coffee, illicit drugs), and use of over-the-counter supplements.
  • Use stress-reduction techniques to reduce potential deleterious effects of stress-induced hypercortisolemia on the fetus, and involve the patient’s partner.
  • See the mentally ill pregnant patient frequently for check-ups on response and/or side effects.
  • Arrange for a child psychiatrist to examine the infant of a seriously mentally ill patient shortly after birth. A newborn’s irritability, crankiness, or insomnia may be perceived as withdrawal symptoms or behavioral teratogenesis, whereas it could very well be a genetically inherited temperament instability from a mother suffering from anxiety, depression, or psychosis.
 

 

Helping the mother without harming the child is like walking a tightrope: it calls upon all our skills, experience, and sound judgment.

P.S. To help you manage potential medico-legal issues such as prescribing during pregnancy, Current Psychiatry welcomes Douglas Mossman, MD, as editor of Malpractice Rx. This month, Dr. Mossman discusses documentation and invites you to submit questions about liability.

Life in the womb is fraught with hazards. Any deviation from a healthy pregnancy can damage a major organ system (first trimester), impair brain structural/functional development (second trimester), or cause prematurity and low birth weight (third trimester).

So many things can go wrong in the intrauterine environment that it is miraculous most babies are born with no apparent physical malformations. But behavioral teratogenesis is more subtle than physical defects and might not manifest until years later. Numerous studies have linked prenatal and obstetric complications to serious psychiatric disorders later in childhood or adulthood.

No wonder, then, that the pharmacologic management of mentally ill pregnant women is a high-stakes challenge. Consider the article in this issue by Louann Brizendine et al on treating anxiety during pregnancy with selective serotonin reuptake inhibitors vs benzodiazepines (page 38).

Risk vs benefit. Psychotropics can have unpredictable, serious effects on fetal growth and development, but fetal repercussions may be equally devastating if we do not stabilize the mentally ill mother and guard her against self-neglect, nonadherence to prenatal care, suicide, or infanticide. Thus, the benefit: risk ratio is sufficiently high to justify pharmacologic intervention during pregnancy—with the requisite caution this treatment deserves.

The greatest risk to the fetus is teratogenicity, which 1 review article1 defined as “the dysgenesis of fetal organs as evidenced either structurally or functionally (eg, brain functions). The typical manifestations of teratogenesis are restricted growth or death of the fetus, carcinogenesis, and malformations, defined as defects in organ structure or function. These abnormalities vary in severity (eg, hypospadias that is mild and may be missed, or is severe, necessitating several corrective operations). Major malformations may be life-threatening and require major surgery or may have serious cosmetic or functional effects.”

Because of teratogenicity concerns, pregnant women are excluded from clinical trials of investigational drugs. Thus, new drugs are not approved for use in these patients, and FDA rankings of drugs’ teratogenic potential (Table) are guided by nonblinded, noncontrolled, naturalistic, after-the-fact observations—the lowest tier of evidence-based medicine.

Table

FDA classification of medications’ teratogenic potential

 

CategoryExamples
A: Controlled studies in pregnant women demonstrate no fetal riskFolic acid, levothyroxine
B: Controlled animal studies have not shown a fetal risk, but there are no studies done on women OR controlled studies in animals have shown a fetal risk that was not reproduced in controlled human studiesAmoxicillin, ceftriaxone
C: Controlled animal studies have demonstrated adverse fetal effects and there are no human studies OR there are no controlled studies in humans or animalsNifedipine, omeprazole
D: Controlled studies in humans demonstrate adverse fetal effects but the benefits of using the drug may be greater than the risksPropylthiouracil
X: Controlled studies in animals and humans have demonstrated adverse fetal effects OR there is evidence of fetal risk based on human experience. The risk of using these drugs outweighs any possible benefit. The drug is absolutely contraindicated in pregnancyMisoprostol, warfarin, isotretinoin
Source: Adapted from Food and Drug Administration. Current categories for drug use in pregnancy (www.fda.gov/fdac/features/2001/301_preg.html)

Proceed with caution. Against this background, I follow these principles when treating pregnant patients:

 

  • Counsel all mentally ill women about the potential risks of conceiving while receiving a psychotropic before they consider pregnancy. Counseling should include all prescription and nonprescription drugs.
  • Obtain a family history of psychiatric disorders from all pregnant patients.
  • Make an accurate psychiatric diagnosis in pregnant patients, and assess the risks of providing vs withholding needed pharmacotherapy.
  • Use nondrug treatments (if evidence-based) before medications. Options include behavioral therapies, interpersonal therapy, supportive therapy, and somatic treatments such as electroconvulsive therapy, repetitive transcranial magnetic stimulation, and light therapy.
  • When using psychotropics, select the lowest-risk agents (Category A) first, and use the lowest efficacious dose.
  • Collaborate with the patient’s obstetrician. I coined the term “psychiatric dystocia” to describe the complicating potential of mental illness on pregnancy.
  • Completely avoid drugs with established teratogenicity, and educate the patient not to take these drugs if another physician prescribes them to her.
  • Prescribe high-dose folate (4 to 5 mg/d) for psychotic, bipolar, or depressed pregnant patients to protect against neural tube defects and enhance fetal CNS development.
  • Regularly check the patient’s nutrition, sleep hygiene, substance use (smoking, alcohol, coffee, illicit drugs), and use of over-the-counter supplements.
  • Use stress-reduction techniques to reduce potential deleterious effects of stress-induced hypercortisolemia on the fetus, and involve the patient’s partner.
  • See the mentally ill pregnant patient frequently for check-ups on response and/or side effects.
  • Arrange for a child psychiatrist to examine the infant of a seriously mentally ill patient shortly after birth. A newborn’s irritability, crankiness, or insomnia may be perceived as withdrawal symptoms or behavioral teratogenesis, whereas it could very well be a genetically inherited temperament instability from a mother suffering from anxiety, depression, or psychosis.
 

 

Helping the mother without harming the child is like walking a tightrope: it calls upon all our skills, experience, and sound judgment.

P.S. To help you manage potential medico-legal issues such as prescribing during pregnancy, Current Psychiatry welcomes Douglas Mossman, MD, as editor of Malpractice Rx. This month, Dr. Mossman discusses documentation and invites you to submit questions about liability.

References

 

1. Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Engl J Med 1998;338:1128-37.

References

 

1. Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Engl J Med 1998;338:1128-37.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
15-16
Page Number
15-16
Publications
Publications
Article Type
Display Headline
Pregnant and mentally ill: A labor-intensive clinical challenge
Display Headline
Pregnant and mentally ill: A labor-intensive clinical challenge
Legacy Keywords
Henry A. Nasrallah MD; Louann Brizendine; behavioral teratogenesis; mentally ill pregnant women; selective serotonin reuptake inhibitors; benzodiazepines; teratogenicity
Legacy Keywords
Henry A. Nasrallah MD; Louann Brizendine; behavioral teratogenesis; mentally ill pregnant women; selective serotonin reuptake inhibitors; benzodiazepines; teratogenicity
Sections
Article PDF Media

One patient’s ‘shot’ at redemption

Article Type
Changed
Tue, 12/11/2018 - 15:30
Display Headline
One patient’s ‘shot’ at redemption

CASE: A ‘purification’

Mr. B, age 61, is in the ICU after shooting himself in the abdomen. The trauma team asks our psychiatry consultation/liaison service to determine if he needs special observation to prevent further self-harm.

Two days ago, Mr. B stood in the parking lot of a nearby hospital, aimed his rifle toward the left upper part of his abdomen, and fired. Bleeding profusely, he dragged himself to the hospital’s emergency room. ER staff stabilized him hemodynamically, then transferred him to our hospital’s regional trauma center, where surgeons performed an emergency laparotomy and found 2 sigmoid colon perforations, with feces floating outside the bowel.

After a partial colectomy and colostomy, Mr. B received broad-spectrum antibiotics, narcotic pain medication, and bowel rest in the ICU. When the trauma team called us, the patient’s condition was stable and he had awakened enough to communicate, although he still needed frequent monitoring.

We visit Mr. B in the ICU and ask him why he shot himself. He denies he was attempting suicide but adds that for months he has been feeling depressed, stressed, and guilty about “all the bad things I’ve done in my life.” Shooting himself helped him forget these negative thoughts.

A devout Roman Catholic, Mr. B has been researching flagellation and other forms of physical penance and considers the shooting a purification. He says he shot himself in the abdomen 2 previous times and felt better for months or years after each shooting.

Four years ago, Mr. B donated his left kidney to an unknown recipient. He does not equate the kidney donation with the shootings but says he felt happy while recuperating. He was later disappointed, however, because the procedure did not help him attract the “attention” he had hoped for.

Mr. B says he had been considering the latest shooting for at least 8 months and had carefully planned it. After studying anatomy textbooks, he figured he could fire into the left upper portion of his abdomen without striking a vital organ.

For several evenings, Mr. B aimed his rifle toward his abdomen but could not bring himself to pull the trigger. On the night of the shooting, he said, he “accidentally” fired at a more damaging angle than he had planned.

Cognitive examination results are mostly normal, although Mr. B has trouble interpreting similarities and proverbs. He appears pale but well-nourished, well-groomed, and serene. He speaks softly, often closing his eyes or staring into the distance. He says he feels “relieved” and “happy” after the shooting but did not anticipate such a severe injury. He denies suicidal thoughts and—because of his current euphoric mood—he hopes he never “needs” to shoot himself again.

The authors’ observations

We first considered delusional disorder and major depressive disorder with psychotic features. Mr. B’s belief that shooting himself would solve his problems seemed delusional, although he did not appear psychotic otherwise. Confusingly, Mr. B’s pre-admission symptoms seemed to suggest major depressive disorder, but he was happy in the ICU.

We explored other diagnoses, such as an odd form of OCD and a personality disorder (especially cluster A, given his strange beliefs), though at this point we had too little information for either diagnosis.

We also wondered if Mr. B’s behavior was normal given his strong belief in Catholic penance. Although some sects of the Catholic Church practice self-flagellation and other forms of self-punishment,1-3 we found no evidence that the church condones or encourages self-shooting. Moreover, Mr. B admitted during questioning that a Catholic clergy member told him shooting oneself is not an appropriate penance.

The authors’ observations

Mr. B was recovering from major abdominal surgery, was taking nothing orally, and claimed to feel fine psychologically. Because he was not grossly psychotic and did not endorse anxiety or depression, we decided against medication but recommended a chaplain consult and planned to visit Mr. B daily to gather more history.

We considered Mr. B a low suicide risk—especially while hospitalized—after he said his “need” to shoot himself had dissipated. He also endorsed no suicidal thoughts or other depressive symptoms, and the nursing staff viewed him as pleasant and compliant. We noted this evidence in the chart and continued to reassess him daily.

HISTORY: Dreams, nightmares

Over the next week, Mr. B shares his life story. He says his parents divorced when he was age 5, and around that time he spent approximately 2 weeks in the hospital after being hit by a truck. He considers those 2 weeks a bright spot in an otherwise turbulent childhood because his parents did not fight and he was showered with gifts and attention.

 

 

Soon after, Mr. B lived with his mother. When he was 9, he heard his divorced parents arguing during a family gathering and prayed for his own death.

Throughout his childhood, Mr. B dreamed of becoming a priest and a war hero. In his early teens, he attended a church youth program where he and other youths were taught that masturbation is a mortal sin. Through high school, Mr. B’s inability to stop masturbating shook his faith and discouraged him from pursuing the priesthood.

In high school, Mr. B did not use alcohol or drugs and excelled academically but had few friends. After graduating, he enlisted in the U.S. Army and hoped to serve in Vietnam, but basic training became too stressful. The relentless harangues from drill instructors reminded him of his parents’ frequent shouting matches during his childhood.

Approximately 2 weeks into basic training, Mr. B shot himself in the abdomen and injured his liver. He underwent laparotomy and cholecystectomy and was discharged from the military. His anxiety dissipated as he recovered, though he later regretted not serving in Vietnam.

Mr. B married at age 44 and was divorced 13 years later. Throughout the marriage, he says, he verbally abused his wife and was emotionally unsupportive. After the divorce, he felt remorse over having mistreated her. His guilt disappeared after he donated his left kidney to an unknown recipient, but the guilt soon returned and drove him to shoot himself a second time in 2005.

Mr. B worked as a special education teacher for 20 years before retiring 4 years ago and has since been pursuing a similar position because he misses going to work. His inability to find a permanent job has led to anxiety, insomnia, increased guilt, and decreased appetite. He says these feelings fueled his desire to shoot himself a third time.

Since his divorce, Mr. B has lived alone and has no family or friends nearby. He feels isolated and is hurt that his family has not acknowledged the cards and notes he sent to them but adds that he did not include his return address on the mailings.

FOLLOW-UP: No relief

After 1 week in the ICU, Mr. B is transferred to the surgical ward. His condition is fair and he continues to receive IV antibiotics; analgesics as needed; omeprazole, 20 mg/d, to prevent a stress ulcer; bowel care medications; and ostomy care and education. We continue to visit him in the surgical ward almost daily. He seems to enjoy our visits, during which he openly discusses his past.

Eleven days after admission, Mr. B says shooting himself has not relieved his negative feelings, and his impending discharge makes him feel anxious with some suicidal thoughts. The surgical team delays discharge after Mr. B develops ileus with nausea and vomiting. The trauma team’s attending physician prescribes an antiemetic, and ileus resolves after 4 days. Mr. B then is discharged in stable condition after he denies intention to harm himself.

Box 1

DSM-IV-TR criteria for delusional disorder

  1. Nonbizarre delusions (involving situations that occur in real life) lasting ≥1 month.
  2. Criterion A for schizophrenia has never been met. (Note: Tactile and olfactory hallucinations may be present in delusional disorder if they are related to the delusional disorder.)
  3. Apart from the impact or ramifications of the delusion(s), functioning is not markedly impaired and behavior is not obviously odd or bizarre.
  4. If mood episodes have occurred concurrently with delusions, their total duration has been brief relative to the duration of the delusional periods.
  5. The disturbance is not due to the direct physiologic effects of a substance or a general medical condition.

Source:Reference 6. Reprinted with permission

The authors’ observations

At first we viewed Mr. B’s idea of shooting himself to solve his problems as a delusion (Box 1), but less than 2 weeks later he denied that his self-injury offered any benefit. Because his original belief was transient, we ruled out delusional disorder and major depression with psychotic features.

Some of Mr. B’s symptoms suggested OCD, including thought-related anxiety that is relieved after performing an action—in his case shooting himself. Whereas obsessions and/or compulsions occur daily in OCD, Mr. B would perform the action and then feel fine for months to years before his anxiety resurfaced. Also, he did not consider his thoughts wrong or obtrusive.

 

 

Box 2

DSM-IV-TR criteria for factitious disorder

  1. Intentional production or feigning of physical or psychological signs or symptoms.
  2. The motivation for the behavior is to assume the sick role.
  3. External incentives for the behavior are absent.

Source:Reference 6. Reprinted with permission

Although Mr. B experienced some depressive symptoms and anxiety just before discharge, we were hesitant to diagnose major depression because his symptoms appeared tied to situational factors. He also did not fit a particular personality disorder, although he showed characteristics of:

  • cluster A (odd ideas, solitary lifestyle)
  • cluster B (self-harm, narcissistic tendencies)
  • and cluster C (avoiding his relatives, dependence on others to meet his needs).
We could have diagnosed personality disorder, not otherwise specified, but we were unsure whether personality explained his pathology or if his personality characteristics warranted diagnosis.

Mr. B’s intentional production of physical symptoms strongly suggested malingering, but we instead diagnosed factitious disorder because he was clearly motivated to play the sick role despite lack of a secondary gain (Box 2). The patient admitted causing the gunshot wound and clearly connected his subsequent emotional relief with both his positive childhood experience in the hospital and his satisfaction after donating a kidney.

Researchers have tried to distinguish between factitious disorder and other types of self-harm. Claes and Vandereycken4 would consider Mr. B’s behavior “self-mutilation” rather than factitious. Turner calls DSM-IV-TR criteria for factitious disorder nebulous and says that lying about symptoms or their origin should be a necessary criterion.5 If so, then Mr. B’s condition might fit no DSM diagnosis.6

The authors’ observations

Although Mr. B’s diagnosis remained elusive, he needed a treatment plan before discharge to prevent another shooting and save his life.

We first considered psychotropics. Because Mr. B’s beliefs did not appear delusional, an antipsychotic would not be a useful first-line treatment. Nor would a benzodiazepine help Mr. B at this point, especially since we did not diagnose primary anxiety.

Although we did not diagnose a major depressive or anxiety-spectrum disorder, we felt an SSRI such as citalopram could help. According to some investigators, SSRIs might benefit patients with over-valued ideas that are not as persistent as delusions.7,8

Additionally, we felt supportive therapy could help Mr. B establish a therapeutic relationship with a provider to whom he could turn during future crises. Should Mr. B contemplate self-harm, the therapist could suggest medications, hospitalization, or other interventions. We also recommended pastoral counseling to increase his support within his faith.

OUTCOME Another shot?

Before discharge, we start citalopram, 10 mg/d, and schedule a follow-up appointment within 2 weeks. We also suggest that Mr. B:

  • move into an apartment near his outpatient doctors
  • get involved with the local Catholic community to build his support network.
When we contact Mr. B 2 months later, he says he discontinued citalopram because he felt no benefit from it. At his initial appointment with a psychiatrist, he denied depressive symptoms and was not scheduled for ongoing therapy. He has not spoken with clergy or other local church members because “I know they would say God has forgiven me.”

Mr. B calls his recent hospitalization upsetting because “I did not get the attention I wanted.” He endorses no immediate plan to shoot himself but voices concern that when his physical problems resolve, he might shoot Himself in the liver—as he had done 40 years ago—to bring himself full circle. “There’s still something attractive about this,” he says.

The authors’ observations

Patients with factitious illness commonly refuse mental health treatment.9

We feel Mr. B needs frequent ongoing appointments in a medical clinic where doctors can provide sufficient attention to counter his persistent self-harm urges. Regular appointments with a primary care physician—regardless of whether Mr. B is medically ill—could help him feel supported.

Related resources

  • Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996.
  • Sutton J, Martinson D. Self-injury Web site: What self-injury is.www.palace.net/~llama/psych/fwhat.html.
Drug brand names

  • Citalopram • Celexa
  • Omeprazole • Prilosec
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. The mysteries of Opus Dei. US News and World Report; December 14, 2003 (health section). Available at: http://www.usnews.com/usnews/culture/articles/031222/22jesus.b.htm. Accessed December 10, 2007.

2. Toke LA. transcribed by Potter DJ Flagellants. In: Catholic encyclopedia, vol. 6. New York: Robert Appleton Co.; 1909. Available at: http://www.newadvent.org/cathen/06089c.htm. Accessed December 10, 2007.

3. Glucklich A. Sacred pain: hurting the body for the sake of the soul. New York: Oxford University Press; 2001.

4. Claes L, Vandereycken W. Self-injurious behavior: differential diagnosis and functional differentiation. Compr Psychiatry 2007;48:137-44.

5. Turner MA. Factitious disorders: reformulating the DSM-IV criteria. Psychosomatics 2006;47:23-32.

6. Diagnostic and statistical manual of mental disorders. 4th ed. text rev. Washington, DC: American Psychiatric Association; 2000.

7. Veale D. Over-valued ideas: a conceptual analysis. Behav Res Ther 2002;40:383-400.

8. Jones E, Watson JP. Delusion, the overvalued idea and religious beliefs: a comparative analysis of their characteristics. Br J Psychiatry 1997;170:381-6.

9. Sutherland AJ, Rodin GM. Factitious disorders in a general hospital setting: clinical features and a review of the literature. Psychosomatics 1990;31:392-9.

Article PDF
Author and Disclosure Information

Willow Naimark, MD
Jeanne Johnson, DDS
Annette Matthews, MD
Dr. Naimark is a second-year psychiatry resident, Ms. Johnson is a medical student, and Dr. Matthews is assistant professor of psychiatry, Oregon Health and Science University, Portland. Dr. Matthews also is staff psychiatrist, Portland VA Medical Center.

Issue
Current Psychiatry - 07(02)
Publications
Page Number
91-97
Legacy Keywords
Willow Naimark MD; Jeanne Johnson DDS; Annette Matthews MD; self-injury; delusional disorder; depression; major depression with psychotic features; major depression without psychotic features; obsessive-compulsive disorder; OCD; personality disorder; factitious disorder; malingering; supportive therapy; insight-oriented therapy
Sections
Author and Disclosure Information

Willow Naimark, MD
Jeanne Johnson, DDS
Annette Matthews, MD
Dr. Naimark is a second-year psychiatry resident, Ms. Johnson is a medical student, and Dr. Matthews is assistant professor of psychiatry, Oregon Health and Science University, Portland. Dr. Matthews also is staff psychiatrist, Portland VA Medical Center.

Author and Disclosure Information

Willow Naimark, MD
Jeanne Johnson, DDS
Annette Matthews, MD
Dr. Naimark is a second-year psychiatry resident, Ms. Johnson is a medical student, and Dr. Matthews is assistant professor of psychiatry, Oregon Health and Science University, Portland. Dr. Matthews also is staff psychiatrist, Portland VA Medical Center.

Article PDF
Article PDF

CASE: A ‘purification’

Mr. B, age 61, is in the ICU after shooting himself in the abdomen. The trauma team asks our psychiatry consultation/liaison service to determine if he needs special observation to prevent further self-harm.

Two days ago, Mr. B stood in the parking lot of a nearby hospital, aimed his rifle toward the left upper part of his abdomen, and fired. Bleeding profusely, he dragged himself to the hospital’s emergency room. ER staff stabilized him hemodynamically, then transferred him to our hospital’s regional trauma center, where surgeons performed an emergency laparotomy and found 2 sigmoid colon perforations, with feces floating outside the bowel.

After a partial colectomy and colostomy, Mr. B received broad-spectrum antibiotics, narcotic pain medication, and bowel rest in the ICU. When the trauma team called us, the patient’s condition was stable and he had awakened enough to communicate, although he still needed frequent monitoring.

We visit Mr. B in the ICU and ask him why he shot himself. He denies he was attempting suicide but adds that for months he has been feeling depressed, stressed, and guilty about “all the bad things I’ve done in my life.” Shooting himself helped him forget these negative thoughts.

A devout Roman Catholic, Mr. B has been researching flagellation and other forms of physical penance and considers the shooting a purification. He says he shot himself in the abdomen 2 previous times and felt better for months or years after each shooting.

Four years ago, Mr. B donated his left kidney to an unknown recipient. He does not equate the kidney donation with the shootings but says he felt happy while recuperating. He was later disappointed, however, because the procedure did not help him attract the “attention” he had hoped for.

Mr. B says he had been considering the latest shooting for at least 8 months and had carefully planned it. After studying anatomy textbooks, he figured he could fire into the left upper portion of his abdomen without striking a vital organ.

For several evenings, Mr. B aimed his rifle toward his abdomen but could not bring himself to pull the trigger. On the night of the shooting, he said, he “accidentally” fired at a more damaging angle than he had planned.

Cognitive examination results are mostly normal, although Mr. B has trouble interpreting similarities and proverbs. He appears pale but well-nourished, well-groomed, and serene. He speaks softly, often closing his eyes or staring into the distance. He says he feels “relieved” and “happy” after the shooting but did not anticipate such a severe injury. He denies suicidal thoughts and—because of his current euphoric mood—he hopes he never “needs” to shoot himself again.

The authors’ observations

We first considered delusional disorder and major depressive disorder with psychotic features. Mr. B’s belief that shooting himself would solve his problems seemed delusional, although he did not appear psychotic otherwise. Confusingly, Mr. B’s pre-admission symptoms seemed to suggest major depressive disorder, but he was happy in the ICU.

We explored other diagnoses, such as an odd form of OCD and a personality disorder (especially cluster A, given his strange beliefs), though at this point we had too little information for either diagnosis.

We also wondered if Mr. B’s behavior was normal given his strong belief in Catholic penance. Although some sects of the Catholic Church practice self-flagellation and other forms of self-punishment,1-3 we found no evidence that the church condones or encourages self-shooting. Moreover, Mr. B admitted during questioning that a Catholic clergy member told him shooting oneself is not an appropriate penance.

The authors’ observations

Mr. B was recovering from major abdominal surgery, was taking nothing orally, and claimed to feel fine psychologically. Because he was not grossly psychotic and did not endorse anxiety or depression, we decided against medication but recommended a chaplain consult and planned to visit Mr. B daily to gather more history.

We considered Mr. B a low suicide risk—especially while hospitalized—after he said his “need” to shoot himself had dissipated. He also endorsed no suicidal thoughts or other depressive symptoms, and the nursing staff viewed him as pleasant and compliant. We noted this evidence in the chart and continued to reassess him daily.

HISTORY: Dreams, nightmares

Over the next week, Mr. B shares his life story. He says his parents divorced when he was age 5, and around that time he spent approximately 2 weeks in the hospital after being hit by a truck. He considers those 2 weeks a bright spot in an otherwise turbulent childhood because his parents did not fight and he was showered with gifts and attention.

 

 

Soon after, Mr. B lived with his mother. When he was 9, he heard his divorced parents arguing during a family gathering and prayed for his own death.

Throughout his childhood, Mr. B dreamed of becoming a priest and a war hero. In his early teens, he attended a church youth program where he and other youths were taught that masturbation is a mortal sin. Through high school, Mr. B’s inability to stop masturbating shook his faith and discouraged him from pursuing the priesthood.

In high school, Mr. B did not use alcohol or drugs and excelled academically but had few friends. After graduating, he enlisted in the U.S. Army and hoped to serve in Vietnam, but basic training became too stressful. The relentless harangues from drill instructors reminded him of his parents’ frequent shouting matches during his childhood.

Approximately 2 weeks into basic training, Mr. B shot himself in the abdomen and injured his liver. He underwent laparotomy and cholecystectomy and was discharged from the military. His anxiety dissipated as he recovered, though he later regretted not serving in Vietnam.

Mr. B married at age 44 and was divorced 13 years later. Throughout the marriage, he says, he verbally abused his wife and was emotionally unsupportive. After the divorce, he felt remorse over having mistreated her. His guilt disappeared after he donated his left kidney to an unknown recipient, but the guilt soon returned and drove him to shoot himself a second time in 2005.

Mr. B worked as a special education teacher for 20 years before retiring 4 years ago and has since been pursuing a similar position because he misses going to work. His inability to find a permanent job has led to anxiety, insomnia, increased guilt, and decreased appetite. He says these feelings fueled his desire to shoot himself a third time.

Since his divorce, Mr. B has lived alone and has no family or friends nearby. He feels isolated and is hurt that his family has not acknowledged the cards and notes he sent to them but adds that he did not include his return address on the mailings.

FOLLOW-UP: No relief

After 1 week in the ICU, Mr. B is transferred to the surgical ward. His condition is fair and he continues to receive IV antibiotics; analgesics as needed; omeprazole, 20 mg/d, to prevent a stress ulcer; bowel care medications; and ostomy care and education. We continue to visit him in the surgical ward almost daily. He seems to enjoy our visits, during which he openly discusses his past.

Eleven days after admission, Mr. B says shooting himself has not relieved his negative feelings, and his impending discharge makes him feel anxious with some suicidal thoughts. The surgical team delays discharge after Mr. B develops ileus with nausea and vomiting. The trauma team’s attending physician prescribes an antiemetic, and ileus resolves after 4 days. Mr. B then is discharged in stable condition after he denies intention to harm himself.

Box 1

DSM-IV-TR criteria for delusional disorder

  1. Nonbizarre delusions (involving situations that occur in real life) lasting ≥1 month.
  2. Criterion A for schizophrenia has never been met. (Note: Tactile and olfactory hallucinations may be present in delusional disorder if they are related to the delusional disorder.)
  3. Apart from the impact or ramifications of the delusion(s), functioning is not markedly impaired and behavior is not obviously odd or bizarre.
  4. If mood episodes have occurred concurrently with delusions, their total duration has been brief relative to the duration of the delusional periods.
  5. The disturbance is not due to the direct physiologic effects of a substance or a general medical condition.

Source:Reference 6. Reprinted with permission

The authors’ observations

At first we viewed Mr. B’s idea of shooting himself to solve his problems as a delusion (Box 1), but less than 2 weeks later he denied that his self-injury offered any benefit. Because his original belief was transient, we ruled out delusional disorder and major depression with psychotic features.

Some of Mr. B’s symptoms suggested OCD, including thought-related anxiety that is relieved after performing an action—in his case shooting himself. Whereas obsessions and/or compulsions occur daily in OCD, Mr. B would perform the action and then feel fine for months to years before his anxiety resurfaced. Also, he did not consider his thoughts wrong or obtrusive.

 

 

Box 2

DSM-IV-TR criteria for factitious disorder

  1. Intentional production or feigning of physical or psychological signs or symptoms.
  2. The motivation for the behavior is to assume the sick role.
  3. External incentives for the behavior are absent.

Source:Reference 6. Reprinted with permission

Although Mr. B experienced some depressive symptoms and anxiety just before discharge, we were hesitant to diagnose major depression because his symptoms appeared tied to situational factors. He also did not fit a particular personality disorder, although he showed characteristics of:

  • cluster A (odd ideas, solitary lifestyle)
  • cluster B (self-harm, narcissistic tendencies)
  • and cluster C (avoiding his relatives, dependence on others to meet his needs).
We could have diagnosed personality disorder, not otherwise specified, but we were unsure whether personality explained his pathology or if his personality characteristics warranted diagnosis.

Mr. B’s intentional production of physical symptoms strongly suggested malingering, but we instead diagnosed factitious disorder because he was clearly motivated to play the sick role despite lack of a secondary gain (Box 2). The patient admitted causing the gunshot wound and clearly connected his subsequent emotional relief with both his positive childhood experience in the hospital and his satisfaction after donating a kidney.

Researchers have tried to distinguish between factitious disorder and other types of self-harm. Claes and Vandereycken4 would consider Mr. B’s behavior “self-mutilation” rather than factitious. Turner calls DSM-IV-TR criteria for factitious disorder nebulous and says that lying about symptoms or their origin should be a necessary criterion.5 If so, then Mr. B’s condition might fit no DSM diagnosis.6

The authors’ observations

Although Mr. B’s diagnosis remained elusive, he needed a treatment plan before discharge to prevent another shooting and save his life.

We first considered psychotropics. Because Mr. B’s beliefs did not appear delusional, an antipsychotic would not be a useful first-line treatment. Nor would a benzodiazepine help Mr. B at this point, especially since we did not diagnose primary anxiety.

Although we did not diagnose a major depressive or anxiety-spectrum disorder, we felt an SSRI such as citalopram could help. According to some investigators, SSRIs might benefit patients with over-valued ideas that are not as persistent as delusions.7,8

Additionally, we felt supportive therapy could help Mr. B establish a therapeutic relationship with a provider to whom he could turn during future crises. Should Mr. B contemplate self-harm, the therapist could suggest medications, hospitalization, or other interventions. We also recommended pastoral counseling to increase his support within his faith.

OUTCOME Another shot?

Before discharge, we start citalopram, 10 mg/d, and schedule a follow-up appointment within 2 weeks. We also suggest that Mr. B:

  • move into an apartment near his outpatient doctors
  • get involved with the local Catholic community to build his support network.
When we contact Mr. B 2 months later, he says he discontinued citalopram because he felt no benefit from it. At his initial appointment with a psychiatrist, he denied depressive symptoms and was not scheduled for ongoing therapy. He has not spoken with clergy or other local church members because “I know they would say God has forgiven me.”

Mr. B calls his recent hospitalization upsetting because “I did not get the attention I wanted.” He endorses no immediate plan to shoot himself but voices concern that when his physical problems resolve, he might shoot Himself in the liver—as he had done 40 years ago—to bring himself full circle. “There’s still something attractive about this,” he says.

The authors’ observations

Patients with factitious illness commonly refuse mental health treatment.9

We feel Mr. B needs frequent ongoing appointments in a medical clinic where doctors can provide sufficient attention to counter his persistent self-harm urges. Regular appointments with a primary care physician—regardless of whether Mr. B is medically ill—could help him feel supported.

Related resources

  • Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996.
  • Sutton J, Martinson D. Self-injury Web site: What self-injury is.www.palace.net/~llama/psych/fwhat.html.
Drug brand names

  • Citalopram • Celexa
  • Omeprazole • Prilosec
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

CASE: A ‘purification’

Mr. B, age 61, is in the ICU after shooting himself in the abdomen. The trauma team asks our psychiatry consultation/liaison service to determine if he needs special observation to prevent further self-harm.

Two days ago, Mr. B stood in the parking lot of a nearby hospital, aimed his rifle toward the left upper part of his abdomen, and fired. Bleeding profusely, he dragged himself to the hospital’s emergency room. ER staff stabilized him hemodynamically, then transferred him to our hospital’s regional trauma center, where surgeons performed an emergency laparotomy and found 2 sigmoid colon perforations, with feces floating outside the bowel.

After a partial colectomy and colostomy, Mr. B received broad-spectrum antibiotics, narcotic pain medication, and bowel rest in the ICU. When the trauma team called us, the patient’s condition was stable and he had awakened enough to communicate, although he still needed frequent monitoring.

We visit Mr. B in the ICU and ask him why he shot himself. He denies he was attempting suicide but adds that for months he has been feeling depressed, stressed, and guilty about “all the bad things I’ve done in my life.” Shooting himself helped him forget these negative thoughts.

A devout Roman Catholic, Mr. B has been researching flagellation and other forms of physical penance and considers the shooting a purification. He says he shot himself in the abdomen 2 previous times and felt better for months or years after each shooting.

Four years ago, Mr. B donated his left kidney to an unknown recipient. He does not equate the kidney donation with the shootings but says he felt happy while recuperating. He was later disappointed, however, because the procedure did not help him attract the “attention” he had hoped for.

Mr. B says he had been considering the latest shooting for at least 8 months and had carefully planned it. After studying anatomy textbooks, he figured he could fire into the left upper portion of his abdomen without striking a vital organ.

For several evenings, Mr. B aimed his rifle toward his abdomen but could not bring himself to pull the trigger. On the night of the shooting, he said, he “accidentally” fired at a more damaging angle than he had planned.

Cognitive examination results are mostly normal, although Mr. B has trouble interpreting similarities and proverbs. He appears pale but well-nourished, well-groomed, and serene. He speaks softly, often closing his eyes or staring into the distance. He says he feels “relieved” and “happy” after the shooting but did not anticipate such a severe injury. He denies suicidal thoughts and—because of his current euphoric mood—he hopes he never “needs” to shoot himself again.

The authors’ observations

We first considered delusional disorder and major depressive disorder with psychotic features. Mr. B’s belief that shooting himself would solve his problems seemed delusional, although he did not appear psychotic otherwise. Confusingly, Mr. B’s pre-admission symptoms seemed to suggest major depressive disorder, but he was happy in the ICU.

We explored other diagnoses, such as an odd form of OCD and a personality disorder (especially cluster A, given his strange beliefs), though at this point we had too little information for either diagnosis.

We also wondered if Mr. B’s behavior was normal given his strong belief in Catholic penance. Although some sects of the Catholic Church practice self-flagellation and other forms of self-punishment,1-3 we found no evidence that the church condones or encourages self-shooting. Moreover, Mr. B admitted during questioning that a Catholic clergy member told him shooting oneself is not an appropriate penance.

The authors’ observations

Mr. B was recovering from major abdominal surgery, was taking nothing orally, and claimed to feel fine psychologically. Because he was not grossly psychotic and did not endorse anxiety or depression, we decided against medication but recommended a chaplain consult and planned to visit Mr. B daily to gather more history.

We considered Mr. B a low suicide risk—especially while hospitalized—after he said his “need” to shoot himself had dissipated. He also endorsed no suicidal thoughts or other depressive symptoms, and the nursing staff viewed him as pleasant and compliant. We noted this evidence in the chart and continued to reassess him daily.

HISTORY: Dreams, nightmares

Over the next week, Mr. B shares his life story. He says his parents divorced when he was age 5, and around that time he spent approximately 2 weeks in the hospital after being hit by a truck. He considers those 2 weeks a bright spot in an otherwise turbulent childhood because his parents did not fight and he was showered with gifts and attention.

 

 

Soon after, Mr. B lived with his mother. When he was 9, he heard his divorced parents arguing during a family gathering and prayed for his own death.

Throughout his childhood, Mr. B dreamed of becoming a priest and a war hero. In his early teens, he attended a church youth program where he and other youths were taught that masturbation is a mortal sin. Through high school, Mr. B’s inability to stop masturbating shook his faith and discouraged him from pursuing the priesthood.

In high school, Mr. B did not use alcohol or drugs and excelled academically but had few friends. After graduating, he enlisted in the U.S. Army and hoped to serve in Vietnam, but basic training became too stressful. The relentless harangues from drill instructors reminded him of his parents’ frequent shouting matches during his childhood.

Approximately 2 weeks into basic training, Mr. B shot himself in the abdomen and injured his liver. He underwent laparotomy and cholecystectomy and was discharged from the military. His anxiety dissipated as he recovered, though he later regretted not serving in Vietnam.

Mr. B married at age 44 and was divorced 13 years later. Throughout the marriage, he says, he verbally abused his wife and was emotionally unsupportive. After the divorce, he felt remorse over having mistreated her. His guilt disappeared after he donated his left kidney to an unknown recipient, but the guilt soon returned and drove him to shoot himself a second time in 2005.

Mr. B worked as a special education teacher for 20 years before retiring 4 years ago and has since been pursuing a similar position because he misses going to work. His inability to find a permanent job has led to anxiety, insomnia, increased guilt, and decreased appetite. He says these feelings fueled his desire to shoot himself a third time.

Since his divorce, Mr. B has lived alone and has no family or friends nearby. He feels isolated and is hurt that his family has not acknowledged the cards and notes he sent to them but adds that he did not include his return address on the mailings.

FOLLOW-UP: No relief

After 1 week in the ICU, Mr. B is transferred to the surgical ward. His condition is fair and he continues to receive IV antibiotics; analgesics as needed; omeprazole, 20 mg/d, to prevent a stress ulcer; bowel care medications; and ostomy care and education. We continue to visit him in the surgical ward almost daily. He seems to enjoy our visits, during which he openly discusses his past.

Eleven days after admission, Mr. B says shooting himself has not relieved his negative feelings, and his impending discharge makes him feel anxious with some suicidal thoughts. The surgical team delays discharge after Mr. B develops ileus with nausea and vomiting. The trauma team’s attending physician prescribes an antiemetic, and ileus resolves after 4 days. Mr. B then is discharged in stable condition after he denies intention to harm himself.

Box 1

DSM-IV-TR criteria for delusional disorder

  1. Nonbizarre delusions (involving situations that occur in real life) lasting ≥1 month.
  2. Criterion A for schizophrenia has never been met. (Note: Tactile and olfactory hallucinations may be present in delusional disorder if they are related to the delusional disorder.)
  3. Apart from the impact or ramifications of the delusion(s), functioning is not markedly impaired and behavior is not obviously odd or bizarre.
  4. If mood episodes have occurred concurrently with delusions, their total duration has been brief relative to the duration of the delusional periods.
  5. The disturbance is not due to the direct physiologic effects of a substance or a general medical condition.

Source:Reference 6. Reprinted with permission

The authors’ observations

At first we viewed Mr. B’s idea of shooting himself to solve his problems as a delusion (Box 1), but less than 2 weeks later he denied that his self-injury offered any benefit. Because his original belief was transient, we ruled out delusional disorder and major depression with psychotic features.

Some of Mr. B’s symptoms suggested OCD, including thought-related anxiety that is relieved after performing an action—in his case shooting himself. Whereas obsessions and/or compulsions occur daily in OCD, Mr. B would perform the action and then feel fine for months to years before his anxiety resurfaced. Also, he did not consider his thoughts wrong or obtrusive.

 

 

Box 2

DSM-IV-TR criteria for factitious disorder

  1. Intentional production or feigning of physical or psychological signs or symptoms.
  2. The motivation for the behavior is to assume the sick role.
  3. External incentives for the behavior are absent.

Source:Reference 6. Reprinted with permission

Although Mr. B experienced some depressive symptoms and anxiety just before discharge, we were hesitant to diagnose major depression because his symptoms appeared tied to situational factors. He also did not fit a particular personality disorder, although he showed characteristics of:

  • cluster A (odd ideas, solitary lifestyle)
  • cluster B (self-harm, narcissistic tendencies)
  • and cluster C (avoiding his relatives, dependence on others to meet his needs).
We could have diagnosed personality disorder, not otherwise specified, but we were unsure whether personality explained his pathology or if his personality characteristics warranted diagnosis.

Mr. B’s intentional production of physical symptoms strongly suggested malingering, but we instead diagnosed factitious disorder because he was clearly motivated to play the sick role despite lack of a secondary gain (Box 2). The patient admitted causing the gunshot wound and clearly connected his subsequent emotional relief with both his positive childhood experience in the hospital and his satisfaction after donating a kidney.

Researchers have tried to distinguish between factitious disorder and other types of self-harm. Claes and Vandereycken4 would consider Mr. B’s behavior “self-mutilation” rather than factitious. Turner calls DSM-IV-TR criteria for factitious disorder nebulous and says that lying about symptoms or their origin should be a necessary criterion.5 If so, then Mr. B’s condition might fit no DSM diagnosis.6

The authors’ observations

Although Mr. B’s diagnosis remained elusive, he needed a treatment plan before discharge to prevent another shooting and save his life.

We first considered psychotropics. Because Mr. B’s beliefs did not appear delusional, an antipsychotic would not be a useful first-line treatment. Nor would a benzodiazepine help Mr. B at this point, especially since we did not diagnose primary anxiety.

Although we did not diagnose a major depressive or anxiety-spectrum disorder, we felt an SSRI such as citalopram could help. According to some investigators, SSRIs might benefit patients with over-valued ideas that are not as persistent as delusions.7,8

Additionally, we felt supportive therapy could help Mr. B establish a therapeutic relationship with a provider to whom he could turn during future crises. Should Mr. B contemplate self-harm, the therapist could suggest medications, hospitalization, or other interventions. We also recommended pastoral counseling to increase his support within his faith.

OUTCOME Another shot?

Before discharge, we start citalopram, 10 mg/d, and schedule a follow-up appointment within 2 weeks. We also suggest that Mr. B:

  • move into an apartment near his outpatient doctors
  • get involved with the local Catholic community to build his support network.
When we contact Mr. B 2 months later, he says he discontinued citalopram because he felt no benefit from it. At his initial appointment with a psychiatrist, he denied depressive symptoms and was not scheduled for ongoing therapy. He has not spoken with clergy or other local church members because “I know they would say God has forgiven me.”

Mr. B calls his recent hospitalization upsetting because “I did not get the attention I wanted.” He endorses no immediate plan to shoot himself but voices concern that when his physical problems resolve, he might shoot Himself in the liver—as he had done 40 years ago—to bring himself full circle. “There’s still something attractive about this,” he says.

The authors’ observations

Patients with factitious illness commonly refuse mental health treatment.9

We feel Mr. B needs frequent ongoing appointments in a medical clinic where doctors can provide sufficient attention to counter his persistent self-harm urges. Regular appointments with a primary care physician—regardless of whether Mr. B is medically ill—could help him feel supported.

Related resources

  • Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996.
  • Sutton J, Martinson D. Self-injury Web site: What self-injury is.www.palace.net/~llama/psych/fwhat.html.
Drug brand names

  • Citalopram • Celexa
  • Omeprazole • Prilosec
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. The mysteries of Opus Dei. US News and World Report; December 14, 2003 (health section). Available at: http://www.usnews.com/usnews/culture/articles/031222/22jesus.b.htm. Accessed December 10, 2007.

2. Toke LA. transcribed by Potter DJ Flagellants. In: Catholic encyclopedia, vol. 6. New York: Robert Appleton Co.; 1909. Available at: http://www.newadvent.org/cathen/06089c.htm. Accessed December 10, 2007.

3. Glucklich A. Sacred pain: hurting the body for the sake of the soul. New York: Oxford University Press; 2001.

4. Claes L, Vandereycken W. Self-injurious behavior: differential diagnosis and functional differentiation. Compr Psychiatry 2007;48:137-44.

5. Turner MA. Factitious disorders: reformulating the DSM-IV criteria. Psychosomatics 2006;47:23-32.

6. Diagnostic and statistical manual of mental disorders. 4th ed. text rev. Washington, DC: American Psychiatric Association; 2000.

7. Veale D. Over-valued ideas: a conceptual analysis. Behav Res Ther 2002;40:383-400.

8. Jones E, Watson JP. Delusion, the overvalued idea and religious beliefs: a comparative analysis of their characteristics. Br J Psychiatry 1997;170:381-6.

9. Sutherland AJ, Rodin GM. Factitious disorders in a general hospital setting: clinical features and a review of the literature. Psychosomatics 1990;31:392-9.

References

1. The mysteries of Opus Dei. US News and World Report; December 14, 2003 (health section). Available at: http://www.usnews.com/usnews/culture/articles/031222/22jesus.b.htm. Accessed December 10, 2007.

2. Toke LA. transcribed by Potter DJ Flagellants. In: Catholic encyclopedia, vol. 6. New York: Robert Appleton Co.; 1909. Available at: http://www.newadvent.org/cathen/06089c.htm. Accessed December 10, 2007.

3. Glucklich A. Sacred pain: hurting the body for the sake of the soul. New York: Oxford University Press; 2001.

4. Claes L, Vandereycken W. Self-injurious behavior: differential diagnosis and functional differentiation. Compr Psychiatry 2007;48:137-44.

5. Turner MA. Factitious disorders: reformulating the DSM-IV criteria. Psychosomatics 2006;47:23-32.

6. Diagnostic and statistical manual of mental disorders. 4th ed. text rev. Washington, DC: American Psychiatric Association; 2000.

7. Veale D. Over-valued ideas: a conceptual analysis. Behav Res Ther 2002;40:383-400.

8. Jones E, Watson JP. Delusion, the overvalued idea and religious beliefs: a comparative analysis of their characteristics. Br J Psychiatry 1997;170:381-6.

9. Sutherland AJ, Rodin GM. Factitious disorders in a general hospital setting: clinical features and a review of the literature. Psychosomatics 1990;31:392-9.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
91-97
Page Number
91-97
Publications
Publications
Article Type
Display Headline
One patient’s ‘shot’ at redemption
Display Headline
One patient’s ‘shot’ at redemption
Legacy Keywords
Willow Naimark MD; Jeanne Johnson DDS; Annette Matthews MD; self-injury; delusional disorder; depression; major depression with psychotic features; major depression without psychotic features; obsessive-compulsive disorder; OCD; personality disorder; factitious disorder; malingering; supportive therapy; insight-oriented therapy
Legacy Keywords
Willow Naimark MD; Jeanne Johnson DDS; Annette Matthews MD; self-injury; delusional disorder; depression; major depression with psychotic features; major depression without psychotic features; obsessive-compulsive disorder; OCD; personality disorder; factitious disorder; malingering; supportive therapy; insight-oriented therapy
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Evaluating teen self-injury: Comorbidities and suicide risk

Article Type
Changed
Tue, 12/11/2018 - 15:07
Display Headline
Evaluating teen self-injury: Comorbidities and suicide risk

Alysha, age 15, is an “A” student and top athlete who feels her parents push her to be perfect. After getting a B on a test, she feels overwhelmed by shame and guilt. She locks herself in the bathroom and begins cutting her arm with a razor blade.

Self-injurious behavior (SIB) in adolescents can be associated with internalizing, externalizing, and substance abuse disorders (Table 1). For most practitioners, such as Alysha, a major goal of SIB is to relieve intolerable stress and negative affect.1

Although this secretive, highly addictive, learned behavior can be difficult to control, some clinical approaches can help these distressed teens and their parents. This article examines the dynamics of SIB, the association between suicidal ideation and SIB, and recommended treatments such as substitute behaviors and dialectic behavioral therapy.

Table 1

Common Axis I disorders among teens with SIB

Axis I disorderPrevalence*
Any internalizing disorder52%
  Major depressive disorder42
  Posttraumatic stress disorder24
  Generalized anxiety disorder16
Any externalizing disorder63
  Conduct disorder49
  Oppositional defiant disorder45
Any substance use disorder60
  Alcohol abuse18
  Alcohol dependence17
  Nicotine dependence39
  Marijuana abuse13
  Marijuana dependence30
  Other substance abuse3
  Other substance dependence6
*Among a sample of 89 adolescents who engaged in SIB
Source: Nock MD, Joiner TE Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):68.
Reprinted with permission from Elsevier.

Growing problem in adolescents

SIB is the deliberate infliction of harm to oneself, either internally or externally, without suicidal intent.1 This behavior is also known as impulsive self-injury, non-suicidal self-injury, self-mutilation, cutting, and self-harm. Once reported primarily in adults with borderline personality disorder, SIB is becoming common among adolescents:

  • Among 663 teens in community-sample survey, 46% engaged in some form of SIB in the past year, and 28% engaged in serious, repetitive behaviors.2
  • 13% to 23% of teenagers have engaged in SIB, according to literature review.3
  • Children as young as 9 have presented with SIB.
Not just cutting. Besides cutting, other methods of self-injury include:

  • burning
  • swallowing objects or substances
  • hitting oneself with the fist or against an object
  • cutting circulation to a digit
  • picking at skin
  • pulling out hair.
Individuals with chronic illnesses can engage in SIB by not complying with treatment, such as a diabetic taking too much or too little insulin or an epileptic not taking medication.

Socially sanctioned behaviors, such as body piercing and tattoos, usually are not considered SIB. They can be used as SIB, however, by teens who impulsively self-pierce or tattoo without appropriate hygiene or anesthetic agents. Their purpose is not to make a fashion statement but to produce pain or discomfort. Cultural behaviors that cause scarring as a rite of passage, such as the Native American Sun Dance, are not considered SIB.1

Despite increasing prevalence among adolescents, SIB remains a solitary behavior. Based on my clinical experience, teens may share ideas about SIB, but they generally don’t practice it in groups.

SIB psychodynamics

Adults and adolescents with SIB frequently have:

  • difficulty regulating emotions
  • unstable interpersonal relations
  • limited coping strategies.1
Adolescents with SIB frequently experience anger, and their self-harm can result from turning this anger inward because they are unable to express it toward others. This is seen in a patient who describes a cutting episode that occurred while you were on vacation but sees no connection between your absence and the SIB.

Shame is also common and can be a major barrier to diagnosing SIB. Adolescents who are ashamed of the behavior will go to great lengths to hide it from others, including clinicians. Despite the shame, many adolescents feel unable to stop engaging in SIB because it fulfills a powerful need.

Adults and adolescents who practice SIB most often report their behavior is motivated by affect regulation and tension release.4 Some adolescents engage in a different, manipulative form of SIB not to relieve tension but as a threat to prevent loss (Box).4

Behavioral reinforcement. An acute stressor—such as parental limits on behavior, feelings of rejection or abandonment by peers, or failing to achieve an unrealistic goal—triggers an escalating, intolerable affect. By experimentation or accident, an adolescent discovers that SIB provides rapid relief of the intolerable state—a calmness that may last for minutes, hours, or days. This relief reinforces the behavior, and the adolescent repeats SIB when faced with the next stressor.

Most individuals with SIB report a similar sequence of events. There is a trigger event, usually involving a real or perceived feeling of loss, rejection, or abandonment. The adolescent tries to resist the impulse to self-harm, feels escalating emotional distress, engages in SIB, and feels immediate relief.5

 

 

Inducing pain or bleeding as a means of relieving intolerable stress may be an attempt to:

  • turn emotional pain into more manageable physical pain
  • direct anger that cannot be expressed at others onto oneself
  • punish oneself for perceived misdeeds.4
SIB tends to escalate over time. Those who engage in it may require more frequent or intensive self-injury to achieve relief. Because the emotional state improves quickly and the adolescent feels a sense of control over the behavior, SIB rapidly can become habitual and difficult to interrupt.

Box

Anger, fear can trigger manipulative SIB

Peter, age 17, is fighting with his girlfriend, who threatens to leave. He grabs a knife and threatens to cut his arm. The girlfriend tries to take the knife. In their struggle, Peter accidentally cuts a tendon in his arm, which results in a permanent loss of function in his hand.

Unlike SIB for affective or tension release, manipulative or “in your face” SIB is not secretive. Adolescents who engage in manipulative SIB do so as a threat to control or induce guilt in others.4 This behavior is triggered primarily by attempts to change another person’s behavior or decision. Unlike the dynamics of impulsive SIB, this type of SIB does not seem to relieve tension; in fact, tension may increase. Manipulative SIB can be particularly dangerous because adolescents may accidentally cause injuries more severe than they intended.

CASE 2: Lingering effects of trauma

Melissa, age 13, has been sexually abused by her foster brother. She briefly returns to the home where her foster brother still resides. He has torn up her room, and her mother—who supports the foster brother—has left the room that way for her to find. Melissa returns to her current residence, cuts her arm 15 times, and pierces her tongue.

In therapy, she denies being angry or upset and does not know why she cut her arm or pierced her tongue.

An adolescent with a history of childhood trauma might have difficulty identifying and expressing internal states and developing trusting relations with others. He or she also can have high levels of anger and shame.

Even an adolescent who experienced neglect or loss of attachment object without overt physical or sexual abuse might have trouble establishing a therapeutic alliance because of difficulty with trust.6 He or she may have little or no capacity to identify emotional states, which limits insight into the behavior.5

Dissociation. Some adolescents use SIB to try to feel something or to bring themselves back from a dissociative state. They report feeling lost, alone, and disconnected from others and themselves. Some report seeing blood as a way of reconnecting with being alive.

Dissociation may occur in adolescents with a history of trauma, particularly in childhood. Abused individuals who engage in SIB may be identifying with the aggressor, attempting to cut away internalized negative images of the abuser or to control anger they are unable to acknowledge.6

Clues prompt further assessment

SIB assessment begins with screening for the behavior. If you find any indicators that suggest SIB (Table 2), question the adolescent about self-injury. Many adolescents want help—and will respond accurately to questions about self-injury—but need to be asked. They usually won’t volunteer the information during an initial evaluation.

Once you have identified SIB, explore the behavior. Document:

  • number, location, and age of injuries
  • depth of cuts (if applicable)
  • signs of infection.
Determine if the adolescent needs medical intervention. Discuss how the adolescent causes the injury, and what precipitates it. If possible, obtain some form of safety contract in which the adolescent agrees to not engage in SIB and to notify a designated adult if he or she feels like engaging in SIB or has done so.

Suicide screening. By definition, SIB does not include an intention to die, and most teenagers with SIB will deny suicidal intent. However, because the line between SIB and passive suicide can be thin, careful screening and ongoing monitoring for suicidal ideation and behavior in teens with SIB is essential. In one study:

  • 70% of adolescents who engaged in SIB had made one suicide attempt
  • 55% had made multiple attempts.7
A separate study found suicidal ideation and depression are keys to identifying adolescents with SIB at risk for suicide attempts.8 Because SIB and suicidal ideation/behaviors can co-occur, SIB safety contracts must cover both.
 

 


Address the parents’ concerns. During your assessment, also focus on the adolescent’s parents. They often are highly distressed, confused, and angry. They typically learn about the SIB from their child’s school counselors or peers and feel betrayed and guilty. They may want to be excessively intrusive and punitive and need support, information, and guidance to address their child’s safety.

Table 2

SIB: Spotting behavioral clues

Adolescents
  • Wearing long sleeves or pants in warm weather
  • Becoming increasingly isolated from family and peers
  • Spending long periods of time in the bathroom or bedroom
  • Deteriorating school performance
  • Decreasing hygiene
Parents
  • Finding blood on teen’s clothing or in unusual places in the home
  • Discovering sharp instruments are missing or placed in unusual locations

Treatment recommendations

Always take SIB in adolescents seriously, and not as something they will “outgrow.” Adolescents with SIB need help modulating affect, stabilizing interpersonal relationships, and developing more adaptive coping strategies and problem-solving skills. Underlying dynamics—especially childhood trauma—must be explored and resolved.

Few evidence-based studies have evaluated SIB treatment in adolescents. Clinicians have extrapolated suggested interventions from the adult literature; however, much of this data was obtained from treating adult women with borderline personality disorder.

No medications are FDA-approved for treating SIB. Use pharmacologic interventions to treat underlying disorders, such as depression or anxiety, so that patients are better able to participate in other therapeutic interventions.

Dialectical behavioral therapy (DBT) is the only therapeutic entity shown in controlled trials to successfully treat SIB. Weekly individual psychotherapy and skills training groups focus on:

  • regulating emotions
  • tolerating distress
  • improving interpersonal relationships
  • reducing identity confusion and maladaptive cognitions.9
Other types of therapy—including psychoanalysis, self psychology, object relations, and interpersonal approaches—have a similar understanding of impulsive SIB and employ similar approaches.

Substitute behaviors. The treatment goal is for patients to substitute less destructive behaviors in response to intense emotional states. Some can use techniques such as snapping a rubber band or rubbing ice against the skin, both of which cause discomfort without injury. Patients can listen to music, create art, write in journals, or engage in other physical activities. Each patient has to find a different behavior that works.

Prevention. Although SIB can be done with any object, most adolescents have a preferred method for causing self-injury and may have a “kit” of equipment. Identify and remove any tools the adolescent uses for self-injury. Because SIB is a highly ritualistic behavior, denying access to the preferred tools can help reduce self-injury frequency and convey that the behavior is unacceptable.

One individual should be designated to monitor the adolescent for SIB. Adolescents are seeking trust and do not respond well to constant questions about their behavior. Because some parents can become intrusive, monitoring may be best assigned to the adolescent’s therapist or a less emotional parent.

CASE 3: Scars provoke relapse

Claudia, a musically talented teen with SIB, withdraws from her choir when they choose costumes with short sleeves. She had not engaged in SIB in >1 year but has scarring and cheloid from the cuts. Years later she starts cutting again after laser treatments fail to remove the scars. She is frustrated because she will always have to wear long sleeves.

Risk of relapse. Therapy for SIB tends to be intense and difficult, with frequent relapses. To overcome SIB, the adolescent must want to stop and work hard at other coping strategies.

Treatment is essential, however, because this behavior can last for decades and leave scars that might interfere with future goals. The longer the adolescent has been dependent on the behavior, the more difficult it is to treat.

Related resources

  • Levenkron S. Cutting: understanding and overcoming self-mutilation. New York: W.W. Norton & Company; 1998.
  • Favazza AR. Bodies under siege: self-mutilation and body modification in culture and psychiatry. 2nd ed. Baltimore: The Johns Hopkins University Press; 1996.
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Simeon D, Favazza AR. Self-injurious behaviors: phenomenology and assessment. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, D.C: American Psychiatric Publishing; 2001:1-28.

2. Lloyd-Richardson EE, Perrine N, Dierker L, Kelley ML. Characteristics and functions of non-suicidal self-injury in a community sample of adolescents. Psychol Med 2007;37(8):1183-92.

3. 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):129-47.

4. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev 2007;27(2):226-39.

5. Leibenluft E, Gardner DL, Cowdry RW. The inner experience of the borderline self-mutilator. J Personal Disord 1987;1:317-24.

6. Guralnik O, Simeon D. Psychodynamic theory and treatment of impulsive self-injurious behaviors. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, DC: American Psychiatric Publishing; 2001:175-97.

7. Nock MD, Joiner TE, Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):65-72.

8. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res 2007;11(1):69-82.

9. Linehan MM, Armstrong HE, Suarez A, et al. Cognitive-behavioral treatment of chronically parasuicidal borderline patients. Arch Gen Psychiatry 1991;48:1060-4.

Article PDF
Author and Disclosure Information

Pamela A. Campbell, MD
Associate professor of psychiatry and pediatrics
University of Cincinnati School of Medicine
Cincinnati Children’s Hospital Medical Center
Cincinnati, OH

Issue
Current Psychiatry - 07(02)
Publications
Page Number
69-76
Legacy Keywords
teen self-injury; suicide risk; Pamela A. Campbell MD; affect modulation; coping skills; major depressive disorder; posttraumatic stress disorder; generalized anxiety disorder; conduct disorder; oppositional defiant disorder; alcohol abuse; alcohol dependence; nicotine dependence; marijuana abuse; marijuana dependence; dialectical behavioral therapy; destructive behaviors
Sections
Author and Disclosure Information

Pamela A. Campbell, MD
Associate professor of psychiatry and pediatrics
University of Cincinnati School of Medicine
Cincinnati Children’s Hospital Medical Center
Cincinnati, OH

Author and Disclosure Information

Pamela A. Campbell, MD
Associate professor of psychiatry and pediatrics
University of Cincinnati School of Medicine
Cincinnati Children’s Hospital Medical Center
Cincinnati, OH

Article PDF
Article PDF

Alysha, age 15, is an “A” student and top athlete who feels her parents push her to be perfect. After getting a B on a test, she feels overwhelmed by shame and guilt. She locks herself in the bathroom and begins cutting her arm with a razor blade.

Self-injurious behavior (SIB) in adolescents can be associated with internalizing, externalizing, and substance abuse disorders (Table 1). For most practitioners, such as Alysha, a major goal of SIB is to relieve intolerable stress and negative affect.1

Although this secretive, highly addictive, learned behavior can be difficult to control, some clinical approaches can help these distressed teens and their parents. This article examines the dynamics of SIB, the association between suicidal ideation and SIB, and recommended treatments such as substitute behaviors and dialectic behavioral therapy.

Table 1

Common Axis I disorders among teens with SIB

Axis I disorderPrevalence*
Any internalizing disorder52%
  Major depressive disorder42
  Posttraumatic stress disorder24
  Generalized anxiety disorder16
Any externalizing disorder63
  Conduct disorder49
  Oppositional defiant disorder45
Any substance use disorder60
  Alcohol abuse18
  Alcohol dependence17
  Nicotine dependence39
  Marijuana abuse13
  Marijuana dependence30
  Other substance abuse3
  Other substance dependence6
*Among a sample of 89 adolescents who engaged in SIB
Source: Nock MD, Joiner TE Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):68.
Reprinted with permission from Elsevier.

Growing problem in adolescents

SIB is the deliberate infliction of harm to oneself, either internally or externally, without suicidal intent.1 This behavior is also known as impulsive self-injury, non-suicidal self-injury, self-mutilation, cutting, and self-harm. Once reported primarily in adults with borderline personality disorder, SIB is becoming common among adolescents:

  • Among 663 teens in community-sample survey, 46% engaged in some form of SIB in the past year, and 28% engaged in serious, repetitive behaviors.2
  • 13% to 23% of teenagers have engaged in SIB, according to literature review.3
  • Children as young as 9 have presented with SIB.
Not just cutting. Besides cutting, other methods of self-injury include:

  • burning
  • swallowing objects or substances
  • hitting oneself with the fist or against an object
  • cutting circulation to a digit
  • picking at skin
  • pulling out hair.
Individuals with chronic illnesses can engage in SIB by not complying with treatment, such as a diabetic taking too much or too little insulin or an epileptic not taking medication.

Socially sanctioned behaviors, such as body piercing and tattoos, usually are not considered SIB. They can be used as SIB, however, by teens who impulsively self-pierce or tattoo without appropriate hygiene or anesthetic agents. Their purpose is not to make a fashion statement but to produce pain or discomfort. Cultural behaviors that cause scarring as a rite of passage, such as the Native American Sun Dance, are not considered SIB.1

Despite increasing prevalence among adolescents, SIB remains a solitary behavior. Based on my clinical experience, teens may share ideas about SIB, but they generally don’t practice it in groups.

SIB psychodynamics

Adults and adolescents with SIB frequently have:

  • difficulty regulating emotions
  • unstable interpersonal relations
  • limited coping strategies.1
Adolescents with SIB frequently experience anger, and their self-harm can result from turning this anger inward because they are unable to express it toward others. This is seen in a patient who describes a cutting episode that occurred while you were on vacation but sees no connection between your absence and the SIB.

Shame is also common and can be a major barrier to diagnosing SIB. Adolescents who are ashamed of the behavior will go to great lengths to hide it from others, including clinicians. Despite the shame, many adolescents feel unable to stop engaging in SIB because it fulfills a powerful need.

Adults and adolescents who practice SIB most often report their behavior is motivated by affect regulation and tension release.4 Some adolescents engage in a different, manipulative form of SIB not to relieve tension but as a threat to prevent loss (Box).4

Behavioral reinforcement. An acute stressor—such as parental limits on behavior, feelings of rejection or abandonment by peers, or failing to achieve an unrealistic goal—triggers an escalating, intolerable affect. By experimentation or accident, an adolescent discovers that SIB provides rapid relief of the intolerable state—a calmness that may last for minutes, hours, or days. This relief reinforces the behavior, and the adolescent repeats SIB when faced with the next stressor.

Most individuals with SIB report a similar sequence of events. There is a trigger event, usually involving a real or perceived feeling of loss, rejection, or abandonment. The adolescent tries to resist the impulse to self-harm, feels escalating emotional distress, engages in SIB, and feels immediate relief.5

 

 

Inducing pain or bleeding as a means of relieving intolerable stress may be an attempt to:

  • turn emotional pain into more manageable physical pain
  • direct anger that cannot be expressed at others onto oneself
  • punish oneself for perceived misdeeds.4
SIB tends to escalate over time. Those who engage in it may require more frequent or intensive self-injury to achieve relief. Because the emotional state improves quickly and the adolescent feels a sense of control over the behavior, SIB rapidly can become habitual and difficult to interrupt.

Box

Anger, fear can trigger manipulative SIB

Peter, age 17, is fighting with his girlfriend, who threatens to leave. He grabs a knife and threatens to cut his arm. The girlfriend tries to take the knife. In their struggle, Peter accidentally cuts a tendon in his arm, which results in a permanent loss of function in his hand.

Unlike SIB for affective or tension release, manipulative or “in your face” SIB is not secretive. Adolescents who engage in manipulative SIB do so as a threat to control or induce guilt in others.4 This behavior is triggered primarily by attempts to change another person’s behavior or decision. Unlike the dynamics of impulsive SIB, this type of SIB does not seem to relieve tension; in fact, tension may increase. Manipulative SIB can be particularly dangerous because adolescents may accidentally cause injuries more severe than they intended.

CASE 2: Lingering effects of trauma

Melissa, age 13, has been sexually abused by her foster brother. She briefly returns to the home where her foster brother still resides. He has torn up her room, and her mother—who supports the foster brother—has left the room that way for her to find. Melissa returns to her current residence, cuts her arm 15 times, and pierces her tongue.

In therapy, she denies being angry or upset and does not know why she cut her arm or pierced her tongue.

An adolescent with a history of childhood trauma might have difficulty identifying and expressing internal states and developing trusting relations with others. He or she also can have high levels of anger and shame.

Even an adolescent who experienced neglect or loss of attachment object without overt physical or sexual abuse might have trouble establishing a therapeutic alliance because of difficulty with trust.6 He or she may have little or no capacity to identify emotional states, which limits insight into the behavior.5

Dissociation. Some adolescents use SIB to try to feel something or to bring themselves back from a dissociative state. They report feeling lost, alone, and disconnected from others and themselves. Some report seeing blood as a way of reconnecting with being alive.

Dissociation may occur in adolescents with a history of trauma, particularly in childhood. Abused individuals who engage in SIB may be identifying with the aggressor, attempting to cut away internalized negative images of the abuser or to control anger they are unable to acknowledge.6

Clues prompt further assessment

SIB assessment begins with screening for the behavior. If you find any indicators that suggest SIB (Table 2), question the adolescent about self-injury. Many adolescents want help—and will respond accurately to questions about self-injury—but need to be asked. They usually won’t volunteer the information during an initial evaluation.

Once you have identified SIB, explore the behavior. Document:

  • number, location, and age of injuries
  • depth of cuts (if applicable)
  • signs of infection.
Determine if the adolescent needs medical intervention. Discuss how the adolescent causes the injury, and what precipitates it. If possible, obtain some form of safety contract in which the adolescent agrees to not engage in SIB and to notify a designated adult if he or she feels like engaging in SIB or has done so.

Suicide screening. By definition, SIB does not include an intention to die, and most teenagers with SIB will deny suicidal intent. However, because the line between SIB and passive suicide can be thin, careful screening and ongoing monitoring for suicidal ideation and behavior in teens with SIB is essential. In one study:

  • 70% of adolescents who engaged in SIB had made one suicide attempt
  • 55% had made multiple attempts.7
A separate study found suicidal ideation and depression are keys to identifying adolescents with SIB at risk for suicide attempts.8 Because SIB and suicidal ideation/behaviors can co-occur, SIB safety contracts must cover both.
 

 


Address the parents’ concerns. During your assessment, also focus on the adolescent’s parents. They often are highly distressed, confused, and angry. They typically learn about the SIB from their child’s school counselors or peers and feel betrayed and guilty. They may want to be excessively intrusive and punitive and need support, information, and guidance to address their child’s safety.

Table 2

SIB: Spotting behavioral clues

Adolescents
  • Wearing long sleeves or pants in warm weather
  • Becoming increasingly isolated from family and peers
  • Spending long periods of time in the bathroom or bedroom
  • Deteriorating school performance
  • Decreasing hygiene
Parents
  • Finding blood on teen’s clothing or in unusual places in the home
  • Discovering sharp instruments are missing or placed in unusual locations

Treatment recommendations

Always take SIB in adolescents seriously, and not as something they will “outgrow.” Adolescents with SIB need help modulating affect, stabilizing interpersonal relationships, and developing more adaptive coping strategies and problem-solving skills. Underlying dynamics—especially childhood trauma—must be explored and resolved.

Few evidence-based studies have evaluated SIB treatment in adolescents. Clinicians have extrapolated suggested interventions from the adult literature; however, much of this data was obtained from treating adult women with borderline personality disorder.

No medications are FDA-approved for treating SIB. Use pharmacologic interventions to treat underlying disorders, such as depression or anxiety, so that patients are better able to participate in other therapeutic interventions.

Dialectical behavioral therapy (DBT) is the only therapeutic entity shown in controlled trials to successfully treat SIB. Weekly individual psychotherapy and skills training groups focus on:

  • regulating emotions
  • tolerating distress
  • improving interpersonal relationships
  • reducing identity confusion and maladaptive cognitions.9
Other types of therapy—including psychoanalysis, self psychology, object relations, and interpersonal approaches—have a similar understanding of impulsive SIB and employ similar approaches.

Substitute behaviors. The treatment goal is for patients to substitute less destructive behaviors in response to intense emotional states. Some can use techniques such as snapping a rubber band or rubbing ice against the skin, both of which cause discomfort without injury. Patients can listen to music, create art, write in journals, or engage in other physical activities. Each patient has to find a different behavior that works.

Prevention. Although SIB can be done with any object, most adolescents have a preferred method for causing self-injury and may have a “kit” of equipment. Identify and remove any tools the adolescent uses for self-injury. Because SIB is a highly ritualistic behavior, denying access to the preferred tools can help reduce self-injury frequency and convey that the behavior is unacceptable.

One individual should be designated to monitor the adolescent for SIB. Adolescents are seeking trust and do not respond well to constant questions about their behavior. Because some parents can become intrusive, monitoring may be best assigned to the adolescent’s therapist or a less emotional parent.

CASE 3: Scars provoke relapse

Claudia, a musically talented teen with SIB, withdraws from her choir when they choose costumes with short sleeves. She had not engaged in SIB in >1 year but has scarring and cheloid from the cuts. Years later she starts cutting again after laser treatments fail to remove the scars. She is frustrated because she will always have to wear long sleeves.

Risk of relapse. Therapy for SIB tends to be intense and difficult, with frequent relapses. To overcome SIB, the adolescent must want to stop and work hard at other coping strategies.

Treatment is essential, however, because this behavior can last for decades and leave scars that might interfere with future goals. The longer the adolescent has been dependent on the behavior, the more difficult it is to treat.

Related resources

  • Levenkron S. Cutting: understanding and overcoming self-mutilation. New York: W.W. Norton & Company; 1998.
  • Favazza AR. Bodies under siege: self-mutilation and body modification in culture and psychiatry. 2nd ed. Baltimore: The Johns Hopkins University Press; 1996.
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Alysha, age 15, is an “A” student and top athlete who feels her parents push her to be perfect. After getting a B on a test, she feels overwhelmed by shame and guilt. She locks herself in the bathroom and begins cutting her arm with a razor blade.

Self-injurious behavior (SIB) in adolescents can be associated with internalizing, externalizing, and substance abuse disorders (Table 1). For most practitioners, such as Alysha, a major goal of SIB is to relieve intolerable stress and negative affect.1

Although this secretive, highly addictive, learned behavior can be difficult to control, some clinical approaches can help these distressed teens and their parents. This article examines the dynamics of SIB, the association between suicidal ideation and SIB, and recommended treatments such as substitute behaviors and dialectic behavioral therapy.

Table 1

Common Axis I disorders among teens with SIB

Axis I disorderPrevalence*
Any internalizing disorder52%
  Major depressive disorder42
  Posttraumatic stress disorder24
  Generalized anxiety disorder16
Any externalizing disorder63
  Conduct disorder49
  Oppositional defiant disorder45
Any substance use disorder60
  Alcohol abuse18
  Alcohol dependence17
  Nicotine dependence39
  Marijuana abuse13
  Marijuana dependence30
  Other substance abuse3
  Other substance dependence6
*Among a sample of 89 adolescents who engaged in SIB
Source: Nock MD, Joiner TE Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):68.
Reprinted with permission from Elsevier.

Growing problem in adolescents

SIB is the deliberate infliction of harm to oneself, either internally or externally, without suicidal intent.1 This behavior is also known as impulsive self-injury, non-suicidal self-injury, self-mutilation, cutting, and self-harm. Once reported primarily in adults with borderline personality disorder, SIB is becoming common among adolescents:

  • Among 663 teens in community-sample survey, 46% engaged in some form of SIB in the past year, and 28% engaged in serious, repetitive behaviors.2
  • 13% to 23% of teenagers have engaged in SIB, according to literature review.3
  • Children as young as 9 have presented with SIB.
Not just cutting. Besides cutting, other methods of self-injury include:

  • burning
  • swallowing objects or substances
  • hitting oneself with the fist or against an object
  • cutting circulation to a digit
  • picking at skin
  • pulling out hair.
Individuals with chronic illnesses can engage in SIB by not complying with treatment, such as a diabetic taking too much or too little insulin or an epileptic not taking medication.

Socially sanctioned behaviors, such as body piercing and tattoos, usually are not considered SIB. They can be used as SIB, however, by teens who impulsively self-pierce or tattoo without appropriate hygiene or anesthetic agents. Their purpose is not to make a fashion statement but to produce pain or discomfort. Cultural behaviors that cause scarring as a rite of passage, such as the Native American Sun Dance, are not considered SIB.1

Despite increasing prevalence among adolescents, SIB remains a solitary behavior. Based on my clinical experience, teens may share ideas about SIB, but they generally don’t practice it in groups.

SIB psychodynamics

Adults and adolescents with SIB frequently have:

  • difficulty regulating emotions
  • unstable interpersonal relations
  • limited coping strategies.1
Adolescents with SIB frequently experience anger, and their self-harm can result from turning this anger inward because they are unable to express it toward others. This is seen in a patient who describes a cutting episode that occurred while you were on vacation but sees no connection between your absence and the SIB.

Shame is also common and can be a major barrier to diagnosing SIB. Adolescents who are ashamed of the behavior will go to great lengths to hide it from others, including clinicians. Despite the shame, many adolescents feel unable to stop engaging in SIB because it fulfills a powerful need.

Adults and adolescents who practice SIB most often report their behavior is motivated by affect regulation and tension release.4 Some adolescents engage in a different, manipulative form of SIB not to relieve tension but as a threat to prevent loss (Box).4

Behavioral reinforcement. An acute stressor—such as parental limits on behavior, feelings of rejection or abandonment by peers, or failing to achieve an unrealistic goal—triggers an escalating, intolerable affect. By experimentation or accident, an adolescent discovers that SIB provides rapid relief of the intolerable state—a calmness that may last for minutes, hours, or days. This relief reinforces the behavior, and the adolescent repeats SIB when faced with the next stressor.

Most individuals with SIB report a similar sequence of events. There is a trigger event, usually involving a real or perceived feeling of loss, rejection, or abandonment. The adolescent tries to resist the impulse to self-harm, feels escalating emotional distress, engages in SIB, and feels immediate relief.5

 

 

Inducing pain or bleeding as a means of relieving intolerable stress may be an attempt to:

  • turn emotional pain into more manageable physical pain
  • direct anger that cannot be expressed at others onto oneself
  • punish oneself for perceived misdeeds.4
SIB tends to escalate over time. Those who engage in it may require more frequent or intensive self-injury to achieve relief. Because the emotional state improves quickly and the adolescent feels a sense of control over the behavior, SIB rapidly can become habitual and difficult to interrupt.

Box

Anger, fear can trigger manipulative SIB

Peter, age 17, is fighting with his girlfriend, who threatens to leave. He grabs a knife and threatens to cut his arm. The girlfriend tries to take the knife. In their struggle, Peter accidentally cuts a tendon in his arm, which results in a permanent loss of function in his hand.

Unlike SIB for affective or tension release, manipulative or “in your face” SIB is not secretive. Adolescents who engage in manipulative SIB do so as a threat to control or induce guilt in others.4 This behavior is triggered primarily by attempts to change another person’s behavior or decision. Unlike the dynamics of impulsive SIB, this type of SIB does not seem to relieve tension; in fact, tension may increase. Manipulative SIB can be particularly dangerous because adolescents may accidentally cause injuries more severe than they intended.

CASE 2: Lingering effects of trauma

Melissa, age 13, has been sexually abused by her foster brother. She briefly returns to the home where her foster brother still resides. He has torn up her room, and her mother—who supports the foster brother—has left the room that way for her to find. Melissa returns to her current residence, cuts her arm 15 times, and pierces her tongue.

In therapy, she denies being angry or upset and does not know why she cut her arm or pierced her tongue.

An adolescent with a history of childhood trauma might have difficulty identifying and expressing internal states and developing trusting relations with others. He or she also can have high levels of anger and shame.

Even an adolescent who experienced neglect or loss of attachment object without overt physical or sexual abuse might have trouble establishing a therapeutic alliance because of difficulty with trust.6 He or she may have little or no capacity to identify emotional states, which limits insight into the behavior.5

Dissociation. Some adolescents use SIB to try to feel something or to bring themselves back from a dissociative state. They report feeling lost, alone, and disconnected from others and themselves. Some report seeing blood as a way of reconnecting with being alive.

Dissociation may occur in adolescents with a history of trauma, particularly in childhood. Abused individuals who engage in SIB may be identifying with the aggressor, attempting to cut away internalized negative images of the abuser or to control anger they are unable to acknowledge.6

Clues prompt further assessment

SIB assessment begins with screening for the behavior. If you find any indicators that suggest SIB (Table 2), question the adolescent about self-injury. Many adolescents want help—and will respond accurately to questions about self-injury—but need to be asked. They usually won’t volunteer the information during an initial evaluation.

Once you have identified SIB, explore the behavior. Document:

  • number, location, and age of injuries
  • depth of cuts (if applicable)
  • signs of infection.
Determine if the adolescent needs medical intervention. Discuss how the adolescent causes the injury, and what precipitates it. If possible, obtain some form of safety contract in which the adolescent agrees to not engage in SIB and to notify a designated adult if he or she feels like engaging in SIB or has done so.

Suicide screening. By definition, SIB does not include an intention to die, and most teenagers with SIB will deny suicidal intent. However, because the line between SIB and passive suicide can be thin, careful screening and ongoing monitoring for suicidal ideation and behavior in teens with SIB is essential. In one study:

  • 70% of adolescents who engaged in SIB had made one suicide attempt
  • 55% had made multiple attempts.7
A separate study found suicidal ideation and depression are keys to identifying adolescents with SIB at risk for suicide attempts.8 Because SIB and suicidal ideation/behaviors can co-occur, SIB safety contracts must cover both.
 

 


Address the parents’ concerns. During your assessment, also focus on the adolescent’s parents. They often are highly distressed, confused, and angry. They typically learn about the SIB from their child’s school counselors or peers and feel betrayed and guilty. They may want to be excessively intrusive and punitive and need support, information, and guidance to address their child’s safety.

Table 2

SIB: Spotting behavioral clues

Adolescents
  • Wearing long sleeves or pants in warm weather
  • Becoming increasingly isolated from family and peers
  • Spending long periods of time in the bathroom or bedroom
  • Deteriorating school performance
  • Decreasing hygiene
Parents
  • Finding blood on teen’s clothing or in unusual places in the home
  • Discovering sharp instruments are missing or placed in unusual locations

Treatment recommendations

Always take SIB in adolescents seriously, and not as something they will “outgrow.” Adolescents with SIB need help modulating affect, stabilizing interpersonal relationships, and developing more adaptive coping strategies and problem-solving skills. Underlying dynamics—especially childhood trauma—must be explored and resolved.

Few evidence-based studies have evaluated SIB treatment in adolescents. Clinicians have extrapolated suggested interventions from the adult literature; however, much of this data was obtained from treating adult women with borderline personality disorder.

No medications are FDA-approved for treating SIB. Use pharmacologic interventions to treat underlying disorders, such as depression or anxiety, so that patients are better able to participate in other therapeutic interventions.

Dialectical behavioral therapy (DBT) is the only therapeutic entity shown in controlled trials to successfully treat SIB. Weekly individual psychotherapy and skills training groups focus on:

  • regulating emotions
  • tolerating distress
  • improving interpersonal relationships
  • reducing identity confusion and maladaptive cognitions.9
Other types of therapy—including psychoanalysis, self psychology, object relations, and interpersonal approaches—have a similar understanding of impulsive SIB and employ similar approaches.

Substitute behaviors. The treatment goal is for patients to substitute less destructive behaviors in response to intense emotional states. Some can use techniques such as snapping a rubber band or rubbing ice against the skin, both of which cause discomfort without injury. Patients can listen to music, create art, write in journals, or engage in other physical activities. Each patient has to find a different behavior that works.

Prevention. Although SIB can be done with any object, most adolescents have a preferred method for causing self-injury and may have a “kit” of equipment. Identify and remove any tools the adolescent uses for self-injury. Because SIB is a highly ritualistic behavior, denying access to the preferred tools can help reduce self-injury frequency and convey that the behavior is unacceptable.

One individual should be designated to monitor the adolescent for SIB. Adolescents are seeking trust and do not respond well to constant questions about their behavior. Because some parents can become intrusive, monitoring may be best assigned to the adolescent’s therapist or a less emotional parent.

CASE 3: Scars provoke relapse

Claudia, a musically talented teen with SIB, withdraws from her choir when they choose costumes with short sleeves. She had not engaged in SIB in >1 year but has scarring and cheloid from the cuts. Years later she starts cutting again after laser treatments fail to remove the scars. She is frustrated because she will always have to wear long sleeves.

Risk of relapse. Therapy for SIB tends to be intense and difficult, with frequent relapses. To overcome SIB, the adolescent must want to stop and work hard at other coping strategies.

Treatment is essential, however, because this behavior can last for decades and leave scars that might interfere with future goals. The longer the adolescent has been dependent on the behavior, the more difficult it is to treat.

Related resources

  • Levenkron S. Cutting: understanding and overcoming self-mutilation. New York: W.W. Norton & Company; 1998.
  • Favazza AR. Bodies under siege: self-mutilation and body modification in culture and psychiatry. 2nd ed. Baltimore: The Johns Hopkins University Press; 1996.
Disclosure

The author reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Simeon D, Favazza AR. Self-injurious behaviors: phenomenology and assessment. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, D.C: American Psychiatric Publishing; 2001:1-28.

2. Lloyd-Richardson EE, Perrine N, Dierker L, Kelley ML. Characteristics and functions of non-suicidal self-injury in a community sample of adolescents. Psychol Med 2007;37(8):1183-92.

3. 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):129-47.

4. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev 2007;27(2):226-39.

5. Leibenluft E, Gardner DL, Cowdry RW. The inner experience of the borderline self-mutilator. J Personal Disord 1987;1:317-24.

6. Guralnik O, Simeon D. Psychodynamic theory and treatment of impulsive self-injurious behaviors. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, DC: American Psychiatric Publishing; 2001:175-97.

7. Nock MD, Joiner TE, Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):65-72.

8. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res 2007;11(1):69-82.

9. Linehan MM, Armstrong HE, Suarez A, et al. Cognitive-behavioral treatment of chronically parasuicidal borderline patients. Arch Gen Psychiatry 1991;48:1060-4.

References

1. Simeon D, Favazza AR. Self-injurious behaviors: phenomenology and assessment. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, D.C: American Psychiatric Publishing; 2001:1-28.

2. Lloyd-Richardson EE, Perrine N, Dierker L, Kelley ML. Characteristics and functions of non-suicidal self-injury in a community sample of adolescents. Psychol Med 2007;37(8):1183-92.

3. 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):129-47.

4. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev 2007;27(2):226-39.

5. Leibenluft E, Gardner DL, Cowdry RW. The inner experience of the borderline self-mutilator. J Personal Disord 1987;1:317-24.

6. Guralnik O, Simeon D. Psychodynamic theory and treatment of impulsive self-injurious behaviors. In: Simeon D, Hollander E, eds. Self-injurious behaviors: assessment and treatment. Washington, DC: American Psychiatric Publishing; 2001:175-97.

7. Nock MD, Joiner TE, Jr, Gordon KL, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relationship to suicide attempts. Psychiatry Res 2006;144(1):65-72.

8. Muehlenkamp JJ, Gutierrez PM. Risk for suicide attempts among adolescents who engage in non-suicidal self-injury. Arch Suicide Res 2007;11(1):69-82.

9. Linehan MM, Armstrong HE, Suarez A, et al. Cognitive-behavioral treatment of chronically parasuicidal borderline patients. Arch Gen Psychiatry 1991;48:1060-4.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
69-76
Page Number
69-76
Publications
Publications
Article Type
Display Headline
Evaluating teen self-injury: Comorbidities and suicide risk
Display Headline
Evaluating teen self-injury: Comorbidities and suicide risk
Legacy Keywords
teen self-injury; suicide risk; Pamela A. Campbell MD; affect modulation; coping skills; major depressive disorder; posttraumatic stress disorder; generalized anxiety disorder; conduct disorder; oppositional defiant disorder; alcohol abuse; alcohol dependence; nicotine dependence; marijuana abuse; marijuana dependence; dialectical behavioral therapy; destructive behaviors
Legacy Keywords
teen self-injury; suicide risk; Pamela A. Campbell MD; affect modulation; coping skills; major depressive disorder; posttraumatic stress disorder; generalized anxiety disorder; conduct disorder; oppositional defiant disorder; alcohol abuse; alcohol dependence; nicotine dependence; marijuana abuse; marijuana dependence; dialectical behavioral therapy; destructive behaviors
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Treating anxiety during pregnancy: Just how safe are SSRIs?

Article Type
Changed
Tue, 12/11/2018 - 15:07
Display Headline
Treating anxiety during pregnancy: Just how safe are SSRIs?

Ms. K, age 25, is 6 weeks pregnant and is taking medications for generalized anxiety disorder (GAD). When she was diagnosed with GAD at age 19, her symptoms included 6 months of excessive anxiety—insomnia, fatigue, difficulty with concentration, and psychomotor agitation—without mood symptoms. These symptoms interfered greatly with her schoolwork and other daily activities.

For 6 years Ms. K has been taking the selective serotonin reuptake inhibitor (SSRI) paroxetine, 15 mg/d, and the benzodiazepine clonazepam, 0.5 mg as needed, with good symptom control. Now that she is pregnant and her primary care doctor has refused to continue these medications, she is seeking treatment and advice.

Not enough is known about how to safely treat anxiety disorders during pregnancy, and physicians are not sure what to do with patients such as Ms. K. Without evidence-based guidelines, we feel anxious about potential risks to mother and fetus as we try to provide appropriate drug therapy.

To help you and your patients weigh the risks and benefits of perinatal treatments for anxiety disorders, this article briefly summarizes the evidence on:

  • anxiety disorders’ natural history during pregnancy
  • how untreated maternal anxiety affects the fetus
  • nonpharmacologic therapies for anxiety disorders
  • a plan to manage fetal risks by staggering SSRI and benzodiazepine use during the first and third trimesters.

Anxiety during pregnancy

Nearly one-third of women experience an anxiety disorder during their lives, with peak onset during childbearing years.1,2 Compared with research on perinatal depression, far fewer studies have examined anxiety disorders’ onset, presentation, prevalence, and treatment.1

The literature includes no studies of the course of preexisting GAD or posttraumatic stress disorder (PTSD) and no evidence that symptoms of preexisting obsessive-compulsive disorder (OCD) change during pregnancy. Some studies of panic disorder show symptoms improving during pregnancy, whereas others do not (Table 1).1

One small study done in late pregnancy found a significant association between the prevalence of an anxiety disorder, maternal primiparity, and comorbid medical conditions. Thus, a woman in her first pregnancy may be at increased risk to develop an anxiety disorder if she has a comorbid medical condition.3 As in the case of Ms. K, however, continuation of preexisting anxiety appears more likely than onset of a new anxiety disorder during pregnancy.

Table 1

How pregnancy affects the course of 4 anxiety disorders

DisorderPrevalenceEffect
Generalized anxiety disorder (GAD)8.5% of women experience GAD during the third trimester, compared with a 5% prevalence in the general populationNo studies have reported on the course of GAD in pregnant women with preexisting disorder
Obsessive-compulsive disorder (OCD)2% to 12% of OCD outpatients of childbearing age report onset during pregnancyPreexisting OCD usually shows no change during pregnancy but may worsen postpartum
Panic disorder (PD)1.3% to 2% in pregnant women, compared with 1.5% to 3.5% in the general populationPanic symptoms in women with preexisting PD may improve during pregnancy and worsen postpartum
Posttraumatic stress disorder (PTSD)2.3% to 7.7% in pregnant women and 0% to 6.9% postpartum, compared with 1% to 14% in the communityNo studies have reported on the course of PTSD in pregnant women with preexisting disorder
Source: References 1,2

Fetal risks from maternal anxiety

Fetal risk from severe maternal anxiety is not zero. Offspring born to high-anxiety mothers exhibit neurobehavioral differences compared with offspring of calmer mothers. Changes in high-anxiety mothers’ offspring include:

  • altered EEG activation and vagal tone
  • increased time in deep sleep and less time in active alert states
  • lower performance on the Brazelton Neonatal Behavior Assessment Scale.4
A cohort study by Teixeira et al5 found an association between maternal anxiety in pregnancy and uterine artery resistance, suggesting a possible mechanism by which a mother’s psychologic state may affect fetal development. High anxiety and self-reported life stress during pregnancy also are associated consistently with abnormal, high-frequency heart rate variability in infants—a finding linked with negative infant behavior and later adult hostility.6

Exposure to maternal high anxiety has been associated with mental developmental delays in infants and increased risk for behavioral and emotional problems in young children.7-10 Anxiety may not directly cause intrauterine growth retardation and preterm delivery, but it is significantly associated with prenatal tobacco, alcohol, and narcotics use—which predicts these and other negative neonatal outcomes.11

Anxiety during pregnancy is a risk factor for postnatal depressive symptoms, independent of depressed mood and family or marital stressors during pregnancy.12 Mothers with postpartum depression appear less able to respond sensitively and competently to their newborns, and these infants may be at increased risk of behavioral, emotional, and cognitive problems.
 

 

7,13

CASE CONTINUED: ‘Stay the course’

Ms. K worries that she could not tolerate recurrence of her anxiety symptoms and wishes to continue both medications. Her husband concurs, but they want to minimize potential risks to their baby. You discuss the options for treating anxiety symptoms during pregnancy, including medications, psychotherapy, and behavioral treatments.

Treatment decisions

Ideally you’ll begin treating anxiety disorders in women of childbearing age with preconception psychoeducation. Explaining the risks of medications if she were to become pregnant and asking about the contraception she is using are de rigueur. Psychotherapy is low risk to the fetus and is considered first choice for treating mild to moderate anxiety in women of childbearing age who plan to become pregnant (Box).1,14-17

Box

Psychotherapy: First choice for anxiety during pregnancy

No studies directly address the efficacy or outcome of any psychotherapy for anxiety in pregnancy. Even so:

  • For mild to moderate anxiety, psychotherapy is the first-line treatment for pregnant women.
  • Interpersonal psychotherapy (IPT) without medications can reduce depressive symptoms in pregnant women with depression.14
  • Cognitive-behavioral therapy (CBT) without medications has shown efficacy for anxiety disorders in psychiatric populations.15,16

Because no evidence suggests that pregnant women require different psychotherapeutic recommendations than other psychiatric patients, consider a course of CBT that targets anxiety symptoms or IPT for a pregnant patient with an anxiety disorder.

Relaxation therapy also has shown efficacy in treating anxiety disorders. In a randomized controlled trial of 110 pregnant women with high-level anxiety, 7 weeks of applied relaxation training sessions was associated with significant reductions in low-weight births, cesarean sections, and instrumental extractions.16,17

Because poor marital relationships are consistent psychosocial predictors of anxiety during pregnancy and postpartum depression,1 recommend family or marital therapy when appropriate.

Psychotherapy alone is inadequate, however, for the many patients—such as Ms. K—who present already pregnant with a history of moderate to severe anxiety. Adjunctive psychotropic therapy—along with various nonmedication therapies—is warranted for patients whose social or occupational functioning would be substantially impaired by suboptimal control of anxiety during pregnancy.

Because Ms. K wishes to continue taking paroxetine and clonazepam, what can you tell her about the risks and benefits of SSRIs and benzodiazepines during pregnancy?

SSRIs in pregnancy

Teratogenicity. Compared with benzodiazepines, SSRIs have been considered agents of choice for use during pregnancy because of a lower risk of teratogenic effects.15 Paroxetine, however, appears to pose a greater risk for teratogenicity than other SSRIs.

An increased risk for fetal ventricular and/or atrial septal defects has been associated with first-trimester exposure to paroxetine, but no other SSRI.18 First trimester exposure to paroxetine at doses averaging 25 mg/d has been associated with statistically significant risks of major congenital anomalies (2-fold increase) and major cardiac anomalies (3-fold increase),19 although other studies have failed to reproduce this finding. A meta-analysis of 7 studies by Bar-Oz et al20 found an association between first-trimester paroxetine exposure and a significant increase in risk for cardiac malformations (odds ratio [OR] 1.72; 95% CI,1.22-2.42).

The overall rate of fetal malformations from SSRIs appears to be low, although most studies have examined only fluoxetine or paroxetine. Some studies have reported various malformations with fluoxetine or sertraline, but others have not. In Finland, a population-based study found no increase in rate of major congenital malformations in offspring of 1,782 women who filled prescriptions for SSRIs during pregnancy, compared with the general population rate of 1% to 3%.21

Neurobehavioral effects. SSRI exposure during fetal life has shown no long-term neurobehavioral effects. A blinded prospective study by Nulman et al22 found no differences in global IQ scores, language development, or behavioral development among children age ≤5 who were exposed in utero to fluoxetine (n=40) or a tricyclic antidepressant (n=46), compared with unexposed children of nondepressed mothers (n=36). Similarly, using reports from teachers and clinical measures of internalizing behaviors, Misri et al10 found no increase in depression, anxiety, or withdrawal in 4-year-olds with prenatal exposure to SSRIs (n=22), compared with nonexposed children (n=14).

Pulmonary hypertension. SSRI exposure in later pregnancy may increase the rate of persistent pulmonary hypertension of the newborn (PPHN), which occurs in 1 to 2 infants per 1,000 live births. PPHN showed a statistically significant association with late prenatal SSRI exposure (OR 6.1) in a study that controlled for maternal smoking, body mass index, and diabetes.23 PPHN occurred in approximately 1% of infants exposed to SSRIs in late pregnancy. PPHN rates were not affected by maternal depression/anxiety, non-SSRI antidepressant exposure throughout pregnancy, or SSRI exposure during early pregnancy only.

 

 

Toxicity and withdrawal syndromes. Infants of women who continue to take SSRIs just before delivery can develop toxicity or withdrawal syndromes. Occurrence of either syndrome depends on SSRI half-life, serum concentration, and the pharmacodynamics of other medications given during pregnancy and labor.24

Discontinuation syndromes can occur in SSRI-exposed neonates within a few hours or days after birth and last up to 1 month after delivery, depending on the infant’s susceptibility.25 Nearly two-thirds of suspected SSRI-induced neonatal withdrawal syndromes have been associated with paroxetine, although all SSRIs appear be associated with some risk.26 Several trials, including a recent prospective study, found prenatal antidepressant use associated with lower gestational age at birth and increased risk of preterm birth.27

A prospective study compared the effects of maternal SSRI use on behavioral state, sleep, motor activity, and heart rate variability in 17 exposed vs 17 nonexposed matched neonates. In the first 1 to 2 weeks of life, SSRI-exposed neonates showed:

  • greater tremulousness
  • less flexible and dampened state regulation
  • more time in uninterrupted REM sleep
  • more frequent startles or sudden arousals
  • greater generalized motor activity
  • greater autonomic dysregulation.28
In a cohort study of 60 neonates exposed to SSRIs in utero, 30% met diagnostic criteria for neonatal abstinence syndrome. The most common discontinuation symptoms were:

  • tremor (37/60)
  • GI disturbances (34/60)—including exaggerated sucking, poor feeding, regurgitation, vomiting, and loose stools
  • sleep disturbance (21/60).
Other symptoms included irritability, constant crying, shivering, increased tone, convulsions, jitteriness, poor gaze control, vomiting, myoclonus, and lethargy.25

Recommendations. The perception that SSRIs have low fetal toxicity has guided prescribing practices in recent years. Newer evidence shows, however, that fetal exposure to SSRIs may have some adverse effects, including lower birth weight and early delivery. First-trimester paroxetine use has been associated with increased risk for fetal ventricular and/or atrial septal defects.

Discuss these risks with the patient when you consider starting or continuing SSRI use during pregnancy.24 If you prescribe an SSRI, use the minimum effective dosage and avoid paroxetine during pregnancy.18

To reduce the risk for PPHN, early delivery, and neonatal withdrawal syndromes, taper and discontinue the SSRI during the third trimester. Restarting the SSRI soon after delivery is the most effective way to prevent recurrence of anxiety symptoms or postpartum depression.

Benzodiazepines

Teratogenicity. Like SSRIs, benzodiazepines cross the placenta to the fetus.29 Benzodiazepine teratogenicity remains controversial.8 Some—but not all—data show a small but significant increased risk for major malformations/oral cleft malformations with first-trimester benzodiazepine exposure.

A Medline literature search from 1966 to 2000 found not enough information to determine whether potential benefits of benzodiazepines to the mother outweigh risks to the fetus.29 An ambitious meta-analysis of >1,400 studies by Dolovich et al30 found a small association between fetal exposure to benzodiazepines and major malformations/cleft palate, but only in pooled data from case-controlled studies. No association was found between fetal exposure to benzodiazepines and malformations/cleft palate in pooled data from cohort studies.

A 32-month, hospital-based surveillance program of 28,565 births found no increase in the rate of major malformations in 43 infants exposed to clonazepam monotherapy—33 (77%) in the first trimester.31 Thus, the risk of major malformations/cleft palate with the use of benzodiazepines in the first trimester appears to be low.

Toxicity and withdrawal syndromes. Neonatal benzodiazepine toxicity and withdrawal syndromes have been reported in studies and case reports. Although these syndromes occur, they do not affect all infants with late third-trimester benzodiazepine exposure. Prevalence rates have not been calculated.32

  • Neonatal toxicity (“floppy infant syndrome”)—characterized by hypothermia, lethargy, poor respiratory effort, and feeding difficulties—occurs after maternal benzodiazepine use just before delivery.8
  • Neonatal withdrawal may be caused by very late, third trimester exposure to benzodiazepines. Symptoms—which can persist ≤3 months after delivery—include restlessness, irritability, abnormal sleep patterns, suckling difficulties, growth retardation, hypertonia, hyperreflexia, tremulousness, apnea, diarrhea, and vomiting.8,29
Recommendations. When possible, avoid benzodiazepines in the first trimester because of possible teratogenicity and then again late in the third trimester before delivery because of neonatal withdrawal syndromes. To reduce as much as possible the small risk of a benzodiazepine-related fetal malformation/cleft palate, wean the mother from benzodiazepines before conception. After the first trimester, the benzodiazepine can be restarted if necessary.29

To minimize neonatal withdrawal, gradually taper the mother’s benzodiazepine before delivery.29 Because the baby’s due date is calculated to be ±2 weeks before delivery, begin this taper 3 to 4 weeks before the due date and discontinue at least 1 week before delivery. Breastfeeding while taking benzodiazepines is not recommended because of the risk of over-sedating the infant.

 

 

A rational approach

Both benzodiazepines and SSRIs are associated with low but demonstrated risks to the fetus when used during pregnancy (Table 2).19,20,23,25,30,33 Use these medications to manage a patient’s anxiety only if the clinical benefit to the mother justifies the potential risks to the fetus.29

A staggered combination of SSRIs during the first 2 trimesters and benzodiazepines during the last 2 trimesters can help balance the risks and benefits of pharmacotherapy of anxiety disorders during pregnancy (Table 3).

Frankly discuss with your patient the risks and benefits in the context of her perceived need for symptom control to sustain her level of functioning. You could document this discussion in the progress note as “R, B, A, and pt C,” signifying that risks, benefits, and alternatives were discussed, and the patient consented. If possible, include the patient’s husband, partner, or parent in this discussion.

Table 2

Risks of SSRIs vs benzodiazepines during pregnancy stages

Pregnancy stage when givenFetal riskSSRIsBenzodiazepines
First trimester*TeratogenicityParoxetine use associated with 2-fold increased risk of major congenital anomalies and 3-fold increased risk of major cardiac anomalies;19 meta-analysis calculated significant risk of cardiac malformations (odds ratio 1.72; population prevalence = 13.4/1,000 births)20,33Meta-analysis of case control studies showed increased risk of major malformations/cleft palate (odds ratio 3.01; population prevalence = 10 to 20/1,000 births); no association seen in cohort studies30
Third trimesterPPHNCase control study showed 3.7% of infants with PPHN were exposed to SSRIs vs 0.7% of controls; adjusted odds ratio 6.1, absolute risk to exposed population = 6 to 12/1,000 births)23 
Perinatal and long-term effectsToxicity/withdrawal syndromesCohort study of 60 infants concluded prevalence of discontinuation syndromes is 30% in neonates with third trimester SSRI exposure25Neonatal toxicity (“floppy infant syndrome”) and neonatal withdrawal reported with maternal benzodiazepine use in late third trimester; prevalence unknown
 Preterm birth, serotonin withdrawal syndromes, CNS effects, long-term neurobehavioral effectsUnknownUnknown
PPHN: persistent pulmonary hypertension of the newborn; SSRIs: selective serotonin reuptake inhibitors
* Available data indicate that first-trimester exposure to SSRIs (other than paroxetine) and benzodiazepines may increase the relative risk for congenital anomalies, but the absolute risk of having a child with an anomaly is small.
Some case reports, but published literature is insufficient to determine prevalence or magnitude of risk.
Table 3

Staggered, combination therapy for anxiety disorders during pregnancy

Pregnancy stageRecommended to manage risks to mother and fetus
First trimester
  • SSRI (not paroxetine)
  • No benzodiazepines
  • Nondrug therapies*
Second trimester
  • SSRI (not paroxetine)
  • Can use benzodiazepine if needed
  • Nondrug therapies*
Third trimester
  • Taper off SSRI by 1 to 2 months before due date
  • Can use benzodiazepine until 2 weeks before due date
  • Nondrug therapies*
SSRI: selective serotonin reuptake inhibitor
* Nondrug therapies can include prenatal exercise, sleep hygiene, relaxation, and psychotherapy (cognitive-behavioral therapy, interpersonal therapy, supportive therapy, family/couples therapy)

CASE CONTINUED: CBT plus medication

Ms. K and her husband are open to adding weekly cognitive-behavioral therapy (CBT) for anxiety as long as she can continue her medications. You discuss the evidence regarding potential neonatal risks with paroxetine and clonazepam treatment. Because Ms. K is 6 weeks pregnant, you outline a plan for a rapid cross-taper off paroxetine and onto fluoxetine, 10 to 30 mg/d, explaining that paroxetine might pose a greater first-trimester risk of major congenital malformations and cardiac malformations. You discuss possible side effects of fluoxetine and explain a plan to taper off fluoxetine during the third trimester to reduce the risk of PPHN, early delivery, and withdrawal in the newborn.

Because Ms. K has been taking clonazepam at only 0.5 mg 1 to 2 times per week, you instruct her to stop taking the benzodiazepine for the next 6 weeks until she is through her first trimester. You also reassure her that she can use clonazepam after the first trimester, if necessary, as long as she agrees to taper off completely 1 to 2 weeks before to her due date.

You refer her to a CBT therapist and emphasize the importance of CBT, relaxation, and sleep hygiene—as well as support from her husband, family, and friends—to reduce her stress and facilitate the medication taper during her third trimester. You plan to see her monthly and co-manage her care with the CBT therapist and Ob/Gyn. You document this discussion in her medical record as evidence of informed consent.

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Paroxetine • Paxil
  • Fluoxetine • Prozac
  • Sertraline • Zoloft

Disclosures

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Ross LE, McLean LM. Anxiety disorders during pregnancy and the postpartum period: a systematic review. J Clin Psychiatry 2006;67(8):1285-98.

2. Labad J, Menchon JM, Alonso P, et al. Female reproductive cycle and obsessive-compulsive disorder. J Clin Psychiatry 2005;66(4):428-35.

3. Adewuya AO, Ola BA, Aloba OO, Mapayi BM. Anxiety disorders among Nigerian women in late pregnancy: a controlled study. Arch Womens Ment Health 2006;9(6):325-8.

4. Field T, Hernandez-Reif M, Diego M, et al. Stability of mood states and biochemistry across pregnancy. Infant Behav Dev 2006;29(2):262-7.

5. Teixeira JM, Fisk NM, Glover V. Association between maternal anxiety in pregnancy and increased uterine artery resistance index: cohort based study. BMJ 1999;318(7177):153-7.

6. Monk C, Myers MM, Sloan RP, et al. Effects of women’s stress-elicited physiological activity and chronic anxiety on fetal heart rate. J Dev Behav Pediatr 2003;24(1):32-8.

7. Egliston KA, McMahon C, Austin MP. Stress in pregnancy and infant HPA axis function: conceptual and methodological issues relating to the use of salivary cortisol as an outcome measure. Psychoneuroendocrinology 2007;32(1):1-13.

8. Levey L, Ragan K, Hower-Hartley A, et al. Psychiatric disorders in pregnancy. Neurol Clin 2004;22(4):863-93.

9. Oberlander TF, Reebye P, Misri S, et al. Externalizing and attentional behaviors in children of depressed mothers treated with a selective serotonin reuptake inhibitor antidepressant during pregnancy. Arch Pediatr Adolesc Med 2007;161(1):22-9.

10. Misri S, Reebye P, Kendrick K, et al. Internalizing behaviors in 4-year-old children exposed in utero to psychotropic medications. Am J Psychiatry 2006;163(6):1026-32.

11. Copper RL, Goldenberg RL, Das A, et al. The Preterm Prediction Study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks’ gestation. Am J Obstet Gynecol 1996;175(5):1286-92.

12. Sutter-Dallay AL, Giaconne-Marcesche V, Glatigny-Dallay E, Verdoux H. Women with anxiety disorders during pregnancy are at increased risk of intense postnatal depressive symptoms: a prospective survey of the MATQUID cohort. Eur Psychiatry 2004;19(8):459-63.

13. Nierop A, Bratsikas A, Zimmermann R, Ehlert U. Are stress-induced cortisol changes during pregnancy associated with postpartum depressive symptoms? Psychosom Med 2006;68(6):931-7.

14. Weissman MM. Recent non-medication trials of interpersonal psychotherapy for depression. Int J Neuropsychopharmacology 2007;10(1):117-22.

15. Ward RK, Zamorski MA. Benefits and risks of psychiatric medications during pregnancy. Am Fam Physician 2002;66(4):629-36.

16. Bastani F, Hidarnia A, Montgomery KS, et al. Does relaxation education in anxious primigravid Iranian women influence adverse pregnancy outcomes? A randomized controlled trial. J Perinat Neonatal Nurs 2006;20(2):138-46.

17. Fricchione G. Generalized anxiety disorder. N Engl J Med 2004;351(7):675-82.

18. Källén BA, Otterblad Olausson P. Maternal use of selective serotonin re-uptake inhibitors in early pregnancy and infant congenital malformations. Birth Defects Res A Clin Mol Teratol 2007;79(4):301-8.

19. Berard A, Ramos E, Rey E, et al. First trimester exposure to paroxetine and risk of cardiac malformations in infants: the importance of dosage. Birth Defects Res B Dev Reprod Toxicol 2007;80(1):18-27.

20. Bar-Oz B, Einarson T, Einarson A, et al. Paroxetine and congenital malformations: meta-analysis and consideration of potential confounding factors. Clin Ther 2005;29(5):918-26.

21. Malm H, Klaukka T, Neuvonen PJ. Risks associated with selective serotonin reuptake inhibitors in pregnancy. Obstet Gynecol 2005;106(6):1289-96.

22. Nulman I, Rovet J, Stewart DE, et al. Child development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: a prospective, controlled study. Am J Psychiatry 2002;159(11):1889-95.

23. Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med 2006;354(6):579-87.

24. Haddad PM, Pal BR, Clarke P, et al. Neonatal symptoms following maternal paroxetine treatment: serotonin toxicity or paroxetine discontinuation syndrome? J Psychopharmacology 2005;19(5):554-7.

25. Levinson-Castiel R, Merlob P, Linder N, et al. Neonatal abstinence syndrome after in utero exposure to selective serotonin reuptake inhibitors in term infants. Arch Pediatr Adolesc Med 2006;160(2):173-6.

26. Sanz EJ, De-las-Cuevas C, Kiuru A, et al. Selective serotonin reuptake inhibitors in pregnant women and neonatal withdrawal syndrome: a database analysis. Lancet 2005;365(9458):482-7.

27. Suri R, Altshuler L, Hellemann G, et al. Effects of antenatal depression and antidepressant treatment on gestational age at birth and risk of preterm birth. Am J Psychiatry 2007;164(8):1206-13.

28. Zeskind PS, Stephens LE. Maternal selective serotonin reuptake inhibitor use during pregnancy and newborn neurobehavior. Pediatrics 2004;113(2):368-75.

29. Iqbal MM, Sobhan T, Ryals T. Effects of commonly used benzodiazepines on the fetus, the neonate, and the nursing infant. Psychiatr Serv 2002;53(1):39-49.

30. Dolovich LR, Addis A, Vaillancourt JM, et al. Benzodiazepine use in pregnancy and major malformations or oral cleft: meta-analysis of cohort and case-control studies. BMJ 1998;317(7162):839-43.

31. McElhatton PR. The effects of benzodiazepine use during pregnancy. Reprod Toxicol 1994;8(6):461-75.

32. Lin AE, Peller AJ, Westgate MN, et al. Clonazepam use in pregnancy and the risk of malformations. Birth Defects Res A Clin Mol Teratol 2004;70(8):534-6.

33. Levy M, James MS, Erickson JD, McClearn AB. Prevalence of birth defects. Birth outcomes Centers for Disease Control and Prevention. Available at: http://www.cdc.gov/reproductivehealth/Products&Pubs/DatatoAction/pdf/birout4.pdf. Accessed January 9, 2008.

Article PDF
Author and Disclosure Information

Deborah B. Raphael, MD
Resident
Department of psychiatry, University of California, San Francisco

Jessica Ross, MD
Postdoctoral fellow
Department of psychiatry, University of California, San Francisco

Louann Brizendine, MD
Clinical professor, director, Women’s Mood and Hormone Clinic
Department of psychiatry, University of California, San Francisco

Issue
Current Psychiatry - 07(02)
Publications
Page Number
39-52
Legacy Keywords
anxiety during pregnancy; SSRIs in pregnancy; Deborah B. Raphael MD; Jessica Ross MD; Louann Brizendine MD; clonazepam; paroxetine; generalized anxiety disorder; posttraumatic stress disorder; PTSD; obsessive-compulsive disorder; OCD; maternal anxiety; teratogenicity; fetal malformations from SSRIs; maternal SSRI use
Sections
Author and Disclosure Information

Deborah B. Raphael, MD
Resident
Department of psychiatry, University of California, San Francisco

Jessica Ross, MD
Postdoctoral fellow
Department of psychiatry, University of California, San Francisco

Louann Brizendine, MD
Clinical professor, director, Women’s Mood and Hormone Clinic
Department of psychiatry, University of California, San Francisco

Author and Disclosure Information

Deborah B. Raphael, MD
Resident
Department of psychiatry, University of California, San Francisco

Jessica Ross, MD
Postdoctoral fellow
Department of psychiatry, University of California, San Francisco

Louann Brizendine, MD
Clinical professor, director, Women’s Mood and Hormone Clinic
Department of psychiatry, University of California, San Francisco

Article PDF
Article PDF

Ms. K, age 25, is 6 weeks pregnant and is taking medications for generalized anxiety disorder (GAD). When she was diagnosed with GAD at age 19, her symptoms included 6 months of excessive anxiety—insomnia, fatigue, difficulty with concentration, and psychomotor agitation—without mood symptoms. These symptoms interfered greatly with her schoolwork and other daily activities.

For 6 years Ms. K has been taking the selective serotonin reuptake inhibitor (SSRI) paroxetine, 15 mg/d, and the benzodiazepine clonazepam, 0.5 mg as needed, with good symptom control. Now that she is pregnant and her primary care doctor has refused to continue these medications, she is seeking treatment and advice.

Not enough is known about how to safely treat anxiety disorders during pregnancy, and physicians are not sure what to do with patients such as Ms. K. Without evidence-based guidelines, we feel anxious about potential risks to mother and fetus as we try to provide appropriate drug therapy.

To help you and your patients weigh the risks and benefits of perinatal treatments for anxiety disorders, this article briefly summarizes the evidence on:

  • anxiety disorders’ natural history during pregnancy
  • how untreated maternal anxiety affects the fetus
  • nonpharmacologic therapies for anxiety disorders
  • a plan to manage fetal risks by staggering SSRI and benzodiazepine use during the first and third trimesters.

Anxiety during pregnancy

Nearly one-third of women experience an anxiety disorder during their lives, with peak onset during childbearing years.1,2 Compared with research on perinatal depression, far fewer studies have examined anxiety disorders’ onset, presentation, prevalence, and treatment.1

The literature includes no studies of the course of preexisting GAD or posttraumatic stress disorder (PTSD) and no evidence that symptoms of preexisting obsessive-compulsive disorder (OCD) change during pregnancy. Some studies of panic disorder show symptoms improving during pregnancy, whereas others do not (Table 1).1

One small study done in late pregnancy found a significant association between the prevalence of an anxiety disorder, maternal primiparity, and comorbid medical conditions. Thus, a woman in her first pregnancy may be at increased risk to develop an anxiety disorder if she has a comorbid medical condition.3 As in the case of Ms. K, however, continuation of preexisting anxiety appears more likely than onset of a new anxiety disorder during pregnancy.

Table 1

How pregnancy affects the course of 4 anxiety disorders

DisorderPrevalenceEffect
Generalized anxiety disorder (GAD)8.5% of women experience GAD during the third trimester, compared with a 5% prevalence in the general populationNo studies have reported on the course of GAD in pregnant women with preexisting disorder
Obsessive-compulsive disorder (OCD)2% to 12% of OCD outpatients of childbearing age report onset during pregnancyPreexisting OCD usually shows no change during pregnancy but may worsen postpartum
Panic disorder (PD)1.3% to 2% in pregnant women, compared with 1.5% to 3.5% in the general populationPanic symptoms in women with preexisting PD may improve during pregnancy and worsen postpartum
Posttraumatic stress disorder (PTSD)2.3% to 7.7% in pregnant women and 0% to 6.9% postpartum, compared with 1% to 14% in the communityNo studies have reported on the course of PTSD in pregnant women with preexisting disorder
Source: References 1,2

Fetal risks from maternal anxiety

Fetal risk from severe maternal anxiety is not zero. Offspring born to high-anxiety mothers exhibit neurobehavioral differences compared with offspring of calmer mothers. Changes in high-anxiety mothers’ offspring include:

  • altered EEG activation and vagal tone
  • increased time in deep sleep and less time in active alert states
  • lower performance on the Brazelton Neonatal Behavior Assessment Scale.4
A cohort study by Teixeira et al5 found an association between maternal anxiety in pregnancy and uterine artery resistance, suggesting a possible mechanism by which a mother’s psychologic state may affect fetal development. High anxiety and self-reported life stress during pregnancy also are associated consistently with abnormal, high-frequency heart rate variability in infants—a finding linked with negative infant behavior and later adult hostility.6

Exposure to maternal high anxiety has been associated with mental developmental delays in infants and increased risk for behavioral and emotional problems in young children.7-10 Anxiety may not directly cause intrauterine growth retardation and preterm delivery, but it is significantly associated with prenatal tobacco, alcohol, and narcotics use—which predicts these and other negative neonatal outcomes.11

Anxiety during pregnancy is a risk factor for postnatal depressive symptoms, independent of depressed mood and family or marital stressors during pregnancy.12 Mothers with postpartum depression appear less able to respond sensitively and competently to their newborns, and these infants may be at increased risk of behavioral, emotional, and cognitive problems.
 

 

7,13

CASE CONTINUED: ‘Stay the course’

Ms. K worries that she could not tolerate recurrence of her anxiety symptoms and wishes to continue both medications. Her husband concurs, but they want to minimize potential risks to their baby. You discuss the options for treating anxiety symptoms during pregnancy, including medications, psychotherapy, and behavioral treatments.

Treatment decisions

Ideally you’ll begin treating anxiety disorders in women of childbearing age with preconception psychoeducation. Explaining the risks of medications if she were to become pregnant and asking about the contraception she is using are de rigueur. Psychotherapy is low risk to the fetus and is considered first choice for treating mild to moderate anxiety in women of childbearing age who plan to become pregnant (Box).1,14-17

Box

Psychotherapy: First choice for anxiety during pregnancy

No studies directly address the efficacy or outcome of any psychotherapy for anxiety in pregnancy. Even so:

  • For mild to moderate anxiety, psychotherapy is the first-line treatment for pregnant women.
  • Interpersonal psychotherapy (IPT) without medications can reduce depressive symptoms in pregnant women with depression.14
  • Cognitive-behavioral therapy (CBT) without medications has shown efficacy for anxiety disorders in psychiatric populations.15,16

Because no evidence suggests that pregnant women require different psychotherapeutic recommendations than other psychiatric patients, consider a course of CBT that targets anxiety symptoms or IPT for a pregnant patient with an anxiety disorder.

Relaxation therapy also has shown efficacy in treating anxiety disorders. In a randomized controlled trial of 110 pregnant women with high-level anxiety, 7 weeks of applied relaxation training sessions was associated with significant reductions in low-weight births, cesarean sections, and instrumental extractions.16,17

Because poor marital relationships are consistent psychosocial predictors of anxiety during pregnancy and postpartum depression,1 recommend family or marital therapy when appropriate.

Psychotherapy alone is inadequate, however, for the many patients—such as Ms. K—who present already pregnant with a history of moderate to severe anxiety. Adjunctive psychotropic therapy—along with various nonmedication therapies—is warranted for patients whose social or occupational functioning would be substantially impaired by suboptimal control of anxiety during pregnancy.

Because Ms. K wishes to continue taking paroxetine and clonazepam, what can you tell her about the risks and benefits of SSRIs and benzodiazepines during pregnancy?

SSRIs in pregnancy

Teratogenicity. Compared with benzodiazepines, SSRIs have been considered agents of choice for use during pregnancy because of a lower risk of teratogenic effects.15 Paroxetine, however, appears to pose a greater risk for teratogenicity than other SSRIs.

An increased risk for fetal ventricular and/or atrial septal defects has been associated with first-trimester exposure to paroxetine, but no other SSRI.18 First trimester exposure to paroxetine at doses averaging 25 mg/d has been associated with statistically significant risks of major congenital anomalies (2-fold increase) and major cardiac anomalies (3-fold increase),19 although other studies have failed to reproduce this finding. A meta-analysis of 7 studies by Bar-Oz et al20 found an association between first-trimester paroxetine exposure and a significant increase in risk for cardiac malformations (odds ratio [OR] 1.72; 95% CI,1.22-2.42).

The overall rate of fetal malformations from SSRIs appears to be low, although most studies have examined only fluoxetine or paroxetine. Some studies have reported various malformations with fluoxetine or sertraline, but others have not. In Finland, a population-based study found no increase in rate of major congenital malformations in offspring of 1,782 women who filled prescriptions for SSRIs during pregnancy, compared with the general population rate of 1% to 3%.21

Neurobehavioral effects. SSRI exposure during fetal life has shown no long-term neurobehavioral effects. A blinded prospective study by Nulman et al22 found no differences in global IQ scores, language development, or behavioral development among children age ≤5 who were exposed in utero to fluoxetine (n=40) or a tricyclic antidepressant (n=46), compared with unexposed children of nondepressed mothers (n=36). Similarly, using reports from teachers and clinical measures of internalizing behaviors, Misri et al10 found no increase in depression, anxiety, or withdrawal in 4-year-olds with prenatal exposure to SSRIs (n=22), compared with nonexposed children (n=14).

Pulmonary hypertension. SSRI exposure in later pregnancy may increase the rate of persistent pulmonary hypertension of the newborn (PPHN), which occurs in 1 to 2 infants per 1,000 live births. PPHN showed a statistically significant association with late prenatal SSRI exposure (OR 6.1) in a study that controlled for maternal smoking, body mass index, and diabetes.23 PPHN occurred in approximately 1% of infants exposed to SSRIs in late pregnancy. PPHN rates were not affected by maternal depression/anxiety, non-SSRI antidepressant exposure throughout pregnancy, or SSRI exposure during early pregnancy only.

 

 

Toxicity and withdrawal syndromes. Infants of women who continue to take SSRIs just before delivery can develop toxicity or withdrawal syndromes. Occurrence of either syndrome depends on SSRI half-life, serum concentration, and the pharmacodynamics of other medications given during pregnancy and labor.24

Discontinuation syndromes can occur in SSRI-exposed neonates within a few hours or days after birth and last up to 1 month after delivery, depending on the infant’s susceptibility.25 Nearly two-thirds of suspected SSRI-induced neonatal withdrawal syndromes have been associated with paroxetine, although all SSRIs appear be associated with some risk.26 Several trials, including a recent prospective study, found prenatal antidepressant use associated with lower gestational age at birth and increased risk of preterm birth.27

A prospective study compared the effects of maternal SSRI use on behavioral state, sleep, motor activity, and heart rate variability in 17 exposed vs 17 nonexposed matched neonates. In the first 1 to 2 weeks of life, SSRI-exposed neonates showed:

  • greater tremulousness
  • less flexible and dampened state regulation
  • more time in uninterrupted REM sleep
  • more frequent startles or sudden arousals
  • greater generalized motor activity
  • greater autonomic dysregulation.28
In a cohort study of 60 neonates exposed to SSRIs in utero, 30% met diagnostic criteria for neonatal abstinence syndrome. The most common discontinuation symptoms were:

  • tremor (37/60)
  • GI disturbances (34/60)—including exaggerated sucking, poor feeding, regurgitation, vomiting, and loose stools
  • sleep disturbance (21/60).
Other symptoms included irritability, constant crying, shivering, increased tone, convulsions, jitteriness, poor gaze control, vomiting, myoclonus, and lethargy.25

Recommendations. The perception that SSRIs have low fetal toxicity has guided prescribing practices in recent years. Newer evidence shows, however, that fetal exposure to SSRIs may have some adverse effects, including lower birth weight and early delivery. First-trimester paroxetine use has been associated with increased risk for fetal ventricular and/or atrial septal defects.

Discuss these risks with the patient when you consider starting or continuing SSRI use during pregnancy.24 If you prescribe an SSRI, use the minimum effective dosage and avoid paroxetine during pregnancy.18

To reduce the risk for PPHN, early delivery, and neonatal withdrawal syndromes, taper and discontinue the SSRI during the third trimester. Restarting the SSRI soon after delivery is the most effective way to prevent recurrence of anxiety symptoms or postpartum depression.

Benzodiazepines

Teratogenicity. Like SSRIs, benzodiazepines cross the placenta to the fetus.29 Benzodiazepine teratogenicity remains controversial.8 Some—but not all—data show a small but significant increased risk for major malformations/oral cleft malformations with first-trimester benzodiazepine exposure.

A Medline literature search from 1966 to 2000 found not enough information to determine whether potential benefits of benzodiazepines to the mother outweigh risks to the fetus.29 An ambitious meta-analysis of >1,400 studies by Dolovich et al30 found a small association between fetal exposure to benzodiazepines and major malformations/cleft palate, but only in pooled data from case-controlled studies. No association was found between fetal exposure to benzodiazepines and malformations/cleft palate in pooled data from cohort studies.

A 32-month, hospital-based surveillance program of 28,565 births found no increase in the rate of major malformations in 43 infants exposed to clonazepam monotherapy—33 (77%) in the first trimester.31 Thus, the risk of major malformations/cleft palate with the use of benzodiazepines in the first trimester appears to be low.

Toxicity and withdrawal syndromes. Neonatal benzodiazepine toxicity and withdrawal syndromes have been reported in studies and case reports. Although these syndromes occur, they do not affect all infants with late third-trimester benzodiazepine exposure. Prevalence rates have not been calculated.32

  • Neonatal toxicity (“floppy infant syndrome”)—characterized by hypothermia, lethargy, poor respiratory effort, and feeding difficulties—occurs after maternal benzodiazepine use just before delivery.8
  • Neonatal withdrawal may be caused by very late, third trimester exposure to benzodiazepines. Symptoms—which can persist ≤3 months after delivery—include restlessness, irritability, abnormal sleep patterns, suckling difficulties, growth retardation, hypertonia, hyperreflexia, tremulousness, apnea, diarrhea, and vomiting.8,29
Recommendations. When possible, avoid benzodiazepines in the first trimester because of possible teratogenicity and then again late in the third trimester before delivery because of neonatal withdrawal syndromes. To reduce as much as possible the small risk of a benzodiazepine-related fetal malformation/cleft palate, wean the mother from benzodiazepines before conception. After the first trimester, the benzodiazepine can be restarted if necessary.29

To minimize neonatal withdrawal, gradually taper the mother’s benzodiazepine before delivery.29 Because the baby’s due date is calculated to be ±2 weeks before delivery, begin this taper 3 to 4 weeks before the due date and discontinue at least 1 week before delivery. Breastfeeding while taking benzodiazepines is not recommended because of the risk of over-sedating the infant.

 

 

A rational approach

Both benzodiazepines and SSRIs are associated with low but demonstrated risks to the fetus when used during pregnancy (Table 2).19,20,23,25,30,33 Use these medications to manage a patient’s anxiety only if the clinical benefit to the mother justifies the potential risks to the fetus.29

A staggered combination of SSRIs during the first 2 trimesters and benzodiazepines during the last 2 trimesters can help balance the risks and benefits of pharmacotherapy of anxiety disorders during pregnancy (Table 3).

Frankly discuss with your patient the risks and benefits in the context of her perceived need for symptom control to sustain her level of functioning. You could document this discussion in the progress note as “R, B, A, and pt C,” signifying that risks, benefits, and alternatives were discussed, and the patient consented. If possible, include the patient’s husband, partner, or parent in this discussion.

Table 2

Risks of SSRIs vs benzodiazepines during pregnancy stages

Pregnancy stage when givenFetal riskSSRIsBenzodiazepines
First trimester*TeratogenicityParoxetine use associated with 2-fold increased risk of major congenital anomalies and 3-fold increased risk of major cardiac anomalies;19 meta-analysis calculated significant risk of cardiac malformations (odds ratio 1.72; population prevalence = 13.4/1,000 births)20,33Meta-analysis of case control studies showed increased risk of major malformations/cleft palate (odds ratio 3.01; population prevalence = 10 to 20/1,000 births); no association seen in cohort studies30
Third trimesterPPHNCase control study showed 3.7% of infants with PPHN were exposed to SSRIs vs 0.7% of controls; adjusted odds ratio 6.1, absolute risk to exposed population = 6 to 12/1,000 births)23 
Perinatal and long-term effectsToxicity/withdrawal syndromesCohort study of 60 infants concluded prevalence of discontinuation syndromes is 30% in neonates with third trimester SSRI exposure25Neonatal toxicity (“floppy infant syndrome”) and neonatal withdrawal reported with maternal benzodiazepine use in late third trimester; prevalence unknown
 Preterm birth, serotonin withdrawal syndromes, CNS effects, long-term neurobehavioral effectsUnknownUnknown
PPHN: persistent pulmonary hypertension of the newborn; SSRIs: selective serotonin reuptake inhibitors
* Available data indicate that first-trimester exposure to SSRIs (other than paroxetine) and benzodiazepines may increase the relative risk for congenital anomalies, but the absolute risk of having a child with an anomaly is small.
Some case reports, but published literature is insufficient to determine prevalence or magnitude of risk.
Table 3

Staggered, combination therapy for anxiety disorders during pregnancy

Pregnancy stageRecommended to manage risks to mother and fetus
First trimester
  • SSRI (not paroxetine)
  • No benzodiazepines
  • Nondrug therapies*
Second trimester
  • SSRI (not paroxetine)
  • Can use benzodiazepine if needed
  • Nondrug therapies*
Third trimester
  • Taper off SSRI by 1 to 2 months before due date
  • Can use benzodiazepine until 2 weeks before due date
  • Nondrug therapies*
SSRI: selective serotonin reuptake inhibitor
* Nondrug therapies can include prenatal exercise, sleep hygiene, relaxation, and psychotherapy (cognitive-behavioral therapy, interpersonal therapy, supportive therapy, family/couples therapy)

CASE CONTINUED: CBT plus medication

Ms. K and her husband are open to adding weekly cognitive-behavioral therapy (CBT) for anxiety as long as she can continue her medications. You discuss the evidence regarding potential neonatal risks with paroxetine and clonazepam treatment. Because Ms. K is 6 weeks pregnant, you outline a plan for a rapid cross-taper off paroxetine and onto fluoxetine, 10 to 30 mg/d, explaining that paroxetine might pose a greater first-trimester risk of major congenital malformations and cardiac malformations. You discuss possible side effects of fluoxetine and explain a plan to taper off fluoxetine during the third trimester to reduce the risk of PPHN, early delivery, and withdrawal in the newborn.

Because Ms. K has been taking clonazepam at only 0.5 mg 1 to 2 times per week, you instruct her to stop taking the benzodiazepine for the next 6 weeks until she is through her first trimester. You also reassure her that she can use clonazepam after the first trimester, if necessary, as long as she agrees to taper off completely 1 to 2 weeks before to her due date.

You refer her to a CBT therapist and emphasize the importance of CBT, relaxation, and sleep hygiene—as well as support from her husband, family, and friends—to reduce her stress and facilitate the medication taper during her third trimester. You plan to see her monthly and co-manage her care with the CBT therapist and Ob/Gyn. You document this discussion in her medical record as evidence of informed consent.

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Paroxetine • Paxil
  • Fluoxetine • Prozac
  • Sertraline • Zoloft

Disclosures

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Ms. K, age 25, is 6 weeks pregnant and is taking medications for generalized anxiety disorder (GAD). When she was diagnosed with GAD at age 19, her symptoms included 6 months of excessive anxiety—insomnia, fatigue, difficulty with concentration, and psychomotor agitation—without mood symptoms. These symptoms interfered greatly with her schoolwork and other daily activities.

For 6 years Ms. K has been taking the selective serotonin reuptake inhibitor (SSRI) paroxetine, 15 mg/d, and the benzodiazepine clonazepam, 0.5 mg as needed, with good symptom control. Now that she is pregnant and her primary care doctor has refused to continue these medications, she is seeking treatment and advice.

Not enough is known about how to safely treat anxiety disorders during pregnancy, and physicians are not sure what to do with patients such as Ms. K. Without evidence-based guidelines, we feel anxious about potential risks to mother and fetus as we try to provide appropriate drug therapy.

To help you and your patients weigh the risks and benefits of perinatal treatments for anxiety disorders, this article briefly summarizes the evidence on:

  • anxiety disorders’ natural history during pregnancy
  • how untreated maternal anxiety affects the fetus
  • nonpharmacologic therapies for anxiety disorders
  • a plan to manage fetal risks by staggering SSRI and benzodiazepine use during the first and third trimesters.

Anxiety during pregnancy

Nearly one-third of women experience an anxiety disorder during their lives, with peak onset during childbearing years.1,2 Compared with research on perinatal depression, far fewer studies have examined anxiety disorders’ onset, presentation, prevalence, and treatment.1

The literature includes no studies of the course of preexisting GAD or posttraumatic stress disorder (PTSD) and no evidence that symptoms of preexisting obsessive-compulsive disorder (OCD) change during pregnancy. Some studies of panic disorder show symptoms improving during pregnancy, whereas others do not (Table 1).1

One small study done in late pregnancy found a significant association between the prevalence of an anxiety disorder, maternal primiparity, and comorbid medical conditions. Thus, a woman in her first pregnancy may be at increased risk to develop an anxiety disorder if she has a comorbid medical condition.3 As in the case of Ms. K, however, continuation of preexisting anxiety appears more likely than onset of a new anxiety disorder during pregnancy.

Table 1

How pregnancy affects the course of 4 anxiety disorders

DisorderPrevalenceEffect
Generalized anxiety disorder (GAD)8.5% of women experience GAD during the third trimester, compared with a 5% prevalence in the general populationNo studies have reported on the course of GAD in pregnant women with preexisting disorder
Obsessive-compulsive disorder (OCD)2% to 12% of OCD outpatients of childbearing age report onset during pregnancyPreexisting OCD usually shows no change during pregnancy but may worsen postpartum
Panic disorder (PD)1.3% to 2% in pregnant women, compared with 1.5% to 3.5% in the general populationPanic symptoms in women with preexisting PD may improve during pregnancy and worsen postpartum
Posttraumatic stress disorder (PTSD)2.3% to 7.7% in pregnant women and 0% to 6.9% postpartum, compared with 1% to 14% in the communityNo studies have reported on the course of PTSD in pregnant women with preexisting disorder
Source: References 1,2

Fetal risks from maternal anxiety

Fetal risk from severe maternal anxiety is not zero. Offspring born to high-anxiety mothers exhibit neurobehavioral differences compared with offspring of calmer mothers. Changes in high-anxiety mothers’ offspring include:

  • altered EEG activation and vagal tone
  • increased time in deep sleep and less time in active alert states
  • lower performance on the Brazelton Neonatal Behavior Assessment Scale.4
A cohort study by Teixeira et al5 found an association between maternal anxiety in pregnancy and uterine artery resistance, suggesting a possible mechanism by which a mother’s psychologic state may affect fetal development. High anxiety and self-reported life stress during pregnancy also are associated consistently with abnormal, high-frequency heart rate variability in infants—a finding linked with negative infant behavior and later adult hostility.6

Exposure to maternal high anxiety has been associated with mental developmental delays in infants and increased risk for behavioral and emotional problems in young children.7-10 Anxiety may not directly cause intrauterine growth retardation and preterm delivery, but it is significantly associated with prenatal tobacco, alcohol, and narcotics use—which predicts these and other negative neonatal outcomes.11

Anxiety during pregnancy is a risk factor for postnatal depressive symptoms, independent of depressed mood and family or marital stressors during pregnancy.12 Mothers with postpartum depression appear less able to respond sensitively and competently to their newborns, and these infants may be at increased risk of behavioral, emotional, and cognitive problems.
 

 

7,13

CASE CONTINUED: ‘Stay the course’

Ms. K worries that she could not tolerate recurrence of her anxiety symptoms and wishes to continue both medications. Her husband concurs, but they want to minimize potential risks to their baby. You discuss the options for treating anxiety symptoms during pregnancy, including medications, psychotherapy, and behavioral treatments.

Treatment decisions

Ideally you’ll begin treating anxiety disorders in women of childbearing age with preconception psychoeducation. Explaining the risks of medications if she were to become pregnant and asking about the contraception she is using are de rigueur. Psychotherapy is low risk to the fetus and is considered first choice for treating mild to moderate anxiety in women of childbearing age who plan to become pregnant (Box).1,14-17

Box

Psychotherapy: First choice for anxiety during pregnancy

No studies directly address the efficacy or outcome of any psychotherapy for anxiety in pregnancy. Even so:

  • For mild to moderate anxiety, psychotherapy is the first-line treatment for pregnant women.
  • Interpersonal psychotherapy (IPT) without medications can reduce depressive symptoms in pregnant women with depression.14
  • Cognitive-behavioral therapy (CBT) without medications has shown efficacy for anxiety disorders in psychiatric populations.15,16

Because no evidence suggests that pregnant women require different psychotherapeutic recommendations than other psychiatric patients, consider a course of CBT that targets anxiety symptoms or IPT for a pregnant patient with an anxiety disorder.

Relaxation therapy also has shown efficacy in treating anxiety disorders. In a randomized controlled trial of 110 pregnant women with high-level anxiety, 7 weeks of applied relaxation training sessions was associated with significant reductions in low-weight births, cesarean sections, and instrumental extractions.16,17

Because poor marital relationships are consistent psychosocial predictors of anxiety during pregnancy and postpartum depression,1 recommend family or marital therapy when appropriate.

Psychotherapy alone is inadequate, however, for the many patients—such as Ms. K—who present already pregnant with a history of moderate to severe anxiety. Adjunctive psychotropic therapy—along with various nonmedication therapies—is warranted for patients whose social or occupational functioning would be substantially impaired by suboptimal control of anxiety during pregnancy.

Because Ms. K wishes to continue taking paroxetine and clonazepam, what can you tell her about the risks and benefits of SSRIs and benzodiazepines during pregnancy?

SSRIs in pregnancy

Teratogenicity. Compared with benzodiazepines, SSRIs have been considered agents of choice for use during pregnancy because of a lower risk of teratogenic effects.15 Paroxetine, however, appears to pose a greater risk for teratogenicity than other SSRIs.

An increased risk for fetal ventricular and/or atrial septal defects has been associated with first-trimester exposure to paroxetine, but no other SSRI.18 First trimester exposure to paroxetine at doses averaging 25 mg/d has been associated with statistically significant risks of major congenital anomalies (2-fold increase) and major cardiac anomalies (3-fold increase),19 although other studies have failed to reproduce this finding. A meta-analysis of 7 studies by Bar-Oz et al20 found an association between first-trimester paroxetine exposure and a significant increase in risk for cardiac malformations (odds ratio [OR] 1.72; 95% CI,1.22-2.42).

The overall rate of fetal malformations from SSRIs appears to be low, although most studies have examined only fluoxetine or paroxetine. Some studies have reported various malformations with fluoxetine or sertraline, but others have not. In Finland, a population-based study found no increase in rate of major congenital malformations in offspring of 1,782 women who filled prescriptions for SSRIs during pregnancy, compared with the general population rate of 1% to 3%.21

Neurobehavioral effects. SSRI exposure during fetal life has shown no long-term neurobehavioral effects. A blinded prospective study by Nulman et al22 found no differences in global IQ scores, language development, or behavioral development among children age ≤5 who were exposed in utero to fluoxetine (n=40) or a tricyclic antidepressant (n=46), compared with unexposed children of nondepressed mothers (n=36). Similarly, using reports from teachers and clinical measures of internalizing behaviors, Misri et al10 found no increase in depression, anxiety, or withdrawal in 4-year-olds with prenatal exposure to SSRIs (n=22), compared with nonexposed children (n=14).

Pulmonary hypertension. SSRI exposure in later pregnancy may increase the rate of persistent pulmonary hypertension of the newborn (PPHN), which occurs in 1 to 2 infants per 1,000 live births. PPHN showed a statistically significant association with late prenatal SSRI exposure (OR 6.1) in a study that controlled for maternal smoking, body mass index, and diabetes.23 PPHN occurred in approximately 1% of infants exposed to SSRIs in late pregnancy. PPHN rates were not affected by maternal depression/anxiety, non-SSRI antidepressant exposure throughout pregnancy, or SSRI exposure during early pregnancy only.

 

 

Toxicity and withdrawal syndromes. Infants of women who continue to take SSRIs just before delivery can develop toxicity or withdrawal syndromes. Occurrence of either syndrome depends on SSRI half-life, serum concentration, and the pharmacodynamics of other medications given during pregnancy and labor.24

Discontinuation syndromes can occur in SSRI-exposed neonates within a few hours or days after birth and last up to 1 month after delivery, depending on the infant’s susceptibility.25 Nearly two-thirds of suspected SSRI-induced neonatal withdrawal syndromes have been associated with paroxetine, although all SSRIs appear be associated with some risk.26 Several trials, including a recent prospective study, found prenatal antidepressant use associated with lower gestational age at birth and increased risk of preterm birth.27

A prospective study compared the effects of maternal SSRI use on behavioral state, sleep, motor activity, and heart rate variability in 17 exposed vs 17 nonexposed matched neonates. In the first 1 to 2 weeks of life, SSRI-exposed neonates showed:

  • greater tremulousness
  • less flexible and dampened state regulation
  • more time in uninterrupted REM sleep
  • more frequent startles or sudden arousals
  • greater generalized motor activity
  • greater autonomic dysregulation.28
In a cohort study of 60 neonates exposed to SSRIs in utero, 30% met diagnostic criteria for neonatal abstinence syndrome. The most common discontinuation symptoms were:

  • tremor (37/60)
  • GI disturbances (34/60)—including exaggerated sucking, poor feeding, regurgitation, vomiting, and loose stools
  • sleep disturbance (21/60).
Other symptoms included irritability, constant crying, shivering, increased tone, convulsions, jitteriness, poor gaze control, vomiting, myoclonus, and lethargy.25

Recommendations. The perception that SSRIs have low fetal toxicity has guided prescribing practices in recent years. Newer evidence shows, however, that fetal exposure to SSRIs may have some adverse effects, including lower birth weight and early delivery. First-trimester paroxetine use has been associated with increased risk for fetal ventricular and/or atrial septal defects.

Discuss these risks with the patient when you consider starting or continuing SSRI use during pregnancy.24 If you prescribe an SSRI, use the minimum effective dosage and avoid paroxetine during pregnancy.18

To reduce the risk for PPHN, early delivery, and neonatal withdrawal syndromes, taper and discontinue the SSRI during the third trimester. Restarting the SSRI soon after delivery is the most effective way to prevent recurrence of anxiety symptoms or postpartum depression.

Benzodiazepines

Teratogenicity. Like SSRIs, benzodiazepines cross the placenta to the fetus.29 Benzodiazepine teratogenicity remains controversial.8 Some—but not all—data show a small but significant increased risk for major malformations/oral cleft malformations with first-trimester benzodiazepine exposure.

A Medline literature search from 1966 to 2000 found not enough information to determine whether potential benefits of benzodiazepines to the mother outweigh risks to the fetus.29 An ambitious meta-analysis of >1,400 studies by Dolovich et al30 found a small association between fetal exposure to benzodiazepines and major malformations/cleft palate, but only in pooled data from case-controlled studies. No association was found between fetal exposure to benzodiazepines and malformations/cleft palate in pooled data from cohort studies.

A 32-month, hospital-based surveillance program of 28,565 births found no increase in the rate of major malformations in 43 infants exposed to clonazepam monotherapy—33 (77%) in the first trimester.31 Thus, the risk of major malformations/cleft palate with the use of benzodiazepines in the first trimester appears to be low.

Toxicity and withdrawal syndromes. Neonatal benzodiazepine toxicity and withdrawal syndromes have been reported in studies and case reports. Although these syndromes occur, they do not affect all infants with late third-trimester benzodiazepine exposure. Prevalence rates have not been calculated.32

  • Neonatal toxicity (“floppy infant syndrome”)—characterized by hypothermia, lethargy, poor respiratory effort, and feeding difficulties—occurs after maternal benzodiazepine use just before delivery.8
  • Neonatal withdrawal may be caused by very late, third trimester exposure to benzodiazepines. Symptoms—which can persist ≤3 months after delivery—include restlessness, irritability, abnormal sleep patterns, suckling difficulties, growth retardation, hypertonia, hyperreflexia, tremulousness, apnea, diarrhea, and vomiting.8,29
Recommendations. When possible, avoid benzodiazepines in the first trimester because of possible teratogenicity and then again late in the third trimester before delivery because of neonatal withdrawal syndromes. To reduce as much as possible the small risk of a benzodiazepine-related fetal malformation/cleft palate, wean the mother from benzodiazepines before conception. After the first trimester, the benzodiazepine can be restarted if necessary.29

To minimize neonatal withdrawal, gradually taper the mother’s benzodiazepine before delivery.29 Because the baby’s due date is calculated to be ±2 weeks before delivery, begin this taper 3 to 4 weeks before the due date and discontinue at least 1 week before delivery. Breastfeeding while taking benzodiazepines is not recommended because of the risk of over-sedating the infant.

 

 

A rational approach

Both benzodiazepines and SSRIs are associated with low but demonstrated risks to the fetus when used during pregnancy (Table 2).19,20,23,25,30,33 Use these medications to manage a patient’s anxiety only if the clinical benefit to the mother justifies the potential risks to the fetus.29

A staggered combination of SSRIs during the first 2 trimesters and benzodiazepines during the last 2 trimesters can help balance the risks and benefits of pharmacotherapy of anxiety disorders during pregnancy (Table 3).

Frankly discuss with your patient the risks and benefits in the context of her perceived need for symptom control to sustain her level of functioning. You could document this discussion in the progress note as “R, B, A, and pt C,” signifying that risks, benefits, and alternatives were discussed, and the patient consented. If possible, include the patient’s husband, partner, or parent in this discussion.

Table 2

Risks of SSRIs vs benzodiazepines during pregnancy stages

Pregnancy stage when givenFetal riskSSRIsBenzodiazepines
First trimester*TeratogenicityParoxetine use associated with 2-fold increased risk of major congenital anomalies and 3-fold increased risk of major cardiac anomalies;19 meta-analysis calculated significant risk of cardiac malformations (odds ratio 1.72; population prevalence = 13.4/1,000 births)20,33Meta-analysis of case control studies showed increased risk of major malformations/cleft palate (odds ratio 3.01; population prevalence = 10 to 20/1,000 births); no association seen in cohort studies30
Third trimesterPPHNCase control study showed 3.7% of infants with PPHN were exposed to SSRIs vs 0.7% of controls; adjusted odds ratio 6.1, absolute risk to exposed population = 6 to 12/1,000 births)23 
Perinatal and long-term effectsToxicity/withdrawal syndromesCohort study of 60 infants concluded prevalence of discontinuation syndromes is 30% in neonates with third trimester SSRI exposure25Neonatal toxicity (“floppy infant syndrome”) and neonatal withdrawal reported with maternal benzodiazepine use in late third trimester; prevalence unknown
 Preterm birth, serotonin withdrawal syndromes, CNS effects, long-term neurobehavioral effectsUnknownUnknown
PPHN: persistent pulmonary hypertension of the newborn; SSRIs: selective serotonin reuptake inhibitors
* Available data indicate that first-trimester exposure to SSRIs (other than paroxetine) and benzodiazepines may increase the relative risk for congenital anomalies, but the absolute risk of having a child with an anomaly is small.
Some case reports, but published literature is insufficient to determine prevalence or magnitude of risk.
Table 3

Staggered, combination therapy for anxiety disorders during pregnancy

Pregnancy stageRecommended to manage risks to mother and fetus
First trimester
  • SSRI (not paroxetine)
  • No benzodiazepines
  • Nondrug therapies*
Second trimester
  • SSRI (not paroxetine)
  • Can use benzodiazepine if needed
  • Nondrug therapies*
Third trimester
  • Taper off SSRI by 1 to 2 months before due date
  • Can use benzodiazepine until 2 weeks before due date
  • Nondrug therapies*
SSRI: selective serotonin reuptake inhibitor
* Nondrug therapies can include prenatal exercise, sleep hygiene, relaxation, and psychotherapy (cognitive-behavioral therapy, interpersonal therapy, supportive therapy, family/couples therapy)

CASE CONTINUED: CBT plus medication

Ms. K and her husband are open to adding weekly cognitive-behavioral therapy (CBT) for anxiety as long as she can continue her medications. You discuss the evidence regarding potential neonatal risks with paroxetine and clonazepam treatment. Because Ms. K is 6 weeks pregnant, you outline a plan for a rapid cross-taper off paroxetine and onto fluoxetine, 10 to 30 mg/d, explaining that paroxetine might pose a greater first-trimester risk of major congenital malformations and cardiac malformations. You discuss possible side effects of fluoxetine and explain a plan to taper off fluoxetine during the third trimester to reduce the risk of PPHN, early delivery, and withdrawal in the newborn.

Because Ms. K has been taking clonazepam at only 0.5 mg 1 to 2 times per week, you instruct her to stop taking the benzodiazepine for the next 6 weeks until she is through her first trimester. You also reassure her that she can use clonazepam after the first trimester, if necessary, as long as she agrees to taper off completely 1 to 2 weeks before to her due date.

You refer her to a CBT therapist and emphasize the importance of CBT, relaxation, and sleep hygiene—as well as support from her husband, family, and friends—to reduce her stress and facilitate the medication taper during her third trimester. You plan to see her monthly and co-manage her care with the CBT therapist and Ob/Gyn. You document this discussion in her medical record as evidence of informed consent.

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Paroxetine • Paxil
  • Fluoxetine • Prozac
  • Sertraline • Zoloft

Disclosures

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Ross LE, McLean LM. Anxiety disorders during pregnancy and the postpartum period: a systematic review. J Clin Psychiatry 2006;67(8):1285-98.

2. Labad J, Menchon JM, Alonso P, et al. Female reproductive cycle and obsessive-compulsive disorder. J Clin Psychiatry 2005;66(4):428-35.

3. Adewuya AO, Ola BA, Aloba OO, Mapayi BM. Anxiety disorders among Nigerian women in late pregnancy: a controlled study. Arch Womens Ment Health 2006;9(6):325-8.

4. Field T, Hernandez-Reif M, Diego M, et al. Stability of mood states and biochemistry across pregnancy. Infant Behav Dev 2006;29(2):262-7.

5. Teixeira JM, Fisk NM, Glover V. Association between maternal anxiety in pregnancy and increased uterine artery resistance index: cohort based study. BMJ 1999;318(7177):153-7.

6. Monk C, Myers MM, Sloan RP, et al. Effects of women’s stress-elicited physiological activity and chronic anxiety on fetal heart rate. J Dev Behav Pediatr 2003;24(1):32-8.

7. Egliston KA, McMahon C, Austin MP. Stress in pregnancy and infant HPA axis function: conceptual and methodological issues relating to the use of salivary cortisol as an outcome measure. Psychoneuroendocrinology 2007;32(1):1-13.

8. Levey L, Ragan K, Hower-Hartley A, et al. Psychiatric disorders in pregnancy. Neurol Clin 2004;22(4):863-93.

9. Oberlander TF, Reebye P, Misri S, et al. Externalizing and attentional behaviors in children of depressed mothers treated with a selective serotonin reuptake inhibitor antidepressant during pregnancy. Arch Pediatr Adolesc Med 2007;161(1):22-9.

10. Misri S, Reebye P, Kendrick K, et al. Internalizing behaviors in 4-year-old children exposed in utero to psychotropic medications. Am J Psychiatry 2006;163(6):1026-32.

11. Copper RL, Goldenberg RL, Das A, et al. The Preterm Prediction Study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks’ gestation. Am J Obstet Gynecol 1996;175(5):1286-92.

12. Sutter-Dallay AL, Giaconne-Marcesche V, Glatigny-Dallay E, Verdoux H. Women with anxiety disorders during pregnancy are at increased risk of intense postnatal depressive symptoms: a prospective survey of the MATQUID cohort. Eur Psychiatry 2004;19(8):459-63.

13. Nierop A, Bratsikas A, Zimmermann R, Ehlert U. Are stress-induced cortisol changes during pregnancy associated with postpartum depressive symptoms? Psychosom Med 2006;68(6):931-7.

14. Weissman MM. Recent non-medication trials of interpersonal psychotherapy for depression. Int J Neuropsychopharmacology 2007;10(1):117-22.

15. Ward RK, Zamorski MA. Benefits and risks of psychiatric medications during pregnancy. Am Fam Physician 2002;66(4):629-36.

16. Bastani F, Hidarnia A, Montgomery KS, et al. Does relaxation education in anxious primigravid Iranian women influence adverse pregnancy outcomes? A randomized controlled trial. J Perinat Neonatal Nurs 2006;20(2):138-46.

17. Fricchione G. Generalized anxiety disorder. N Engl J Med 2004;351(7):675-82.

18. Källén BA, Otterblad Olausson P. Maternal use of selective serotonin re-uptake inhibitors in early pregnancy and infant congenital malformations. Birth Defects Res A Clin Mol Teratol 2007;79(4):301-8.

19. Berard A, Ramos E, Rey E, et al. First trimester exposure to paroxetine and risk of cardiac malformations in infants: the importance of dosage. Birth Defects Res B Dev Reprod Toxicol 2007;80(1):18-27.

20. Bar-Oz B, Einarson T, Einarson A, et al. Paroxetine and congenital malformations: meta-analysis and consideration of potential confounding factors. Clin Ther 2005;29(5):918-26.

21. Malm H, Klaukka T, Neuvonen PJ. Risks associated with selective serotonin reuptake inhibitors in pregnancy. Obstet Gynecol 2005;106(6):1289-96.

22. Nulman I, Rovet J, Stewart DE, et al. Child development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: a prospective, controlled study. Am J Psychiatry 2002;159(11):1889-95.

23. Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med 2006;354(6):579-87.

24. Haddad PM, Pal BR, Clarke P, et al. Neonatal symptoms following maternal paroxetine treatment: serotonin toxicity or paroxetine discontinuation syndrome? J Psychopharmacology 2005;19(5):554-7.

25. Levinson-Castiel R, Merlob P, Linder N, et al. Neonatal abstinence syndrome after in utero exposure to selective serotonin reuptake inhibitors in term infants. Arch Pediatr Adolesc Med 2006;160(2):173-6.

26. Sanz EJ, De-las-Cuevas C, Kiuru A, et al. Selective serotonin reuptake inhibitors in pregnant women and neonatal withdrawal syndrome: a database analysis. Lancet 2005;365(9458):482-7.

27. Suri R, Altshuler L, Hellemann G, et al. Effects of antenatal depression and antidepressant treatment on gestational age at birth and risk of preterm birth. Am J Psychiatry 2007;164(8):1206-13.

28. Zeskind PS, Stephens LE. Maternal selective serotonin reuptake inhibitor use during pregnancy and newborn neurobehavior. Pediatrics 2004;113(2):368-75.

29. Iqbal MM, Sobhan T, Ryals T. Effects of commonly used benzodiazepines on the fetus, the neonate, and the nursing infant. Psychiatr Serv 2002;53(1):39-49.

30. Dolovich LR, Addis A, Vaillancourt JM, et al. Benzodiazepine use in pregnancy and major malformations or oral cleft: meta-analysis of cohort and case-control studies. BMJ 1998;317(7162):839-43.

31. McElhatton PR. The effects of benzodiazepine use during pregnancy. Reprod Toxicol 1994;8(6):461-75.

32. Lin AE, Peller AJ, Westgate MN, et al. Clonazepam use in pregnancy and the risk of malformations. Birth Defects Res A Clin Mol Teratol 2004;70(8):534-6.

33. Levy M, James MS, Erickson JD, McClearn AB. Prevalence of birth defects. Birth outcomes Centers for Disease Control and Prevention. Available at: http://www.cdc.gov/reproductivehealth/Products&Pubs/DatatoAction/pdf/birout4.pdf. Accessed January 9, 2008.

References

1. Ross LE, McLean LM. Anxiety disorders during pregnancy and the postpartum period: a systematic review. J Clin Psychiatry 2006;67(8):1285-98.

2. Labad J, Menchon JM, Alonso P, et al. Female reproductive cycle and obsessive-compulsive disorder. J Clin Psychiatry 2005;66(4):428-35.

3. Adewuya AO, Ola BA, Aloba OO, Mapayi BM. Anxiety disorders among Nigerian women in late pregnancy: a controlled study. Arch Womens Ment Health 2006;9(6):325-8.

4. Field T, Hernandez-Reif M, Diego M, et al. Stability of mood states and biochemistry across pregnancy. Infant Behav Dev 2006;29(2):262-7.

5. Teixeira JM, Fisk NM, Glover V. Association between maternal anxiety in pregnancy and increased uterine artery resistance index: cohort based study. BMJ 1999;318(7177):153-7.

6. Monk C, Myers MM, Sloan RP, et al. Effects of women’s stress-elicited physiological activity and chronic anxiety on fetal heart rate. J Dev Behav Pediatr 2003;24(1):32-8.

7. Egliston KA, McMahon C, Austin MP. Stress in pregnancy and infant HPA axis function: conceptual and methodological issues relating to the use of salivary cortisol as an outcome measure. Psychoneuroendocrinology 2007;32(1):1-13.

8. Levey L, Ragan K, Hower-Hartley A, et al. Psychiatric disorders in pregnancy. Neurol Clin 2004;22(4):863-93.

9. Oberlander TF, Reebye P, Misri S, et al. Externalizing and attentional behaviors in children of depressed mothers treated with a selective serotonin reuptake inhibitor antidepressant during pregnancy. Arch Pediatr Adolesc Med 2007;161(1):22-9.

10. Misri S, Reebye P, Kendrick K, et al. Internalizing behaviors in 4-year-old children exposed in utero to psychotropic medications. Am J Psychiatry 2006;163(6):1026-32.

11. Copper RL, Goldenberg RL, Das A, et al. The Preterm Prediction Study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks’ gestation. Am J Obstet Gynecol 1996;175(5):1286-92.

12. Sutter-Dallay AL, Giaconne-Marcesche V, Glatigny-Dallay E, Verdoux H. Women with anxiety disorders during pregnancy are at increased risk of intense postnatal depressive symptoms: a prospective survey of the MATQUID cohort. Eur Psychiatry 2004;19(8):459-63.

13. Nierop A, Bratsikas A, Zimmermann R, Ehlert U. Are stress-induced cortisol changes during pregnancy associated with postpartum depressive symptoms? Psychosom Med 2006;68(6):931-7.

14. Weissman MM. Recent non-medication trials of interpersonal psychotherapy for depression. Int J Neuropsychopharmacology 2007;10(1):117-22.

15. Ward RK, Zamorski MA. Benefits and risks of psychiatric medications during pregnancy. Am Fam Physician 2002;66(4):629-36.

16. Bastani F, Hidarnia A, Montgomery KS, et al. Does relaxation education in anxious primigravid Iranian women influence adverse pregnancy outcomes? A randomized controlled trial. J Perinat Neonatal Nurs 2006;20(2):138-46.

17. Fricchione G. Generalized anxiety disorder. N Engl J Med 2004;351(7):675-82.

18. Källén BA, Otterblad Olausson P. Maternal use of selective serotonin re-uptake inhibitors in early pregnancy and infant congenital malformations. Birth Defects Res A Clin Mol Teratol 2007;79(4):301-8.

19. Berard A, Ramos E, Rey E, et al. First trimester exposure to paroxetine and risk of cardiac malformations in infants: the importance of dosage. Birth Defects Res B Dev Reprod Toxicol 2007;80(1):18-27.

20. Bar-Oz B, Einarson T, Einarson A, et al. Paroxetine and congenital malformations: meta-analysis and consideration of potential confounding factors. Clin Ther 2005;29(5):918-26.

21. Malm H, Klaukka T, Neuvonen PJ. Risks associated with selective serotonin reuptake inhibitors in pregnancy. Obstet Gynecol 2005;106(6):1289-96.

22. Nulman I, Rovet J, Stewart DE, et al. Child development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: a prospective, controlled study. Am J Psychiatry 2002;159(11):1889-95.

23. Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med 2006;354(6):579-87.

24. Haddad PM, Pal BR, Clarke P, et al. Neonatal symptoms following maternal paroxetine treatment: serotonin toxicity or paroxetine discontinuation syndrome? J Psychopharmacology 2005;19(5):554-7.

25. Levinson-Castiel R, Merlob P, Linder N, et al. Neonatal abstinence syndrome after in utero exposure to selective serotonin reuptake inhibitors in term infants. Arch Pediatr Adolesc Med 2006;160(2):173-6.

26. Sanz EJ, De-las-Cuevas C, Kiuru A, et al. Selective serotonin reuptake inhibitors in pregnant women and neonatal withdrawal syndrome: a database analysis. Lancet 2005;365(9458):482-7.

27. Suri R, Altshuler L, Hellemann G, et al. Effects of antenatal depression and antidepressant treatment on gestational age at birth and risk of preterm birth. Am J Psychiatry 2007;164(8):1206-13.

28. Zeskind PS, Stephens LE. Maternal selective serotonin reuptake inhibitor use during pregnancy and newborn neurobehavior. Pediatrics 2004;113(2):368-75.

29. Iqbal MM, Sobhan T, Ryals T. Effects of commonly used benzodiazepines on the fetus, the neonate, and the nursing infant. Psychiatr Serv 2002;53(1):39-49.

30. Dolovich LR, Addis A, Vaillancourt JM, et al. Benzodiazepine use in pregnancy and major malformations or oral cleft: meta-analysis of cohort and case-control studies. BMJ 1998;317(7162):839-43.

31. McElhatton PR. The effects of benzodiazepine use during pregnancy. Reprod Toxicol 1994;8(6):461-75.

32. Lin AE, Peller AJ, Westgate MN, et al. Clonazepam use in pregnancy and the risk of malformations. Birth Defects Res A Clin Mol Teratol 2004;70(8):534-6.

33. Levy M, James MS, Erickson JD, McClearn AB. Prevalence of birth defects. Birth outcomes Centers for Disease Control and Prevention. Available at: http://www.cdc.gov/reproductivehealth/Products&Pubs/DatatoAction/pdf/birout4.pdf. Accessed January 9, 2008.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
39-52
Page Number
39-52
Publications
Publications
Article Type
Display Headline
Treating anxiety during pregnancy: Just how safe are SSRIs?
Display Headline
Treating anxiety during pregnancy: Just how safe are SSRIs?
Legacy Keywords
anxiety during pregnancy; SSRIs in pregnancy; Deborah B. Raphael MD; Jessica Ross MD; Louann Brizendine MD; clonazepam; paroxetine; generalized anxiety disorder; posttraumatic stress disorder; PTSD; obsessive-compulsive disorder; OCD; maternal anxiety; teratogenicity; fetal malformations from SSRIs; maternal SSRI use
Legacy Keywords
anxiety during pregnancy; SSRIs in pregnancy; Deborah B. Raphael MD; Jessica Ross MD; Louann Brizendine MD; clonazepam; paroxetine; generalized anxiety disorder; posttraumatic stress disorder; PTSD; obsessive-compulsive disorder; OCD; maternal anxiety; teratogenicity; fetal malformations from SSRIs; maternal SSRI use
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Treating alcohol dependence: When and how to use 4 medications

Article Type
Changed
Tue, 12/11/2018 - 15:07
Display Headline
Treating alcohol dependence: When and how to use 4 medications

Mr. G, age 38, is an investment banker referred for evaluation of an alcohol use disorder. Three years ago his internist diagnosed Mr. G with major depression and prescribed a selective serotonin reuptake inhibitor. Mr. G’s mood has improved, but his drinking is out of control and is affecting his work and marriage.

Mr. G describes his father as an alcoholic and says he has noticed worrisome similarities in himself. Since his teenage years, he recalls always being able to drink more than his peers. Amnesia episodes began in college during heavy drinking days and now occur almost weekly. Most recently his driver’s license was suspended after he was arrested for driving while impaired by alcohol.

He has attempted to stop drinking 3 times in the last 6 months and feels frustrated because he continues to relapse. During his last quit attempt, he remained abstinent for 3 months and believes his mood was unchanged during that time.

For motivated patients such as Mr. G, National Institute on Alcohol Abuse and Alcoholism (NIAAA) guidelines (updated in 2007) consider medications first-line treatment for alcohol dependence, along with psychotherapies and mutual-help groups such as Alcoholics Anonymous.1 Medications with evidence of efficacy include FDA-approved disulfiram, naltrexone, and acamprosate, and off-label topiramate.

Each drug’s pharmacology is different; some may be beneficial during early abstinence, whereas others are more effective for maintaining abstinence. Because many physicians have had little or no experience using these medications,2,3 we discuss dosing recommendations and side effect profiles—important clinical differences to guide drug selection and administration.

Box

Alcohol dependence: Diagnostic criteria

Alcohol use disorders describe a maladaptive pattern of alcohol use that causes clinically significant impairment or distress. Alcohol dependence is manifested by ≥3 of the following symptoms in a 12-month period:

  • Tolerance
  • Withdrawal
  • Often drinking alcohol in larger amounts or over a longer time period than intended
  • Persistent desire or unsuccessful efforts to cut down or control alcohol use
  • Spending a great deal of time in activities necessary to obtain, use, or recover from alcohol’s effects
  • Giving up or reducing important social, occupational, or recreational activities because of alcohol use
  • Continuing alcohol use despite knowing you have a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by alcohol.

Source: Adapted from DSM-IV-TR

CASE CONTINUED: Which came first?

When Mr. G presented with depressive symptoms, his internist informed him that alcohol’s effects can mimic depression and advised him to cut back his consumption. Mr. G temporarily reduced his drinking but continued to experience depressed mood, sleep disturbances, difficulty concentrating, fatigue, and poor appetite. The internist then prescribed escitalopram, 10 mg/d, and Mr. G says his depressive symptoms improved. He has not attempted suicide or required psychiatric hospitalization.

After a thorough evaluation—including a detailed assessment of his drinking history, other substance use, and mood symptoms—you diagnose Mr. G with alcohol dependence without physiological dependence and a primary major depressive disorder (MDD).

Mr. G’s diagnosis of alcohol dependence is based on evidence in the last 12 months of tolerance, repeated loss of control over the amount he drinks, multiple failed attempts to stop drinking, repeated negative consequences to his work productivity and personal relationships, and continued drinking despite knowing that alcohol consumption sometimes intensifies his mood symptoms (Box). Physiological dependence is unlikely because he did not experience withdrawal during a recent period of abstinence.

The mood component. Particularly in psychiatric patients, alcohol dependence often coexists with and affects the treatment of other psychopathologies:

  • 1 in 3 adults experience an alcohol use disorder during their lifetimes.4
  • 1 in 3 adults with an alcohol use disorder have a comorbid psychiatric disorder.5
  • Alcohol dependence doubles the risk of major depression and triples the lifetime risk of any mood disorder.4
A thorough evaluation—both medical and psychiatric—is necessary to distinguish a primary mood disorder from a substance-induced mood disorder. Aspects of a patient’s history that may support a substance-induced disorder diagnosis include:

  • Mood symptoms appear after the onset of a substance use disorder.
  • Mood symptoms are absent during abstinence.
  • Mood symptoms are consistent with the effects of the drug being used.6
Substance-induced mood symptoms usually become less intense and eventually resolve as abstinence is maintained. Primary mood symptoms may not be affected by abstinence, and interventions—including pharmacotherapy—are more likely to be required.

In evaluating Mr. G’s drinking history, you determined that his mood symptoms began before he started consuming alcohol regularly and have persisted during periods of abstinence. Thus, primary MDD is a more likely diagnosis than substance-induced MDD.

 

 

CASE CONTINUED: Planning treatment

A combination of behavioral therapy and pharmacotherapy is appropriate for treating Mr. G’s alcohol dependence. When you discuss the diagnosis with him, he endorses a goal of abstinence. For behavioral therapy, he says he would like to try Alcoholics Anonymous, which helped a friend “turn his life around.”

He has become accustomed to taking escitalopram once daily but is hesitant to take any medication that requires more frequent dosing. He also worries that medication might impair his work performance, which requires extensive periods of concentration. Your goal—as you consider available medications—is to develop a treatment plan that incorporates Mr. G’s preferences and addresses his concerns.

Disulfiram

Disulfiram, an irreversible aldehyde dehydrogenase inhibitor, is indicated for maintaining enforced sobriety in patients with chronic alcohol dependence (Table 1). Aldehyde dehydrogenase inhibition disrupts alcohol-to-acetate metabolism, which leads to acetaldehyde accumulation. If a disulfiram-treated patient ingests alcohol, increased acetaldehyde levels lead to the unpleasant “disulfiram-ethanol reaction,” with diaphoresis, flushing, nausea, vomiting, headache, tachycardia, and hypotension. The reaction’s severity is proportional to the disulfiram dose and amount of alcohol consumed.

Patients taking disulfiram must abstain from all alcohol, including over-the-counter cold remedies and mouthwashes containing alcohol. Advise patients that ingesting small amounts of alcohol can induce symptoms, even days after taking disulfiram.

Efficacy. Clinical trial results with disulfiram have been mixed. In the largest controlled study to date, Fuller et al7 found no significant difference in rates of total abstinence, time to first drink, employment, or social stability measures at 1 year among 605 men who received counseling plus disulfiram, 250 mg/d, or placebo. Other disulfiram studies have found a modest decrease in the frequency of drinking but no effect on abstinence rates.8,9

Because medication adherence is the strongest predictor of outcome with disulfiram,10 monitoring for adherence and stressing its importance to patients may increase the drug’s efficacy. Disulfiram may be most effective in highly motivated patients with stable social support or as an adjuvant to an outpatient treatment program.

Administration. Disulfiram is available in 250-mg tablets and is usually dosed from 125 to 500 mg/d. Treatment can begin after patients abstain from alcohol for ≥12 hours and have a serum alcohol concentration of zero.

Side effects. Drowsiness is a common complaint with disulfiram; this adverse effect is frequently self-limited and can be reduced by evening dosing.

Subclinical liver enzyme elevations have been reported in 25% of patients taking disulfiram.11 Although rare, potentially fatal hepatotoxicity has been reported12,13 (with a dose as low as 200 mg/d12), typically occurring early in treatment and associated with jaundice and fever. One study estimated the risk of dying of hepatotoxicity caused by disulfiram to be 1 in 30,000 patients/year.14

A recent Swedish study13 reviewed data from 1966 through 2002 and found 82 cases of drug-induced liver injury associated with disulfiram. By comparing these findings with sales figures from 1972 to 2002, the authors report an incidence of disulfiram-induced liver injury of about 1 case per 1.3 million estimated average daily doses.

Order liver function tests at baseline, then retest 10 to 14 days after starting disulfiram and again approximately 4 weeks later. Thereafter, monitoring once every 3 to 6 months is generally sufficient in patients without liver disease symptoms.

Other serious adverse events associated with disulfiram therapy include optic neuritis, peripheral neuritis, cholestatic hepatitis, seizures, and arrhythmias. Psychosis also can occur, generally with dosages ≥500 mg/d. Avoid concomitant use of disulfiram and metronidazole, which can cause acute psychosis.

Disulfiram-related inhibition of cytochrome P-450 can increase serum levels and toxicity risk of medications metabolized in the liver, such as warfarin, phenytoin, and isoniazid. Patients taking concomitant warfarin and disulfiram require close monitoring for increases in the international normalized ratio (INR).

Contraindications. Disulfiram is contraindicated in patients with ischemic heart disease and those who are pregnant. Also avoid disulfiram in patients with cerebrovascular disease, diabetes mellitus, psychosis, or cognitive impairment.

Recommendation. Disulfiram is a valid option for treating alcohol dependence in a select group of highly motivated patients who are medically and psychiatrically stable and in whom adherence can be closely monitored.

Table 1

Disulfiram: Fast facts

Mechanism: Acetaldehyde accumulates when aldehyde dehydrogenase is inhibited
FDA-approved for alcohol dependence: Yes
Dosing: 125 to 500 mg once daily
Effect: Aversive reaction to alcohol
Potential side effects: Liver toxicity, seizures, arrhythmia, peripheral neuropathy, psychosis
Contraindications: Concurrent alcohol consumption, severe cardiac disease, psychosis, pregnancy
Comments: Many drug interactions, including warfarin, metronidazole, and phenytoin; monitor liver function for toxicity
 

 

Naltrexone

Naltrexone is a μ-opioid receptor antagonist thought to reduce alcohol’s reinforcing effects by interfering with β-endorphin pathways. It is indicated for treating alcohol dependence and has been shown to reduce relapse and number of drinking days in alcohol-dependent patients (Table 2).15

Naltrexone also has reduced alcohol consumption in healthy volunteers, social drinkers, and other nondependent drinkers.16 Its effect may have a genetic component, as suggested by greater benefit in persons with a family history of alcoholism.17

Efficacy. Most studies investigated naltrexone as part of a comprehensive treatment program that included behavioral therapies.18 Recently, the randomized, placebo-controlled Combining Medications and Behavioral Interventions (COMBINE) study found that using either naltrexone or behavioral therapy improved abstinence, and combing naltrexone with behavioral therapy was not more effective than either treatment alone.19

Administration. Oral naltrexone is usually started at 25 mg and increased over 2 to 3 days to 50 or 100 mg/d. The standard dose is 50 mg/d, although the COMBINE study reported efficacy at 100 mg/d.19

Oral naltrexone is most helpful for patients who adhere to 70% to 90% of the medication.20 The extended-release form (a 380-mg IM dose given every 4 weeks) provides an option to monitor adherence.21

Side effects. Naltrexone toxicity can cause hepatocellular injury. Do not administer this drug to patients with acute hepatitis or end-stage liver disease. When prescribing naltrexone, check patients’ liver function monthly for the first 3 months, then once every 3 months thereafter.22 Less serious, common side effects include nausea, myalgia, and headache.

Naltrexone antagonizes opioid receptors and causes withdrawal symptoms in patients who are physically dependent on opioids. Therefore, do not give naltrexone to patients who require opioids for chronic pain. If your patient is using an opioid but could switch to other pain medication, discontinue the opioid for at least 7 days and consider a urine toxicology or naltrexone challenge before starting naltrexone.

Urine drug tests are inexpensive and easy to use but have limitations. Many standard “dipsticks” will detect heroin, morphine, and codeine but not oxycodone, hydrocodone, or other synthetic opioids. Specific tests are available to detect oxycodone, hydrocodone, hydromorphone, buprenorphine, and methadone. Some synthetic opioids (such as fentanyl) remain difficult to detect, however, because of their low concentration and rapid metabolism.

Table 2

Naltrexone: Fast facts

Mechanism: Opioid receptor antagonism interferes with β-endorphin pathways
FDA-approved for alcohol dependence: Yes
Dosing: Oral 50 mg once daily (recent evidence suggests safety and efficacy at 100 mg once daily); IM 380 mg once every 4 weeks
Effect: Decreases frequency and severity of relapse
Potential side effects: Nausea, myalgia, headache, dizziness
Contraindications: Opioid use, acute hepatitis, liver failure
Comments: Monitor liver function; once-monthly dosing may improve adherence

Acamprosate

Acamprosate is structurally similar to gamma-aminobutyric acid (GABA) and is thought to inhibit the glutamatergic system. This attenuation by acamprosate reduces the glutamatergic hyperactivity normally seen after chronic alcohol exposure.

Acamprosate is indicated for relapse prevention in patients with alcohol dependence who have stopped drinking (Table 3). Multiple randomized studies have demonstrated its efficacy in improving abstinence rates as compared with placebo.23,24 Other studies, however, failed to show improved abstinence with acamprosate.25 Some of the negative studies included patients who recently relapsed or had only a few days of abstinence before starting acamprosate.19,26 Therefore, acamprosate might be most effective when used to maintain abstinence and less effective—if at all—to initiate abstinence.

Administration. Acamprosate is available in 333-mg tablets, with a recommended dosage of 666 mg tid. Side effects tend to be transient and mild. Reduce the dose to 333 mg tid for patients with moderate renal insufficiency (creatinine clearance [CrCl] 30 to 50 mL/min), and do not use acamprosate in patients with severe renal insufficiency (CrCl

Side effects. Acamprosate was well-tolerated in clinical trials; diarrhea and other GI side effects were the most commonly reported adverse events.

In some placebo-controlled studies, patients taking acamprosate reported more frequent suicidal thoughts and attempts compared with patients taking placebo.27 These events were extremely rare, and no direct relationship with acamprosate therapy has been established. Nonetheless, monitor patients for depression and suicidal thoughts during acamprosate therapy.

Table 3

Acamprosate: Fast facts

Mechanism: Structurally similar to GABA; thought to inhibit the glutamatergic system
FDA-approved for alcohol dependence: Yes
Dosing: 666 mg tid
Effect: Increases abstinence
Potential side effects: Nausea, diarrhea, suicidal thoughts
Contraindications: Severe renal disease
Comments: Monitor patients for suicidal thoughts and depression

Topiramate

Topiramate potentiates GABA and inhibits excitatory glutamate transmission—properties believed to lead to decreased dopamine release at the nucleus accumbens in response to alcohol consumption. Although topiramate is not FDA-approved for alcohol dependence, limited data comparing this anticonvulsant with placebo have shown a reduction in drinking and increased abstinence (Table 4).28,29

 

 

Administration. Start with 25 mg/d and increase over several weeks to 300 mg/d, given in divided doses.

Side effects include dizziness, paresthesia, somnolence, difficulty concentrating, and weight loss. Because topiramate is excreted renally, reduce doses by 50% in patients with CrCl

Other renal side effects include an elevated risk of nephrolithiasis. Topiramate’s inhibition of carbonic anhydrase can reduce bicarbonate levels, leading to a nonanion gap metabolic acidosis.

Table 4

Topiramate: Fast facts

Mechanism: Potentiates GABA and inhibits glutamate receptor subtypes
FDA-approved for alcohol dependence: No
Dosing: 300 mg/d in divided doses
Effect: Decreases craving and drinking
Potential side effects: Metabolic acidosis, psychomotor slowing, dizziness, difficulty concentrating, paresthesia, weight loss, nephrolithiasis, hyperammonemia with concomitant use of valproic acid
Contraindications: None known other than hypersensitivity (as with all drugs)
Comments: Dose titration requires several weeks; avoid abrupt withdrawal; may reduce effectiveness of oral contraceptives

CASE CONTINUED: Implementing a treatment plan

You start Mr. G on oral naltrexone, 25 mg/d, and titrate to 100 mg/d. Although no optimum treatment duration has been established, you plan to follow NIAAA recommendations that Mr. G use naltrexone at least 3 months, with the possibility of continuing 1 year or longer if he responds well.1

You schedule weekly visits for the first month to monitor for side effects and to make any necessary modifications in behavioral and pharmacologic treatment. You also continue escitalopram, 10 mg, which has successfully controlled Mr. G’s MDD symptoms.

Medications for alcohol dependence generally have been studied as adjuncts to behavioral therapies. The COMBINE study of 1,383 alcohol-abstinent patients found naltrexone with medical management or cognitive-behavioral therapy alone to be equivalent in efficacy.19 The medical management provided a supportive environment, encouraged medication compliance, provided empathy to build a therapeutic relationship, and promoted self-help groups as an adjunct to treatment.30 Thus, medication has a role in treating alcohol dependence, but behavioral therapy remains an important part of comprehensive substance abuse treatment.

When choosing medications, consider the agents’ clinically relevant differences:

  • Naltrexone and—less conclusively—topiramate have shown benefit for alcoholdependent patients starting treatment and for relapse prevention.
  • Acamprosate may help prevent relapse in abstinent patients.
  • Disulfiram remains a valid option in highly motivated patients with social support available to ensure medication adherence.
Because Mr. G is starting therapy after recent alcohol use, medications such as naltrexone and topiramate that have shown benefit early in treatment (in addition to relapse prevention) are preferred. Of these 2 drugs, naltrexone is the better choice for Mr. G—who is concerned about his work performance—because difficulty concentrating is a common side effect of topiramate. Oral naltrexone would be preferred as initial therapy for Mr. G because he has expressed comfort taking once-daily oral medication. The more expensive oncemonthly naltrexone depot formulation could be a second-line treatment if adherence becomes an issue.

Hypersensitivity is considered a contraindication for any medication. Mr. G tolerates well an initial dose of 25 mg/d, followed by increases to 50 mg and then 100 mg over several days. You titrate oral naltrexone to 100 mg/d—even though it is commonly prescribed at 50 mg/d—because recent evidence suggests efficacy and safety at the higher dosage.19

Related resources

Drug brand names

  • Acamprosate • Campral
  • Disulfiram • Antabuse
  • Escitalopram • Lexapro
  • Naltrexone, oral • ReVia
  • Naltrexone, extended-release • Vivitrol
  • Topiramate • Topamax
  • Valproic acid • Depakote
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. National Institutes of Health. Helping patients who drink too much: a clinician’s guide. Bethesda, MD: National Institute on Alcohol Abuse and Alcoholism; 2007. NIH Publication 07-3769. Available at: http://www.niaaa.nih.gov/Publications/EducationTrainingMaterials/guide.htm. Accessed January 3, 2008.

2. Substance Abuse and Mental Health Services Administration. National Survey of Substance Abuse Treatment Services: 2005. Data on substance abuse treatment facilities, (DASIS Series: S-34). Rockville, MD: Office of Applied Studies, 2006. DHHS Publication (SMA) 06-4206.

3. Substance Abuse and Mental Health Services Administration. Treatment episode data set (TEDS) highlights 2005: National admissions to substance abuse treatment services. Rockville, MD: Office of Applied Studies. 2006. DHHS Publication (SMA) 07-4229.

4. Hasin DS, Stinson FS, Ogburn EO, Grant BF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Arch Gen Psychiatry 2007;64(7):830-42.

5. Regier DA, Farmer ME, Rae DS, et al. Comorbidity of mental disorders with alcohol and other drug abuse. Results from the Epidemiologic Catchment Area (ECA) study. JAMA. 1990;264(19):2511-8.

6. Mariani JJ, Levin FR. Treatment of comorbid conditions of substance abuse. Directions in Psychiatry 2005;25(2):129-39.

7. Fuller RK, Branchey L, Brightwell DR, et al. Disulfiram treatment of alcoholism. A Veteran’s Administration cooperative study. JAMA. 1986;256:1449-55.

8. Fuller RK, Roth HP. Disulfiram for the treatment of alcoholism. An evaluation of 128 men. Ann Intern Med. 1979;90:901-4.

9. Schuckit MA. A one-year follow-up of men alcoholics given disulfiram. J Stud Alcohol 1985;46:191-5.

10. Collins GB, McAllister MS, Adury K. Drug adjuncts for treating alcohol dependence. Cleve Clin J Med 2006;73(7):641-4.

11. Goyer PF, Major LF. Hepatotoxicty in disulfiram treated patients. J Stud Alcohol 1979;40:133-7.

12. Ranek L, Buch Andreasen P. Disulfiram hepatotoxicity. Br Med J 1977;2(6079):94-6.

13. Björnsson E, Nordlinder H, Olsson R. Clinical characteristics and prognostic markers in disulfiram-induced liver injury. J Hepatol 2006;44(4):791-7.

14. Enghusen PH, Loft S, Anderson JR, et al. Disulfiram therapy—adverse drug reactions and interactions. Acta Psychiatr Scand 1992;86(369):59-66.

15. Volpicelli JR, Volpicelli LA, O’Brien CP. Medical management of alcohol dependence: clinical use and limitations of naltrexone treatment. Alcohol Alcohol 1995;30:789.-

16. de Wit H, Svenson J, York A. Non-specific effect of naltrexone on ethanol consumption in social drinkers. Psychopharmacology (Berl) 1999;146:33.-

17. Monterosso JR, Flannery BA, Pettinati HM, et al. Predicting treatment response to naltrexone: the influence of craving and family history. Am J Addict 2001;10:258-68.

18. Srisurapanont M, Jarusuraisin N. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev 2005;(1):CD001867.-

19. Anton RF, O’Malley SS, Ciraulo DA, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295:2003-17.

20. Monti PM, Rohsenow DJ, Swift RM, et al. Naltrexone and cue exposure with coping and communication skills training for alcoholics: treatment process and 1-year outcomes. Alcol Clin Exp Res 2001;25:1634-47.

21. Garbutt JC, Kranzler HR, O’Malley SS, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293(13):1617-25.

22. Doering PL. Substance-related disorders: alcohol, nicotine, and caffeine. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. 4th ed. Stamford, CT: Appleton & Lange; 1999.

23. Mann K, Lehert P, Morgan MY. The efficacy of acamprosate in the maintenance of abstinence in alcohol-dependent individuals: results of a meta-analysis. Alcohol Clin Exp Res 2004;28(1):51-63.

24. Paille FM, Guelfi JD, Perkins AC, et al. Double-blind randomized multicentre trial of acamprosate in maintaining abstinence from alcohol. Alcohol Alcohol 1995;30:239-47.

25. Chick J, Howlett H, Morgan MY, Ritson B. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol Alcohol 2000;35(2):176-87.

26. Mason BJ, Goodman AM, Chabac S, Lehert P. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind, placebo-controlled trial: the role of patient motivation. J Psychiatr Res 2006;40(5):383-93.

27. Mason BJ. Treatment of alcohol-dependent outpatients with acamprosate: a clinical review. J Clin Psychiatry 2001;62(suppl 20):42-8.

28. Johnson BA, Ait-Daoud N, Bowden CL, et al. Oral topiramate for treatment of alcohol dependence: a randomized controlled trial. Lancet 2003;361(9370):1677-85.

29. Johnson BA, Rosenthal N, Capece JA, et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA 2007;298(14):1641-51.

30. COMBINE Study Research Group. Testing combined pharmacotherapies and behavioral interventions in alcohol dependence: rationale and methods. Alcohol Clin Exp Res 2003;27:1107-22.

Article PDF
Author and Disclosure Information

Soteri Polydorou, MD
Postdoctoral fellow division on substance abuse, department of psychiatry
Assistant in clinical medicine, department of medicine
College of Physicians and Surgeons,
Columbia University,
New York, NY

Frances R. Levin, MD
Fellowship director, division on substance abuse,
Kennedy-Leavy Professor of Clinical Psychiatry, department of psychiatry
College of Physicians and Surgeons,
Columbia University,
New York, NY

Issue
Current Psychiatry - 07(02)
Publications
Page Number
25-37
Legacy Keywords
alcohol dependence; Soteri Polydorou MD; Frances R. Levin MD; disulfiram; naltrexone; acamprosate; topiramate; alcohol use disorders; substance-induced mood symptoms
Sections
Author and Disclosure Information

Soteri Polydorou, MD
Postdoctoral fellow division on substance abuse, department of psychiatry
Assistant in clinical medicine, department of medicine
College of Physicians and Surgeons,
Columbia University,
New York, NY

Frances R. Levin, MD
Fellowship director, division on substance abuse,
Kennedy-Leavy Professor of Clinical Psychiatry, department of psychiatry
College of Physicians and Surgeons,
Columbia University,
New York, NY

Author and Disclosure Information

Soteri Polydorou, MD
Postdoctoral fellow division on substance abuse, department of psychiatry
Assistant in clinical medicine, department of medicine
College of Physicians and Surgeons,
Columbia University,
New York, NY

Frances R. Levin, MD
Fellowship director, division on substance abuse,
Kennedy-Leavy Professor of Clinical Psychiatry, department of psychiatry
College of Physicians and Surgeons,
Columbia University,
New York, NY

Article PDF
Article PDF

Mr. G, age 38, is an investment banker referred for evaluation of an alcohol use disorder. Three years ago his internist diagnosed Mr. G with major depression and prescribed a selective serotonin reuptake inhibitor. Mr. G’s mood has improved, but his drinking is out of control and is affecting his work and marriage.

Mr. G describes his father as an alcoholic and says he has noticed worrisome similarities in himself. Since his teenage years, he recalls always being able to drink more than his peers. Amnesia episodes began in college during heavy drinking days and now occur almost weekly. Most recently his driver’s license was suspended after he was arrested for driving while impaired by alcohol.

He has attempted to stop drinking 3 times in the last 6 months and feels frustrated because he continues to relapse. During his last quit attempt, he remained abstinent for 3 months and believes his mood was unchanged during that time.

For motivated patients such as Mr. G, National Institute on Alcohol Abuse and Alcoholism (NIAAA) guidelines (updated in 2007) consider medications first-line treatment for alcohol dependence, along with psychotherapies and mutual-help groups such as Alcoholics Anonymous.1 Medications with evidence of efficacy include FDA-approved disulfiram, naltrexone, and acamprosate, and off-label topiramate.

Each drug’s pharmacology is different; some may be beneficial during early abstinence, whereas others are more effective for maintaining abstinence. Because many physicians have had little or no experience using these medications,2,3 we discuss dosing recommendations and side effect profiles—important clinical differences to guide drug selection and administration.

Box

Alcohol dependence: Diagnostic criteria

Alcohol use disorders describe a maladaptive pattern of alcohol use that causes clinically significant impairment or distress. Alcohol dependence is manifested by ≥3 of the following symptoms in a 12-month period:

  • Tolerance
  • Withdrawal
  • Often drinking alcohol in larger amounts or over a longer time period than intended
  • Persistent desire or unsuccessful efforts to cut down or control alcohol use
  • Spending a great deal of time in activities necessary to obtain, use, or recover from alcohol’s effects
  • Giving up or reducing important social, occupational, or recreational activities because of alcohol use
  • Continuing alcohol use despite knowing you have a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by alcohol.

Source: Adapted from DSM-IV-TR

CASE CONTINUED: Which came first?

When Mr. G presented with depressive symptoms, his internist informed him that alcohol’s effects can mimic depression and advised him to cut back his consumption. Mr. G temporarily reduced his drinking but continued to experience depressed mood, sleep disturbances, difficulty concentrating, fatigue, and poor appetite. The internist then prescribed escitalopram, 10 mg/d, and Mr. G says his depressive symptoms improved. He has not attempted suicide or required psychiatric hospitalization.

After a thorough evaluation—including a detailed assessment of his drinking history, other substance use, and mood symptoms—you diagnose Mr. G with alcohol dependence without physiological dependence and a primary major depressive disorder (MDD).

Mr. G’s diagnosis of alcohol dependence is based on evidence in the last 12 months of tolerance, repeated loss of control over the amount he drinks, multiple failed attempts to stop drinking, repeated negative consequences to his work productivity and personal relationships, and continued drinking despite knowing that alcohol consumption sometimes intensifies his mood symptoms (Box). Physiological dependence is unlikely because he did not experience withdrawal during a recent period of abstinence.

The mood component. Particularly in psychiatric patients, alcohol dependence often coexists with and affects the treatment of other psychopathologies:

  • 1 in 3 adults experience an alcohol use disorder during their lifetimes.4
  • 1 in 3 adults with an alcohol use disorder have a comorbid psychiatric disorder.5
  • Alcohol dependence doubles the risk of major depression and triples the lifetime risk of any mood disorder.4
A thorough evaluation—both medical and psychiatric—is necessary to distinguish a primary mood disorder from a substance-induced mood disorder. Aspects of a patient’s history that may support a substance-induced disorder diagnosis include:

  • Mood symptoms appear after the onset of a substance use disorder.
  • Mood symptoms are absent during abstinence.
  • Mood symptoms are consistent with the effects of the drug being used.6
Substance-induced mood symptoms usually become less intense and eventually resolve as abstinence is maintained. Primary mood symptoms may not be affected by abstinence, and interventions—including pharmacotherapy—are more likely to be required.

In evaluating Mr. G’s drinking history, you determined that his mood symptoms began before he started consuming alcohol regularly and have persisted during periods of abstinence. Thus, primary MDD is a more likely diagnosis than substance-induced MDD.

 

 

CASE CONTINUED: Planning treatment

A combination of behavioral therapy and pharmacotherapy is appropriate for treating Mr. G’s alcohol dependence. When you discuss the diagnosis with him, he endorses a goal of abstinence. For behavioral therapy, he says he would like to try Alcoholics Anonymous, which helped a friend “turn his life around.”

He has become accustomed to taking escitalopram once daily but is hesitant to take any medication that requires more frequent dosing. He also worries that medication might impair his work performance, which requires extensive periods of concentration. Your goal—as you consider available medications—is to develop a treatment plan that incorporates Mr. G’s preferences and addresses his concerns.

Disulfiram

Disulfiram, an irreversible aldehyde dehydrogenase inhibitor, is indicated for maintaining enforced sobriety in patients with chronic alcohol dependence (Table 1). Aldehyde dehydrogenase inhibition disrupts alcohol-to-acetate metabolism, which leads to acetaldehyde accumulation. If a disulfiram-treated patient ingests alcohol, increased acetaldehyde levels lead to the unpleasant “disulfiram-ethanol reaction,” with diaphoresis, flushing, nausea, vomiting, headache, tachycardia, and hypotension. The reaction’s severity is proportional to the disulfiram dose and amount of alcohol consumed.

Patients taking disulfiram must abstain from all alcohol, including over-the-counter cold remedies and mouthwashes containing alcohol. Advise patients that ingesting small amounts of alcohol can induce symptoms, even days after taking disulfiram.

Efficacy. Clinical trial results with disulfiram have been mixed. In the largest controlled study to date, Fuller et al7 found no significant difference in rates of total abstinence, time to first drink, employment, or social stability measures at 1 year among 605 men who received counseling plus disulfiram, 250 mg/d, or placebo. Other disulfiram studies have found a modest decrease in the frequency of drinking but no effect on abstinence rates.8,9

Because medication adherence is the strongest predictor of outcome with disulfiram,10 monitoring for adherence and stressing its importance to patients may increase the drug’s efficacy. Disulfiram may be most effective in highly motivated patients with stable social support or as an adjuvant to an outpatient treatment program.

Administration. Disulfiram is available in 250-mg tablets and is usually dosed from 125 to 500 mg/d. Treatment can begin after patients abstain from alcohol for ≥12 hours and have a serum alcohol concentration of zero.

Side effects. Drowsiness is a common complaint with disulfiram; this adverse effect is frequently self-limited and can be reduced by evening dosing.

Subclinical liver enzyme elevations have been reported in 25% of patients taking disulfiram.11 Although rare, potentially fatal hepatotoxicity has been reported12,13 (with a dose as low as 200 mg/d12), typically occurring early in treatment and associated with jaundice and fever. One study estimated the risk of dying of hepatotoxicity caused by disulfiram to be 1 in 30,000 patients/year.14

A recent Swedish study13 reviewed data from 1966 through 2002 and found 82 cases of drug-induced liver injury associated with disulfiram. By comparing these findings with sales figures from 1972 to 2002, the authors report an incidence of disulfiram-induced liver injury of about 1 case per 1.3 million estimated average daily doses.

Order liver function tests at baseline, then retest 10 to 14 days after starting disulfiram and again approximately 4 weeks later. Thereafter, monitoring once every 3 to 6 months is generally sufficient in patients without liver disease symptoms.

Other serious adverse events associated with disulfiram therapy include optic neuritis, peripheral neuritis, cholestatic hepatitis, seizures, and arrhythmias. Psychosis also can occur, generally with dosages ≥500 mg/d. Avoid concomitant use of disulfiram and metronidazole, which can cause acute psychosis.

Disulfiram-related inhibition of cytochrome P-450 can increase serum levels and toxicity risk of medications metabolized in the liver, such as warfarin, phenytoin, and isoniazid. Patients taking concomitant warfarin and disulfiram require close monitoring for increases in the international normalized ratio (INR).

Contraindications. Disulfiram is contraindicated in patients with ischemic heart disease and those who are pregnant. Also avoid disulfiram in patients with cerebrovascular disease, diabetes mellitus, psychosis, or cognitive impairment.

Recommendation. Disulfiram is a valid option for treating alcohol dependence in a select group of highly motivated patients who are medically and psychiatrically stable and in whom adherence can be closely monitored.

Table 1

Disulfiram: Fast facts

Mechanism: Acetaldehyde accumulates when aldehyde dehydrogenase is inhibited
FDA-approved for alcohol dependence: Yes
Dosing: 125 to 500 mg once daily
Effect: Aversive reaction to alcohol
Potential side effects: Liver toxicity, seizures, arrhythmia, peripheral neuropathy, psychosis
Contraindications: Concurrent alcohol consumption, severe cardiac disease, psychosis, pregnancy
Comments: Many drug interactions, including warfarin, metronidazole, and phenytoin; monitor liver function for toxicity
 

 

Naltrexone

Naltrexone is a μ-opioid receptor antagonist thought to reduce alcohol’s reinforcing effects by interfering with β-endorphin pathways. It is indicated for treating alcohol dependence and has been shown to reduce relapse and number of drinking days in alcohol-dependent patients (Table 2).15

Naltrexone also has reduced alcohol consumption in healthy volunteers, social drinkers, and other nondependent drinkers.16 Its effect may have a genetic component, as suggested by greater benefit in persons with a family history of alcoholism.17

Efficacy. Most studies investigated naltrexone as part of a comprehensive treatment program that included behavioral therapies.18 Recently, the randomized, placebo-controlled Combining Medications and Behavioral Interventions (COMBINE) study found that using either naltrexone or behavioral therapy improved abstinence, and combing naltrexone with behavioral therapy was not more effective than either treatment alone.19

Administration. Oral naltrexone is usually started at 25 mg and increased over 2 to 3 days to 50 or 100 mg/d. The standard dose is 50 mg/d, although the COMBINE study reported efficacy at 100 mg/d.19

Oral naltrexone is most helpful for patients who adhere to 70% to 90% of the medication.20 The extended-release form (a 380-mg IM dose given every 4 weeks) provides an option to monitor adherence.21

Side effects. Naltrexone toxicity can cause hepatocellular injury. Do not administer this drug to patients with acute hepatitis or end-stage liver disease. When prescribing naltrexone, check patients’ liver function monthly for the first 3 months, then once every 3 months thereafter.22 Less serious, common side effects include nausea, myalgia, and headache.

Naltrexone antagonizes opioid receptors and causes withdrawal symptoms in patients who are physically dependent on opioids. Therefore, do not give naltrexone to patients who require opioids for chronic pain. If your patient is using an opioid but could switch to other pain medication, discontinue the opioid for at least 7 days and consider a urine toxicology or naltrexone challenge before starting naltrexone.

Urine drug tests are inexpensive and easy to use but have limitations. Many standard “dipsticks” will detect heroin, morphine, and codeine but not oxycodone, hydrocodone, or other synthetic opioids. Specific tests are available to detect oxycodone, hydrocodone, hydromorphone, buprenorphine, and methadone. Some synthetic opioids (such as fentanyl) remain difficult to detect, however, because of their low concentration and rapid metabolism.

Table 2

Naltrexone: Fast facts

Mechanism: Opioid receptor antagonism interferes with β-endorphin pathways
FDA-approved for alcohol dependence: Yes
Dosing: Oral 50 mg once daily (recent evidence suggests safety and efficacy at 100 mg once daily); IM 380 mg once every 4 weeks
Effect: Decreases frequency and severity of relapse
Potential side effects: Nausea, myalgia, headache, dizziness
Contraindications: Opioid use, acute hepatitis, liver failure
Comments: Monitor liver function; once-monthly dosing may improve adherence

Acamprosate

Acamprosate is structurally similar to gamma-aminobutyric acid (GABA) and is thought to inhibit the glutamatergic system. This attenuation by acamprosate reduces the glutamatergic hyperactivity normally seen after chronic alcohol exposure.

Acamprosate is indicated for relapse prevention in patients with alcohol dependence who have stopped drinking (Table 3). Multiple randomized studies have demonstrated its efficacy in improving abstinence rates as compared with placebo.23,24 Other studies, however, failed to show improved abstinence with acamprosate.25 Some of the negative studies included patients who recently relapsed or had only a few days of abstinence before starting acamprosate.19,26 Therefore, acamprosate might be most effective when used to maintain abstinence and less effective—if at all—to initiate abstinence.

Administration. Acamprosate is available in 333-mg tablets, with a recommended dosage of 666 mg tid. Side effects tend to be transient and mild. Reduce the dose to 333 mg tid for patients with moderate renal insufficiency (creatinine clearance [CrCl] 30 to 50 mL/min), and do not use acamprosate in patients with severe renal insufficiency (CrCl

Side effects. Acamprosate was well-tolerated in clinical trials; diarrhea and other GI side effects were the most commonly reported adverse events.

In some placebo-controlled studies, patients taking acamprosate reported more frequent suicidal thoughts and attempts compared with patients taking placebo.27 These events were extremely rare, and no direct relationship with acamprosate therapy has been established. Nonetheless, monitor patients for depression and suicidal thoughts during acamprosate therapy.

Table 3

Acamprosate: Fast facts

Mechanism: Structurally similar to GABA; thought to inhibit the glutamatergic system
FDA-approved for alcohol dependence: Yes
Dosing: 666 mg tid
Effect: Increases abstinence
Potential side effects: Nausea, diarrhea, suicidal thoughts
Contraindications: Severe renal disease
Comments: Monitor patients for suicidal thoughts and depression

Topiramate

Topiramate potentiates GABA and inhibits excitatory glutamate transmission—properties believed to lead to decreased dopamine release at the nucleus accumbens in response to alcohol consumption. Although topiramate is not FDA-approved for alcohol dependence, limited data comparing this anticonvulsant with placebo have shown a reduction in drinking and increased abstinence (Table 4).28,29

 

 

Administration. Start with 25 mg/d and increase over several weeks to 300 mg/d, given in divided doses.

Side effects include dizziness, paresthesia, somnolence, difficulty concentrating, and weight loss. Because topiramate is excreted renally, reduce doses by 50% in patients with CrCl

Other renal side effects include an elevated risk of nephrolithiasis. Topiramate’s inhibition of carbonic anhydrase can reduce bicarbonate levels, leading to a nonanion gap metabolic acidosis.

Table 4

Topiramate: Fast facts

Mechanism: Potentiates GABA and inhibits glutamate receptor subtypes
FDA-approved for alcohol dependence: No
Dosing: 300 mg/d in divided doses
Effect: Decreases craving and drinking
Potential side effects: Metabolic acidosis, psychomotor slowing, dizziness, difficulty concentrating, paresthesia, weight loss, nephrolithiasis, hyperammonemia with concomitant use of valproic acid
Contraindications: None known other than hypersensitivity (as with all drugs)
Comments: Dose titration requires several weeks; avoid abrupt withdrawal; may reduce effectiveness of oral contraceptives

CASE CONTINUED: Implementing a treatment plan

You start Mr. G on oral naltrexone, 25 mg/d, and titrate to 100 mg/d. Although no optimum treatment duration has been established, you plan to follow NIAAA recommendations that Mr. G use naltrexone at least 3 months, with the possibility of continuing 1 year or longer if he responds well.1

You schedule weekly visits for the first month to monitor for side effects and to make any necessary modifications in behavioral and pharmacologic treatment. You also continue escitalopram, 10 mg, which has successfully controlled Mr. G’s MDD symptoms.

Medications for alcohol dependence generally have been studied as adjuncts to behavioral therapies. The COMBINE study of 1,383 alcohol-abstinent patients found naltrexone with medical management or cognitive-behavioral therapy alone to be equivalent in efficacy.19 The medical management provided a supportive environment, encouraged medication compliance, provided empathy to build a therapeutic relationship, and promoted self-help groups as an adjunct to treatment.30 Thus, medication has a role in treating alcohol dependence, but behavioral therapy remains an important part of comprehensive substance abuse treatment.

When choosing medications, consider the agents’ clinically relevant differences:

  • Naltrexone and—less conclusively—topiramate have shown benefit for alcoholdependent patients starting treatment and for relapse prevention.
  • Acamprosate may help prevent relapse in abstinent patients.
  • Disulfiram remains a valid option in highly motivated patients with social support available to ensure medication adherence.
Because Mr. G is starting therapy after recent alcohol use, medications such as naltrexone and topiramate that have shown benefit early in treatment (in addition to relapse prevention) are preferred. Of these 2 drugs, naltrexone is the better choice for Mr. G—who is concerned about his work performance—because difficulty concentrating is a common side effect of topiramate. Oral naltrexone would be preferred as initial therapy for Mr. G because he has expressed comfort taking once-daily oral medication. The more expensive oncemonthly naltrexone depot formulation could be a second-line treatment if adherence becomes an issue.

Hypersensitivity is considered a contraindication for any medication. Mr. G tolerates well an initial dose of 25 mg/d, followed by increases to 50 mg and then 100 mg over several days. You titrate oral naltrexone to 100 mg/d—even though it is commonly prescribed at 50 mg/d—because recent evidence suggests efficacy and safety at the higher dosage.19

Related resources

Drug brand names

  • Acamprosate • Campral
  • Disulfiram • Antabuse
  • Escitalopram • Lexapro
  • Naltrexone, oral • ReVia
  • Naltrexone, extended-release • Vivitrol
  • Topiramate • Topamax
  • Valproic acid • Depakote
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Mr. G, age 38, is an investment banker referred for evaluation of an alcohol use disorder. Three years ago his internist diagnosed Mr. G with major depression and prescribed a selective serotonin reuptake inhibitor. Mr. G’s mood has improved, but his drinking is out of control and is affecting his work and marriage.

Mr. G describes his father as an alcoholic and says he has noticed worrisome similarities in himself. Since his teenage years, he recalls always being able to drink more than his peers. Amnesia episodes began in college during heavy drinking days and now occur almost weekly. Most recently his driver’s license was suspended after he was arrested for driving while impaired by alcohol.

He has attempted to stop drinking 3 times in the last 6 months and feels frustrated because he continues to relapse. During his last quit attempt, he remained abstinent for 3 months and believes his mood was unchanged during that time.

For motivated patients such as Mr. G, National Institute on Alcohol Abuse and Alcoholism (NIAAA) guidelines (updated in 2007) consider medications first-line treatment for alcohol dependence, along with psychotherapies and mutual-help groups such as Alcoholics Anonymous.1 Medications with evidence of efficacy include FDA-approved disulfiram, naltrexone, and acamprosate, and off-label topiramate.

Each drug’s pharmacology is different; some may be beneficial during early abstinence, whereas others are more effective for maintaining abstinence. Because many physicians have had little or no experience using these medications,2,3 we discuss dosing recommendations and side effect profiles—important clinical differences to guide drug selection and administration.

Box

Alcohol dependence: Diagnostic criteria

Alcohol use disorders describe a maladaptive pattern of alcohol use that causes clinically significant impairment or distress. Alcohol dependence is manifested by ≥3 of the following symptoms in a 12-month period:

  • Tolerance
  • Withdrawal
  • Often drinking alcohol in larger amounts or over a longer time period than intended
  • Persistent desire or unsuccessful efforts to cut down or control alcohol use
  • Spending a great deal of time in activities necessary to obtain, use, or recover from alcohol’s effects
  • Giving up or reducing important social, occupational, or recreational activities because of alcohol use
  • Continuing alcohol use despite knowing you have a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by alcohol.

Source: Adapted from DSM-IV-TR

CASE CONTINUED: Which came first?

When Mr. G presented with depressive symptoms, his internist informed him that alcohol’s effects can mimic depression and advised him to cut back his consumption. Mr. G temporarily reduced his drinking but continued to experience depressed mood, sleep disturbances, difficulty concentrating, fatigue, and poor appetite. The internist then prescribed escitalopram, 10 mg/d, and Mr. G says his depressive symptoms improved. He has not attempted suicide or required psychiatric hospitalization.

After a thorough evaluation—including a detailed assessment of his drinking history, other substance use, and mood symptoms—you diagnose Mr. G with alcohol dependence without physiological dependence and a primary major depressive disorder (MDD).

Mr. G’s diagnosis of alcohol dependence is based on evidence in the last 12 months of tolerance, repeated loss of control over the amount he drinks, multiple failed attempts to stop drinking, repeated negative consequences to his work productivity and personal relationships, and continued drinking despite knowing that alcohol consumption sometimes intensifies his mood symptoms (Box). Physiological dependence is unlikely because he did not experience withdrawal during a recent period of abstinence.

The mood component. Particularly in psychiatric patients, alcohol dependence often coexists with and affects the treatment of other psychopathologies:

  • 1 in 3 adults experience an alcohol use disorder during their lifetimes.4
  • 1 in 3 adults with an alcohol use disorder have a comorbid psychiatric disorder.5
  • Alcohol dependence doubles the risk of major depression and triples the lifetime risk of any mood disorder.4
A thorough evaluation—both medical and psychiatric—is necessary to distinguish a primary mood disorder from a substance-induced mood disorder. Aspects of a patient’s history that may support a substance-induced disorder diagnosis include:

  • Mood symptoms appear after the onset of a substance use disorder.
  • Mood symptoms are absent during abstinence.
  • Mood symptoms are consistent with the effects of the drug being used.6
Substance-induced mood symptoms usually become less intense and eventually resolve as abstinence is maintained. Primary mood symptoms may not be affected by abstinence, and interventions—including pharmacotherapy—are more likely to be required.

In evaluating Mr. G’s drinking history, you determined that his mood symptoms began before he started consuming alcohol regularly and have persisted during periods of abstinence. Thus, primary MDD is a more likely diagnosis than substance-induced MDD.

 

 

CASE CONTINUED: Planning treatment

A combination of behavioral therapy and pharmacotherapy is appropriate for treating Mr. G’s alcohol dependence. When you discuss the diagnosis with him, he endorses a goal of abstinence. For behavioral therapy, he says he would like to try Alcoholics Anonymous, which helped a friend “turn his life around.”

He has become accustomed to taking escitalopram once daily but is hesitant to take any medication that requires more frequent dosing. He also worries that medication might impair his work performance, which requires extensive periods of concentration. Your goal—as you consider available medications—is to develop a treatment plan that incorporates Mr. G’s preferences and addresses his concerns.

Disulfiram

Disulfiram, an irreversible aldehyde dehydrogenase inhibitor, is indicated for maintaining enforced sobriety in patients with chronic alcohol dependence (Table 1). Aldehyde dehydrogenase inhibition disrupts alcohol-to-acetate metabolism, which leads to acetaldehyde accumulation. If a disulfiram-treated patient ingests alcohol, increased acetaldehyde levels lead to the unpleasant “disulfiram-ethanol reaction,” with diaphoresis, flushing, nausea, vomiting, headache, tachycardia, and hypotension. The reaction’s severity is proportional to the disulfiram dose and amount of alcohol consumed.

Patients taking disulfiram must abstain from all alcohol, including over-the-counter cold remedies and mouthwashes containing alcohol. Advise patients that ingesting small amounts of alcohol can induce symptoms, even days after taking disulfiram.

Efficacy. Clinical trial results with disulfiram have been mixed. In the largest controlled study to date, Fuller et al7 found no significant difference in rates of total abstinence, time to first drink, employment, or social stability measures at 1 year among 605 men who received counseling plus disulfiram, 250 mg/d, or placebo. Other disulfiram studies have found a modest decrease in the frequency of drinking but no effect on abstinence rates.8,9

Because medication adherence is the strongest predictor of outcome with disulfiram,10 monitoring for adherence and stressing its importance to patients may increase the drug’s efficacy. Disulfiram may be most effective in highly motivated patients with stable social support or as an adjuvant to an outpatient treatment program.

Administration. Disulfiram is available in 250-mg tablets and is usually dosed from 125 to 500 mg/d. Treatment can begin after patients abstain from alcohol for ≥12 hours and have a serum alcohol concentration of zero.

Side effects. Drowsiness is a common complaint with disulfiram; this adverse effect is frequently self-limited and can be reduced by evening dosing.

Subclinical liver enzyme elevations have been reported in 25% of patients taking disulfiram.11 Although rare, potentially fatal hepatotoxicity has been reported12,13 (with a dose as low as 200 mg/d12), typically occurring early in treatment and associated with jaundice and fever. One study estimated the risk of dying of hepatotoxicity caused by disulfiram to be 1 in 30,000 patients/year.14

A recent Swedish study13 reviewed data from 1966 through 2002 and found 82 cases of drug-induced liver injury associated with disulfiram. By comparing these findings with sales figures from 1972 to 2002, the authors report an incidence of disulfiram-induced liver injury of about 1 case per 1.3 million estimated average daily doses.

Order liver function tests at baseline, then retest 10 to 14 days after starting disulfiram and again approximately 4 weeks later. Thereafter, monitoring once every 3 to 6 months is generally sufficient in patients without liver disease symptoms.

Other serious adverse events associated with disulfiram therapy include optic neuritis, peripheral neuritis, cholestatic hepatitis, seizures, and arrhythmias. Psychosis also can occur, generally with dosages ≥500 mg/d. Avoid concomitant use of disulfiram and metronidazole, which can cause acute psychosis.

Disulfiram-related inhibition of cytochrome P-450 can increase serum levels and toxicity risk of medications metabolized in the liver, such as warfarin, phenytoin, and isoniazid. Patients taking concomitant warfarin and disulfiram require close monitoring for increases in the international normalized ratio (INR).

Contraindications. Disulfiram is contraindicated in patients with ischemic heart disease and those who are pregnant. Also avoid disulfiram in patients with cerebrovascular disease, diabetes mellitus, psychosis, or cognitive impairment.

Recommendation. Disulfiram is a valid option for treating alcohol dependence in a select group of highly motivated patients who are medically and psychiatrically stable and in whom adherence can be closely monitored.

Table 1

Disulfiram: Fast facts

Mechanism: Acetaldehyde accumulates when aldehyde dehydrogenase is inhibited
FDA-approved for alcohol dependence: Yes
Dosing: 125 to 500 mg once daily
Effect: Aversive reaction to alcohol
Potential side effects: Liver toxicity, seizures, arrhythmia, peripheral neuropathy, psychosis
Contraindications: Concurrent alcohol consumption, severe cardiac disease, psychosis, pregnancy
Comments: Many drug interactions, including warfarin, metronidazole, and phenytoin; monitor liver function for toxicity
 

 

Naltrexone

Naltrexone is a μ-opioid receptor antagonist thought to reduce alcohol’s reinforcing effects by interfering with β-endorphin pathways. It is indicated for treating alcohol dependence and has been shown to reduce relapse and number of drinking days in alcohol-dependent patients (Table 2).15

Naltrexone also has reduced alcohol consumption in healthy volunteers, social drinkers, and other nondependent drinkers.16 Its effect may have a genetic component, as suggested by greater benefit in persons with a family history of alcoholism.17

Efficacy. Most studies investigated naltrexone as part of a comprehensive treatment program that included behavioral therapies.18 Recently, the randomized, placebo-controlled Combining Medications and Behavioral Interventions (COMBINE) study found that using either naltrexone or behavioral therapy improved abstinence, and combing naltrexone with behavioral therapy was not more effective than either treatment alone.19

Administration. Oral naltrexone is usually started at 25 mg and increased over 2 to 3 days to 50 or 100 mg/d. The standard dose is 50 mg/d, although the COMBINE study reported efficacy at 100 mg/d.19

Oral naltrexone is most helpful for patients who adhere to 70% to 90% of the medication.20 The extended-release form (a 380-mg IM dose given every 4 weeks) provides an option to monitor adherence.21

Side effects. Naltrexone toxicity can cause hepatocellular injury. Do not administer this drug to patients with acute hepatitis or end-stage liver disease. When prescribing naltrexone, check patients’ liver function monthly for the first 3 months, then once every 3 months thereafter.22 Less serious, common side effects include nausea, myalgia, and headache.

Naltrexone antagonizes opioid receptors and causes withdrawal symptoms in patients who are physically dependent on opioids. Therefore, do not give naltrexone to patients who require opioids for chronic pain. If your patient is using an opioid but could switch to other pain medication, discontinue the opioid for at least 7 days and consider a urine toxicology or naltrexone challenge before starting naltrexone.

Urine drug tests are inexpensive and easy to use but have limitations. Many standard “dipsticks” will detect heroin, morphine, and codeine but not oxycodone, hydrocodone, or other synthetic opioids. Specific tests are available to detect oxycodone, hydrocodone, hydromorphone, buprenorphine, and methadone. Some synthetic opioids (such as fentanyl) remain difficult to detect, however, because of their low concentration and rapid metabolism.

Table 2

Naltrexone: Fast facts

Mechanism: Opioid receptor antagonism interferes with β-endorphin pathways
FDA-approved for alcohol dependence: Yes
Dosing: Oral 50 mg once daily (recent evidence suggests safety and efficacy at 100 mg once daily); IM 380 mg once every 4 weeks
Effect: Decreases frequency and severity of relapse
Potential side effects: Nausea, myalgia, headache, dizziness
Contraindications: Opioid use, acute hepatitis, liver failure
Comments: Monitor liver function; once-monthly dosing may improve adherence

Acamprosate

Acamprosate is structurally similar to gamma-aminobutyric acid (GABA) and is thought to inhibit the glutamatergic system. This attenuation by acamprosate reduces the glutamatergic hyperactivity normally seen after chronic alcohol exposure.

Acamprosate is indicated for relapse prevention in patients with alcohol dependence who have stopped drinking (Table 3). Multiple randomized studies have demonstrated its efficacy in improving abstinence rates as compared with placebo.23,24 Other studies, however, failed to show improved abstinence with acamprosate.25 Some of the negative studies included patients who recently relapsed or had only a few days of abstinence before starting acamprosate.19,26 Therefore, acamprosate might be most effective when used to maintain abstinence and less effective—if at all—to initiate abstinence.

Administration. Acamprosate is available in 333-mg tablets, with a recommended dosage of 666 mg tid. Side effects tend to be transient and mild. Reduce the dose to 333 mg tid for patients with moderate renal insufficiency (creatinine clearance [CrCl] 30 to 50 mL/min), and do not use acamprosate in patients with severe renal insufficiency (CrCl

Side effects. Acamprosate was well-tolerated in clinical trials; diarrhea and other GI side effects were the most commonly reported adverse events.

In some placebo-controlled studies, patients taking acamprosate reported more frequent suicidal thoughts and attempts compared with patients taking placebo.27 These events were extremely rare, and no direct relationship with acamprosate therapy has been established. Nonetheless, monitor patients for depression and suicidal thoughts during acamprosate therapy.

Table 3

Acamprosate: Fast facts

Mechanism: Structurally similar to GABA; thought to inhibit the glutamatergic system
FDA-approved for alcohol dependence: Yes
Dosing: 666 mg tid
Effect: Increases abstinence
Potential side effects: Nausea, diarrhea, suicidal thoughts
Contraindications: Severe renal disease
Comments: Monitor patients for suicidal thoughts and depression

Topiramate

Topiramate potentiates GABA and inhibits excitatory glutamate transmission—properties believed to lead to decreased dopamine release at the nucleus accumbens in response to alcohol consumption. Although topiramate is not FDA-approved for alcohol dependence, limited data comparing this anticonvulsant with placebo have shown a reduction in drinking and increased abstinence (Table 4).28,29

 

 

Administration. Start with 25 mg/d and increase over several weeks to 300 mg/d, given in divided doses.

Side effects include dizziness, paresthesia, somnolence, difficulty concentrating, and weight loss. Because topiramate is excreted renally, reduce doses by 50% in patients with CrCl

Other renal side effects include an elevated risk of nephrolithiasis. Topiramate’s inhibition of carbonic anhydrase can reduce bicarbonate levels, leading to a nonanion gap metabolic acidosis.

Table 4

Topiramate: Fast facts

Mechanism: Potentiates GABA and inhibits glutamate receptor subtypes
FDA-approved for alcohol dependence: No
Dosing: 300 mg/d in divided doses
Effect: Decreases craving and drinking
Potential side effects: Metabolic acidosis, psychomotor slowing, dizziness, difficulty concentrating, paresthesia, weight loss, nephrolithiasis, hyperammonemia with concomitant use of valproic acid
Contraindications: None known other than hypersensitivity (as with all drugs)
Comments: Dose titration requires several weeks; avoid abrupt withdrawal; may reduce effectiveness of oral contraceptives

CASE CONTINUED: Implementing a treatment plan

You start Mr. G on oral naltrexone, 25 mg/d, and titrate to 100 mg/d. Although no optimum treatment duration has been established, you plan to follow NIAAA recommendations that Mr. G use naltrexone at least 3 months, with the possibility of continuing 1 year or longer if he responds well.1

You schedule weekly visits for the first month to monitor for side effects and to make any necessary modifications in behavioral and pharmacologic treatment. You also continue escitalopram, 10 mg, which has successfully controlled Mr. G’s MDD symptoms.

Medications for alcohol dependence generally have been studied as adjuncts to behavioral therapies. The COMBINE study of 1,383 alcohol-abstinent patients found naltrexone with medical management or cognitive-behavioral therapy alone to be equivalent in efficacy.19 The medical management provided a supportive environment, encouraged medication compliance, provided empathy to build a therapeutic relationship, and promoted self-help groups as an adjunct to treatment.30 Thus, medication has a role in treating alcohol dependence, but behavioral therapy remains an important part of comprehensive substance abuse treatment.

When choosing medications, consider the agents’ clinically relevant differences:

  • Naltrexone and—less conclusively—topiramate have shown benefit for alcoholdependent patients starting treatment and for relapse prevention.
  • Acamprosate may help prevent relapse in abstinent patients.
  • Disulfiram remains a valid option in highly motivated patients with social support available to ensure medication adherence.
Because Mr. G is starting therapy after recent alcohol use, medications such as naltrexone and topiramate that have shown benefit early in treatment (in addition to relapse prevention) are preferred. Of these 2 drugs, naltrexone is the better choice for Mr. G—who is concerned about his work performance—because difficulty concentrating is a common side effect of topiramate. Oral naltrexone would be preferred as initial therapy for Mr. G because he has expressed comfort taking once-daily oral medication. The more expensive oncemonthly naltrexone depot formulation could be a second-line treatment if adherence becomes an issue.

Hypersensitivity is considered a contraindication for any medication. Mr. G tolerates well an initial dose of 25 mg/d, followed by increases to 50 mg and then 100 mg over several days. You titrate oral naltrexone to 100 mg/d—even though it is commonly prescribed at 50 mg/d—because recent evidence suggests efficacy and safety at the higher dosage.19

Related resources

Drug brand names

  • Acamprosate • Campral
  • Disulfiram • Antabuse
  • Escitalopram • Lexapro
  • Naltrexone, oral • ReVia
  • Naltrexone, extended-release • Vivitrol
  • Topiramate • Topamax
  • Valproic acid • Depakote
Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. National Institutes of Health. Helping patients who drink too much: a clinician’s guide. Bethesda, MD: National Institute on Alcohol Abuse and Alcoholism; 2007. NIH Publication 07-3769. Available at: http://www.niaaa.nih.gov/Publications/EducationTrainingMaterials/guide.htm. Accessed January 3, 2008.

2. Substance Abuse and Mental Health Services Administration. National Survey of Substance Abuse Treatment Services: 2005. Data on substance abuse treatment facilities, (DASIS Series: S-34). Rockville, MD: Office of Applied Studies, 2006. DHHS Publication (SMA) 06-4206.

3. Substance Abuse and Mental Health Services Administration. Treatment episode data set (TEDS) highlights 2005: National admissions to substance abuse treatment services. Rockville, MD: Office of Applied Studies. 2006. DHHS Publication (SMA) 07-4229.

4. Hasin DS, Stinson FS, Ogburn EO, Grant BF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Arch Gen Psychiatry 2007;64(7):830-42.

5. Regier DA, Farmer ME, Rae DS, et al. Comorbidity of mental disorders with alcohol and other drug abuse. Results from the Epidemiologic Catchment Area (ECA) study. JAMA. 1990;264(19):2511-8.

6. Mariani JJ, Levin FR. Treatment of comorbid conditions of substance abuse. Directions in Psychiatry 2005;25(2):129-39.

7. Fuller RK, Branchey L, Brightwell DR, et al. Disulfiram treatment of alcoholism. A Veteran’s Administration cooperative study. JAMA. 1986;256:1449-55.

8. Fuller RK, Roth HP. Disulfiram for the treatment of alcoholism. An evaluation of 128 men. Ann Intern Med. 1979;90:901-4.

9. Schuckit MA. A one-year follow-up of men alcoholics given disulfiram. J Stud Alcohol 1985;46:191-5.

10. Collins GB, McAllister MS, Adury K. Drug adjuncts for treating alcohol dependence. Cleve Clin J Med 2006;73(7):641-4.

11. Goyer PF, Major LF. Hepatotoxicty in disulfiram treated patients. J Stud Alcohol 1979;40:133-7.

12. Ranek L, Buch Andreasen P. Disulfiram hepatotoxicity. Br Med J 1977;2(6079):94-6.

13. Björnsson E, Nordlinder H, Olsson R. Clinical characteristics and prognostic markers in disulfiram-induced liver injury. J Hepatol 2006;44(4):791-7.

14. Enghusen PH, Loft S, Anderson JR, et al. Disulfiram therapy—adverse drug reactions and interactions. Acta Psychiatr Scand 1992;86(369):59-66.

15. Volpicelli JR, Volpicelli LA, O’Brien CP. Medical management of alcohol dependence: clinical use and limitations of naltrexone treatment. Alcohol Alcohol 1995;30:789.-

16. de Wit H, Svenson J, York A. Non-specific effect of naltrexone on ethanol consumption in social drinkers. Psychopharmacology (Berl) 1999;146:33.-

17. Monterosso JR, Flannery BA, Pettinati HM, et al. Predicting treatment response to naltrexone: the influence of craving and family history. Am J Addict 2001;10:258-68.

18. Srisurapanont M, Jarusuraisin N. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev 2005;(1):CD001867.-

19. Anton RF, O’Malley SS, Ciraulo DA, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295:2003-17.

20. Monti PM, Rohsenow DJ, Swift RM, et al. Naltrexone and cue exposure with coping and communication skills training for alcoholics: treatment process and 1-year outcomes. Alcol Clin Exp Res 2001;25:1634-47.

21. Garbutt JC, Kranzler HR, O’Malley SS, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293(13):1617-25.

22. Doering PL. Substance-related disorders: alcohol, nicotine, and caffeine. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. 4th ed. Stamford, CT: Appleton & Lange; 1999.

23. Mann K, Lehert P, Morgan MY. The efficacy of acamprosate in the maintenance of abstinence in alcohol-dependent individuals: results of a meta-analysis. Alcohol Clin Exp Res 2004;28(1):51-63.

24. Paille FM, Guelfi JD, Perkins AC, et al. Double-blind randomized multicentre trial of acamprosate in maintaining abstinence from alcohol. Alcohol Alcohol 1995;30:239-47.

25. Chick J, Howlett H, Morgan MY, Ritson B. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol Alcohol 2000;35(2):176-87.

26. Mason BJ, Goodman AM, Chabac S, Lehert P. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind, placebo-controlled trial: the role of patient motivation. J Psychiatr Res 2006;40(5):383-93.

27. Mason BJ. Treatment of alcohol-dependent outpatients with acamprosate: a clinical review. J Clin Psychiatry 2001;62(suppl 20):42-8.

28. Johnson BA, Ait-Daoud N, Bowden CL, et al. Oral topiramate for treatment of alcohol dependence: a randomized controlled trial. Lancet 2003;361(9370):1677-85.

29. Johnson BA, Rosenthal N, Capece JA, et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA 2007;298(14):1641-51.

30. COMBINE Study Research Group. Testing combined pharmacotherapies and behavioral interventions in alcohol dependence: rationale and methods. Alcohol Clin Exp Res 2003;27:1107-22.

References

1. National Institutes of Health. Helping patients who drink too much: a clinician’s guide. Bethesda, MD: National Institute on Alcohol Abuse and Alcoholism; 2007. NIH Publication 07-3769. Available at: http://www.niaaa.nih.gov/Publications/EducationTrainingMaterials/guide.htm. Accessed January 3, 2008.

2. Substance Abuse and Mental Health Services Administration. National Survey of Substance Abuse Treatment Services: 2005. Data on substance abuse treatment facilities, (DASIS Series: S-34). Rockville, MD: Office of Applied Studies, 2006. DHHS Publication (SMA) 06-4206.

3. Substance Abuse and Mental Health Services Administration. Treatment episode data set (TEDS) highlights 2005: National admissions to substance abuse treatment services. Rockville, MD: Office of Applied Studies. 2006. DHHS Publication (SMA) 07-4229.

4. Hasin DS, Stinson FS, Ogburn EO, Grant BF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States. Arch Gen Psychiatry 2007;64(7):830-42.

5. Regier DA, Farmer ME, Rae DS, et al. Comorbidity of mental disorders with alcohol and other drug abuse. Results from the Epidemiologic Catchment Area (ECA) study. JAMA. 1990;264(19):2511-8.

6. Mariani JJ, Levin FR. Treatment of comorbid conditions of substance abuse. Directions in Psychiatry 2005;25(2):129-39.

7. Fuller RK, Branchey L, Brightwell DR, et al. Disulfiram treatment of alcoholism. A Veteran’s Administration cooperative study. JAMA. 1986;256:1449-55.

8. Fuller RK, Roth HP. Disulfiram for the treatment of alcoholism. An evaluation of 128 men. Ann Intern Med. 1979;90:901-4.

9. Schuckit MA. A one-year follow-up of men alcoholics given disulfiram. J Stud Alcohol 1985;46:191-5.

10. Collins GB, McAllister MS, Adury K. Drug adjuncts for treating alcohol dependence. Cleve Clin J Med 2006;73(7):641-4.

11. Goyer PF, Major LF. Hepatotoxicty in disulfiram treated patients. J Stud Alcohol 1979;40:133-7.

12. Ranek L, Buch Andreasen P. Disulfiram hepatotoxicity. Br Med J 1977;2(6079):94-6.

13. Björnsson E, Nordlinder H, Olsson R. Clinical characteristics and prognostic markers in disulfiram-induced liver injury. J Hepatol 2006;44(4):791-7.

14. Enghusen PH, Loft S, Anderson JR, et al. Disulfiram therapy—adverse drug reactions and interactions. Acta Psychiatr Scand 1992;86(369):59-66.

15. Volpicelli JR, Volpicelli LA, O’Brien CP. Medical management of alcohol dependence: clinical use and limitations of naltrexone treatment. Alcohol Alcohol 1995;30:789.-

16. de Wit H, Svenson J, York A. Non-specific effect of naltrexone on ethanol consumption in social drinkers. Psychopharmacology (Berl) 1999;146:33.-

17. Monterosso JR, Flannery BA, Pettinati HM, et al. Predicting treatment response to naltrexone: the influence of craving and family history. Am J Addict 2001;10:258-68.

18. Srisurapanont M, Jarusuraisin N. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev 2005;(1):CD001867.-

19. Anton RF, O’Malley SS, Ciraulo DA, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295:2003-17.

20. Monti PM, Rohsenow DJ, Swift RM, et al. Naltrexone and cue exposure with coping and communication skills training for alcoholics: treatment process and 1-year outcomes. Alcol Clin Exp Res 2001;25:1634-47.

21. Garbutt JC, Kranzler HR, O’Malley SS, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293(13):1617-25.

22. Doering PL. Substance-related disorders: alcohol, nicotine, and caffeine. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. 4th ed. Stamford, CT: Appleton & Lange; 1999.

23. Mann K, Lehert P, Morgan MY. The efficacy of acamprosate in the maintenance of abstinence in alcohol-dependent individuals: results of a meta-analysis. Alcohol Clin Exp Res 2004;28(1):51-63.

24. Paille FM, Guelfi JD, Perkins AC, et al. Double-blind randomized multicentre trial of acamprosate in maintaining abstinence from alcohol. Alcohol Alcohol 1995;30:239-47.

25. Chick J, Howlett H, Morgan MY, Ritson B. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol Alcohol 2000;35(2):176-87.

26. Mason BJ, Goodman AM, Chabac S, Lehert P. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind, placebo-controlled trial: the role of patient motivation. J Psychiatr Res 2006;40(5):383-93.

27. Mason BJ. Treatment of alcohol-dependent outpatients with acamprosate: a clinical review. J Clin Psychiatry 2001;62(suppl 20):42-8.

28. Johnson BA, Ait-Daoud N, Bowden CL, et al. Oral topiramate for treatment of alcohol dependence: a randomized controlled trial. Lancet 2003;361(9370):1677-85.

29. Johnson BA, Rosenthal N, Capece JA, et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA 2007;298(14):1641-51.

30. COMBINE Study Research Group. Testing combined pharmacotherapies and behavioral interventions in alcohol dependence: rationale and methods. Alcohol Clin Exp Res 2003;27:1107-22.

Issue
Current Psychiatry - 07(02)
Issue
Current Psychiatry - 07(02)
Page Number
25-37
Page Number
25-37
Publications
Publications
Article Type
Display Headline
Treating alcohol dependence: When and how to use 4 medications
Display Headline
Treating alcohol dependence: When and how to use 4 medications
Legacy Keywords
alcohol dependence; Soteri Polydorou MD; Frances R. Levin MD; disulfiram; naltrexone; acamprosate; topiramate; alcohol use disorders; substance-induced mood symptoms
Legacy Keywords
alcohol dependence; Soteri Polydorou MD; Frances R. Levin MD; disulfiram; naltrexone; acamprosate; topiramate; alcohol use disorders; substance-induced mood symptoms
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Is your patient in marijuana withdrawal?

Article Type
Changed
Tue, 12/11/2018 - 15:21
Display Headline
Is your patient in marijuana withdrawal?

Marijuana-dependent patients often report that withdrawal symptoms hinder their attempts to quit and trigger relapse. Up to 95% of marijuana users report withdrawal symptoms during abstinence, and you can gauge a patient’s withdrawal risk by knowing how much cannabis he or she has used.1

Light to heavy use

Using greater amounts of marijuana, meeting criteria for abuse or dependence, and a history of chronic use (>10 years) increase the risk of developing clinically significant withdrawal symptoms.1,2

  • Light use —smoking ≤2 joints/day on fewer than 4 days a week—is associated with low withdrawal risk.1
  • Moderate use—2 to 3 joints almost daily—results in significant withdrawal symptoms in 50% to 60% of marijuana users, with the higher rate associated with chronic use.1,2
  • Heavy use—≥4 joints daily or almost daily—usually results in significant withdrawal symptoms.1

Symptoms

Marijuana withdrawal is associated with neurovegetative symptoms, such as loss of appetite that can result in transient weight loss; trouble sleeping or sleep disrupted by strange dreams; and physical malaise, such as abdominal discomfort, chills, and feeling “shaky.”1 Patients may also report psychiatric symptoms such as anxiety, irritability, or depressed mood.2

Most studies show irritability, appetite changes, sleep disruption, and anxiety occur more frequently than craving, abdominal discomfort, and increased sex drive during marijuana abstinence.1 One preliminary study found that women were more likely to describe abdominal discomfort and men were more likely to report marijuana craving and increased sex drive during withdrawal.3

Withdrawal symptoms usually begin 24 to 48 hours after patients’ last marijuana use, and most resolve within 2 to 4 weeks. In some cases, anxiety and irritability can persist >4 weeks.

Recommendations

Determine if your patient’s marijuana use is light, moderate, or heavy by asking about lifetime and current use. Based on these answers, the risk of withdrawal is 50%.

If irritability is prominent in a light user, consider causes other than withdrawal. In a patient with chronic anxiety symptoms and light marijuana use, consider starting medication for the anxiety. Provide psycho-education and supportive therapy to help a patient with heavy marijuana use and acute anxiety get through the withdrawal period.

References

1. Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and signifi cance of cannabis withdrawal syndrome. Am J Psychiatry 2004;161(11):1967-77.

2. Kouri EM, Pope HG, Jr. Abstinence symptoms during withdrawal from chronic marijuana use. Exp Clin Psychopharmacol 2000;8(4):483-92.

3. Copersino ML, Boyd SJ, Tashkin DP, et.al. Gender differences in the experience of spontaneous cannabis quitting. Presented at: Annual Meeting of the College on Problems of Drug Dependence; June 17-22, 2006; Scottsdale, AZ.

Article PDF
Author and Disclosure Information

Marina Goldman, MD
Dimitri Markov, MD
Elisabeth J.S. Kunkel, MD
Ron Ehrman, PhD
Dr. Goldman is an addiction fellow at the University of Pennsylvania. Dr. Markov is assistant professor of psychiatry and Dr. Kunkel is professor of psychiatry at Thomas Jefferson University. Dr. Ehrman is associate clinical professor of psychology, University of Pennsylvania.

Issue
Current Psychiatry - 07(01)
Publications
Page Number
110-110
Legacy Keywords
marijuana withdrawal; marijuana-dependent patients; Marina Goldman MD; Dimitri Markov MD; Elisabeth JS Kunkel MD; Ron Ehrman PhD
Sections
Author and Disclosure Information

Marina Goldman, MD
Dimitri Markov, MD
Elisabeth J.S. Kunkel, MD
Ron Ehrman, PhD
Dr. Goldman is an addiction fellow at the University of Pennsylvania. Dr. Markov is assistant professor of psychiatry and Dr. Kunkel is professor of psychiatry at Thomas Jefferson University. Dr. Ehrman is associate clinical professor of psychology, University of Pennsylvania.

Author and Disclosure Information

Marina Goldman, MD
Dimitri Markov, MD
Elisabeth J.S. Kunkel, MD
Ron Ehrman, PhD
Dr. Goldman is an addiction fellow at the University of Pennsylvania. Dr. Markov is assistant professor of psychiatry and Dr. Kunkel is professor of psychiatry at Thomas Jefferson University. Dr. Ehrman is associate clinical professor of psychology, University of Pennsylvania.

Article PDF
Article PDF

Marijuana-dependent patients often report that withdrawal symptoms hinder their attempts to quit and trigger relapse. Up to 95% of marijuana users report withdrawal symptoms during abstinence, and you can gauge a patient’s withdrawal risk by knowing how much cannabis he or she has used.1

Light to heavy use

Using greater amounts of marijuana, meeting criteria for abuse or dependence, and a history of chronic use (>10 years) increase the risk of developing clinically significant withdrawal symptoms.1,2

  • Light use —smoking ≤2 joints/day on fewer than 4 days a week—is associated with low withdrawal risk.1
  • Moderate use—2 to 3 joints almost daily—results in significant withdrawal symptoms in 50% to 60% of marijuana users, with the higher rate associated with chronic use.1,2
  • Heavy use—≥4 joints daily or almost daily—usually results in significant withdrawal symptoms.1

Symptoms

Marijuana withdrawal is associated with neurovegetative symptoms, such as loss of appetite that can result in transient weight loss; trouble sleeping or sleep disrupted by strange dreams; and physical malaise, such as abdominal discomfort, chills, and feeling “shaky.”1 Patients may also report psychiatric symptoms such as anxiety, irritability, or depressed mood.2

Most studies show irritability, appetite changes, sleep disruption, and anxiety occur more frequently than craving, abdominal discomfort, and increased sex drive during marijuana abstinence.1 One preliminary study found that women were more likely to describe abdominal discomfort and men were more likely to report marijuana craving and increased sex drive during withdrawal.3

Withdrawal symptoms usually begin 24 to 48 hours after patients’ last marijuana use, and most resolve within 2 to 4 weeks. In some cases, anxiety and irritability can persist >4 weeks.

Recommendations

Determine if your patient’s marijuana use is light, moderate, or heavy by asking about lifetime and current use. Based on these answers, the risk of withdrawal is 50%.

If irritability is prominent in a light user, consider causes other than withdrawal. In a patient with chronic anxiety symptoms and light marijuana use, consider starting medication for the anxiety. Provide psycho-education and supportive therapy to help a patient with heavy marijuana use and acute anxiety get through the withdrawal period.

Marijuana-dependent patients often report that withdrawal symptoms hinder their attempts to quit and trigger relapse. Up to 95% of marijuana users report withdrawal symptoms during abstinence, and you can gauge a patient’s withdrawal risk by knowing how much cannabis he or she has used.1

Light to heavy use

Using greater amounts of marijuana, meeting criteria for abuse or dependence, and a history of chronic use (>10 years) increase the risk of developing clinically significant withdrawal symptoms.1,2

  • Light use —smoking ≤2 joints/day on fewer than 4 days a week—is associated with low withdrawal risk.1
  • Moderate use—2 to 3 joints almost daily—results in significant withdrawal symptoms in 50% to 60% of marijuana users, with the higher rate associated with chronic use.1,2
  • Heavy use—≥4 joints daily or almost daily—usually results in significant withdrawal symptoms.1

Symptoms

Marijuana withdrawal is associated with neurovegetative symptoms, such as loss of appetite that can result in transient weight loss; trouble sleeping or sleep disrupted by strange dreams; and physical malaise, such as abdominal discomfort, chills, and feeling “shaky.”1 Patients may also report psychiatric symptoms such as anxiety, irritability, or depressed mood.2

Most studies show irritability, appetite changes, sleep disruption, and anxiety occur more frequently than craving, abdominal discomfort, and increased sex drive during marijuana abstinence.1 One preliminary study found that women were more likely to describe abdominal discomfort and men were more likely to report marijuana craving and increased sex drive during withdrawal.3

Withdrawal symptoms usually begin 24 to 48 hours after patients’ last marijuana use, and most resolve within 2 to 4 weeks. In some cases, anxiety and irritability can persist >4 weeks.

Recommendations

Determine if your patient’s marijuana use is light, moderate, or heavy by asking about lifetime and current use. Based on these answers, the risk of withdrawal is 50%.

If irritability is prominent in a light user, consider causes other than withdrawal. In a patient with chronic anxiety symptoms and light marijuana use, consider starting medication for the anxiety. Provide psycho-education and supportive therapy to help a patient with heavy marijuana use and acute anxiety get through the withdrawal period.

References

1. Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and signifi cance of cannabis withdrawal syndrome. Am J Psychiatry 2004;161(11):1967-77.

2. Kouri EM, Pope HG, Jr. Abstinence symptoms during withdrawal from chronic marijuana use. Exp Clin Psychopharmacol 2000;8(4):483-92.

3. Copersino ML, Boyd SJ, Tashkin DP, et.al. Gender differences in the experience of spontaneous cannabis quitting. Presented at: Annual Meeting of the College on Problems of Drug Dependence; June 17-22, 2006; Scottsdale, AZ.

References

1. Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and signifi cance of cannabis withdrawal syndrome. Am J Psychiatry 2004;161(11):1967-77.

2. Kouri EM, Pope HG, Jr. Abstinence symptoms during withdrawal from chronic marijuana use. Exp Clin Psychopharmacol 2000;8(4):483-92.

3. Copersino ML, Boyd SJ, Tashkin DP, et.al. Gender differences in the experience of spontaneous cannabis quitting. Presented at: Annual Meeting of the College on Problems of Drug Dependence; June 17-22, 2006; Scottsdale, AZ.

Issue
Current Psychiatry - 07(01)
Issue
Current Psychiatry - 07(01)
Page Number
110-110
Page Number
110-110
Publications
Publications
Article Type
Display Headline
Is your patient in marijuana withdrawal?
Display Headline
Is your patient in marijuana withdrawal?
Legacy Keywords
marijuana withdrawal; marijuana-dependent patients; Marina Goldman MD; Dimitri Markov MD; Elisabeth JS Kunkel MD; Ron Ehrman PhD
Legacy Keywords
marijuana withdrawal; marijuana-dependent patients; Marina Goldman MD; Dimitri Markov MD; Elisabeth JS Kunkel MD; Ron Ehrman PhD
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Depression or chronic fatigue syndrome?

Article Type
Changed
Tue, 12/11/2018 - 15:21
Display Headline
Depression or chronic fatigue syndrome?

Chronic fatigue syndrome (CFS) is characterized by prolonged, debilitating fatigue that does not improve with rest and may be worsened by physical and mental activity. This fatigue must be present for at least 6 months. This syndrome affects more than 1 million individuals in the U.S. and shares symptoms with many medical and psychiatric illnesses, particularly depression.

Keeping in mind that a patient who presents with fatigue could have depression and CFS, follow DSM-IV-TR criteria for major depressive disorder to help identify clinical factors that distinguish the 2 disorders.1

1 Depressed mood

Individuals with major depressive disorder report being depressed most of time and often cannot provide a reason or identify a loss to explain their depressed mood. CFS patients are more likely to report feeling discouraged and depressed because a lack of energy leaves them unable to complete tasks.

2 Diminished interest in activities

Depressed patients typically report a loss or lack of interest in pleasurable activities. CFS patients say they would engage in favorite activities more if their energy level would allow it.2

3 Weight loss and decreased appetite

A depressed patient may report no interest in food or may overeat. CFS patients are interested in food but find shopping and meal preparation fatiguing. Their nutritional intake may consist of fast food or easily prepared meals.

4 Insomnia

Depressed patients will report poor sleep or excessive sleep (insomnia or hypersomnia). CFS patients experience unrefreshing sleep regardless of how long they sleep.

5 Fatigue or loss of energy

Patients with major depressive disorder will report fatigue regardless of the task. They often view all tasks as equally difficult. CFS patients present with overwhelming fatigue. They express a desire to do more but are physically unable. These patients experience postexertional malaise typically worsening 12 to 48 hours after an activity and lasting for days to weeks.2 Therefore, CFS patients may avoid normal activity.3

6 Feelings of worthlessness or guilt

Guilt in major depressive disorder is often delusional and broad, extending to all areas of life. CFS patients will report guilt caused by their inability to be more active. They may feel they are letting their family or co-workers down. Some people—including some health care providers—do not regard CFS as a “real disease;” others may criticize these patients’ work and daily activity level, therefore increasing guilt.

7 Diminished ability to think or concentrate

In depressed patients, this symptom should improve with antidepressant therapy. In patients with CFS, antidepressants often do not improve concentration or memory.

8 Thoughts of death and suicide

Depressed patients often will have suicidal ideation and believe life is hopeless. CFS patients can become depressed and suicidal because of the condition’s prolonged debilitating symptoms. They may qualify suicidal thoughts with, “If I have to live like this for the rest of my life, I’d rather be dead.”

9 Medical complaints

Depressed and CFS patients often present with medical complaints and require a medical workup. Chronic symptoms— such as muscle pain, headache, multijoint pain without swelling or redness, sore throat, and tender lymph nodes—constitute some of the core symptom criteria of CFS.3 These symptoms may overlap with depressed patients’ somatic complaints.

History of symptom onset and complete medical workups are important to make an accurate diagnosis. Rule out other medical disorders such as fibromyalgia, chronic mononucleosis, hypothyroidism, and subacute infections in patients who present with prolonged fatigue.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed, text revision 2000: Washington, DC: American Psychiatric Association; 349-56.

2. LaFerney M. Diagnosing depression: clinical depression can be difficult to differentiate from other illnesses and medication side effects. Advance for Nurses 2007;7(8):33.-

3. Recognition and management of chronic fatigue syndrome: resource guide for health care professionals. Atlanta, GA: Centers for Dis-ease Control and Prevention; 2006.

Article PDF
Author and Disclosure Information

Michael C. LaFerney, APRN, BC
Mr. LaFerney is psychiatric clinical nurse specialist, Arbour SeniorCare, Rockland, MA.

Issue
Current Psychiatry - 07(01)
Publications
Page Number
91-94
Legacy Keywords
Chronic fatigue syndrome; major depressive disorder; Michael C. LaFerney
Sections
Author and Disclosure Information

Michael C. LaFerney, APRN, BC
Mr. LaFerney is psychiatric clinical nurse specialist, Arbour SeniorCare, Rockland, MA.

Author and Disclosure Information

Michael C. LaFerney, APRN, BC
Mr. LaFerney is psychiatric clinical nurse specialist, Arbour SeniorCare, Rockland, MA.

Article PDF
Article PDF

Chronic fatigue syndrome (CFS) is characterized by prolonged, debilitating fatigue that does not improve with rest and may be worsened by physical and mental activity. This fatigue must be present for at least 6 months. This syndrome affects more than 1 million individuals in the U.S. and shares symptoms with many medical and psychiatric illnesses, particularly depression.

Keeping in mind that a patient who presents with fatigue could have depression and CFS, follow DSM-IV-TR criteria for major depressive disorder to help identify clinical factors that distinguish the 2 disorders.1

1 Depressed mood

Individuals with major depressive disorder report being depressed most of time and often cannot provide a reason or identify a loss to explain their depressed mood. CFS patients are more likely to report feeling discouraged and depressed because a lack of energy leaves them unable to complete tasks.

2 Diminished interest in activities

Depressed patients typically report a loss or lack of interest in pleasurable activities. CFS patients say they would engage in favorite activities more if their energy level would allow it.2

3 Weight loss and decreased appetite

A depressed patient may report no interest in food or may overeat. CFS patients are interested in food but find shopping and meal preparation fatiguing. Their nutritional intake may consist of fast food or easily prepared meals.

4 Insomnia

Depressed patients will report poor sleep or excessive sleep (insomnia or hypersomnia). CFS patients experience unrefreshing sleep regardless of how long they sleep.

5 Fatigue or loss of energy

Patients with major depressive disorder will report fatigue regardless of the task. They often view all tasks as equally difficult. CFS patients present with overwhelming fatigue. They express a desire to do more but are physically unable. These patients experience postexertional malaise typically worsening 12 to 48 hours after an activity and lasting for days to weeks.2 Therefore, CFS patients may avoid normal activity.3

6 Feelings of worthlessness or guilt

Guilt in major depressive disorder is often delusional and broad, extending to all areas of life. CFS patients will report guilt caused by their inability to be more active. They may feel they are letting their family or co-workers down. Some people—including some health care providers—do not regard CFS as a “real disease;” others may criticize these patients’ work and daily activity level, therefore increasing guilt.

7 Diminished ability to think or concentrate

In depressed patients, this symptom should improve with antidepressant therapy. In patients with CFS, antidepressants often do not improve concentration or memory.

8 Thoughts of death and suicide

Depressed patients often will have suicidal ideation and believe life is hopeless. CFS patients can become depressed and suicidal because of the condition’s prolonged debilitating symptoms. They may qualify suicidal thoughts with, “If I have to live like this for the rest of my life, I’d rather be dead.”

9 Medical complaints

Depressed and CFS patients often present with medical complaints and require a medical workup. Chronic symptoms— such as muscle pain, headache, multijoint pain without swelling or redness, sore throat, and tender lymph nodes—constitute some of the core symptom criteria of CFS.3 These symptoms may overlap with depressed patients’ somatic complaints.

History of symptom onset and complete medical workups are important to make an accurate diagnosis. Rule out other medical disorders such as fibromyalgia, chronic mononucleosis, hypothyroidism, and subacute infections in patients who present with prolonged fatigue.

Chronic fatigue syndrome (CFS) is characterized by prolonged, debilitating fatigue that does not improve with rest and may be worsened by physical and mental activity. This fatigue must be present for at least 6 months. This syndrome affects more than 1 million individuals in the U.S. and shares symptoms with many medical and psychiatric illnesses, particularly depression.

Keeping in mind that a patient who presents with fatigue could have depression and CFS, follow DSM-IV-TR criteria for major depressive disorder to help identify clinical factors that distinguish the 2 disorders.1

1 Depressed mood

Individuals with major depressive disorder report being depressed most of time and often cannot provide a reason or identify a loss to explain their depressed mood. CFS patients are more likely to report feeling discouraged and depressed because a lack of energy leaves them unable to complete tasks.

2 Diminished interest in activities

Depressed patients typically report a loss or lack of interest in pleasurable activities. CFS patients say they would engage in favorite activities more if their energy level would allow it.2

3 Weight loss and decreased appetite

A depressed patient may report no interest in food or may overeat. CFS patients are interested in food but find shopping and meal preparation fatiguing. Their nutritional intake may consist of fast food or easily prepared meals.

4 Insomnia

Depressed patients will report poor sleep or excessive sleep (insomnia or hypersomnia). CFS patients experience unrefreshing sleep regardless of how long they sleep.

5 Fatigue or loss of energy

Patients with major depressive disorder will report fatigue regardless of the task. They often view all tasks as equally difficult. CFS patients present with overwhelming fatigue. They express a desire to do more but are physically unable. These patients experience postexertional malaise typically worsening 12 to 48 hours after an activity and lasting for days to weeks.2 Therefore, CFS patients may avoid normal activity.3

6 Feelings of worthlessness or guilt

Guilt in major depressive disorder is often delusional and broad, extending to all areas of life. CFS patients will report guilt caused by their inability to be more active. They may feel they are letting their family or co-workers down. Some people—including some health care providers—do not regard CFS as a “real disease;” others may criticize these patients’ work and daily activity level, therefore increasing guilt.

7 Diminished ability to think or concentrate

In depressed patients, this symptom should improve with antidepressant therapy. In patients with CFS, antidepressants often do not improve concentration or memory.

8 Thoughts of death and suicide

Depressed patients often will have suicidal ideation and believe life is hopeless. CFS patients can become depressed and suicidal because of the condition’s prolonged debilitating symptoms. They may qualify suicidal thoughts with, “If I have to live like this for the rest of my life, I’d rather be dead.”

9 Medical complaints

Depressed and CFS patients often present with medical complaints and require a medical workup. Chronic symptoms— such as muscle pain, headache, multijoint pain without swelling or redness, sore throat, and tender lymph nodes—constitute some of the core symptom criteria of CFS.3 These symptoms may overlap with depressed patients’ somatic complaints.

History of symptom onset and complete medical workups are important to make an accurate diagnosis. Rule out other medical disorders such as fibromyalgia, chronic mononucleosis, hypothyroidism, and subacute infections in patients who present with prolonged fatigue.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed, text revision 2000: Washington, DC: American Psychiatric Association; 349-56.

2. LaFerney M. Diagnosing depression: clinical depression can be difficult to differentiate from other illnesses and medication side effects. Advance for Nurses 2007;7(8):33.-

3. Recognition and management of chronic fatigue syndrome: resource guide for health care professionals. Atlanta, GA: Centers for Dis-ease Control and Prevention; 2006.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed, text revision 2000: Washington, DC: American Psychiatric Association; 349-56.

2. LaFerney M. Diagnosing depression: clinical depression can be difficult to differentiate from other illnesses and medication side effects. Advance for Nurses 2007;7(8):33.-

3. Recognition and management of chronic fatigue syndrome: resource guide for health care professionals. Atlanta, GA: Centers for Dis-ease Control and Prevention; 2006.

Issue
Current Psychiatry - 07(01)
Issue
Current Psychiatry - 07(01)
Page Number
91-94
Page Number
91-94
Publications
Publications
Article Type
Display Headline
Depression or chronic fatigue syndrome?
Display Headline
Depression or chronic fatigue syndrome?
Legacy Keywords
Chronic fatigue syndrome; major depressive disorder; Michael C. LaFerney
Legacy Keywords
Chronic fatigue syndrome; major depressive disorder; Michael C. LaFerney
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media