Pharmacological treatment of neuropsychiatric symptoms of dementia. A review of the evidence
Context: Neuropsychiatric symptoms of dementia are common and associated with poor outcomes for patients and caregivers. Although nonpharmacological interventions should be the first line of treatment, a wide variety of pharmacological agents are used in the management of neuropsychiatric symptoms; therefore, concise, current, evidence-based recommendations are needed.

Objective: To evaluate the efficacy of pharmacological agents used in the treatment of neuropsychiatric symptoms of dementia.

Evidence Acquisition: A systematic review of English-language articles published from 1966 to July 2004 using MEDLINE, the Cochrane Database of Systematic Reviews, and a manual search of bibliographies was conducted. Inclusion criteria were double-blind, placebo-controlled, randomized controlled trials (RCTs) or meta-analyses of any drug therapy for patients with dementia that included neuropsychiatric outcomes. Trials reporting only depression outcomes were excluded. Data on the inclusion criteria, patients, methods, results, and quality of each study were independently abstracted. Twenty-nine articles met inclusion criteria.

Evidence Synthesis: For typical antipsychotics, 2 meta-analyses and 2 RCTs were included. Generally, no difference among specific agents was found, efficacy was small at best, and adverse effects were common. Six RCTs with atypical antipsychotics were included; results showed modest, statistically significant efficacy of olanzapine and risperidone, with minimal adverse effects at lower doses. Atypical antipsychotics are associated with an increased risk of stroke. There have been no RCTs designed to directly compare the efficacy of typical and atypical antipsychotics. Five trials of antidepressants were included; results showed no efficacy for treating neuropsychiatric symptoms other than depression, with the exception of 1 study of citalopram. For mood stabilizers, 3 RCTs investigating valproate showed no efficacy. Two small RCTs of carbamazepine had conflicting results. Two meta-analyses and 6 RCTs of cholinesterase inhibitors generally showed small, although statistically significant, efficacy. Two RCTs of memantine also had conflicting results for treatment of neuropsychiatric symptoms.

Conclusions: Pharmacological therapies are not particularly effective for management of neuropsychiatric symptoms of dementia. Of the agents reviewed, the atypical antipsychotics risperidone and olanzapine currently have the best evidence for efficacy. However, the effects are modest and further complicated by an increased risk of stroke. Additional trials of cholinesterase inhibitors enrolling patients with high levels of neuropsychiatric symptoms may be warranted.


Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neuropsychiatric symptoms of dementia. A review of the evidence. JAMA 2005;293:596-608.


INTRODUCTION

Up to 50% of community-dwelling elderly individuals older than 85 years have dementia, with Alzheimer disease (AD), vascular dementia, and dementia with Lewy bodies accounting for most cases.1-2 Although cognitive deficits are the clinical hallmark of dementing illnesses, noncognitive symptoms are common and can dominate disease presentation. These include an array of neuropsychiatric symptoms, such as agitation, aggression, delusions, hallucinations, repetitive vocalizations, and wandering, among other symptoms. Neuropsychiatric symptoms have been observed in 60% to 98% of patients with dementia,3-7 especially in later stages, and are associated with caregiver stress and depression, as well as reduced caregiver employment and income.8-13

Neuropsychiatric symptoms are also associated with increased hospital lengths of stay14 and commonly lead to nursing home placement.15-17 Federal expenditures for dementia are expected to triple in the next 10 years18 and 30% of the cost of caring for patients with AD is attributed directly to the management of neuropsychiatric symptoms.19 Thus, interventions aimed at treating neuropsychiatric symptoms could have a tremendous impact on patients, caregivers, and society.

Although there are multiple classes of drugs in use for neuropsychiatric symptoms, including antipsychotics, anticonvulsants, antidepressants, anxiolytics, cholinesterase inhibitors, and N-methyl-D-aspartate–receptor modulators, there is no clear standard of care and treatment is often based on local pharmacotherapy customs. We provide a comprehensive systematic review of pharmacological interventions for neuropsychiatric symptoms of dementia. Our goal is to provide the generalist physician with a clinically useful, evidence-based assessment of available pharmacological interventions for neuropsychiatric symptoms.


EVIDENCE ACQUISITION

To identify articles, we systematically searched the MEDLINE database for English-language articles published between 1966 and July 2004, the Cochrane Database of Systematic Reviews, and performed a manual search of the reference lists of relevant retrieved articles. In MEDLINE, we combined the results of searches in 3 separate domains: dementia (MeSH terms dementia; Alzheimer disease; dementia, vascular; or Lewy body disease), neuropsychiatric symptoms (behavior; neurobehavioral manifestations; perceptual disorders; psychomotor disorders; mood disorders; or keyword neuropsychiatric), and drug therapy (cholinesterase inhibitors; tranquilizing agents; serotonin uptake inhibitors; anticonvulsants; valproic acid; benzodiazepines; trazodone; memantine; or psychotropic drugs). A total of 253 articles were identified through database searches. The titles and abstracts were read by 2 authors (K.M.S. and K.F.H.) and if the article appeared to meet inclusion criteria or if we were uncertain, the full study was obtained. A total of 187 articles were excluded based on reviewing titles and abstracts; therefore, 66 articles were obtained for full review and an additional 12 articles were identified in the manual search of references.

Studies were selected for inclusion in our systematic review if they met all of the following inclusion criteria: double-blind, placebo-controlled, randomized controlled trials (RCTs) or meta-analyses of RCTs; intervention consisting of any drug therapy for patients with dementia (generally defined in accordance with Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) criteria and including AD, vascular dementia, mixed, or dementia with Lewy bodies); and outcomes for neuropsychiatric symptoms were reported (eg, hallucinations, delusions, combativeness, verbal aggression, psychomotor agitation, wandering). Trials reporting only depression outcomes were excluded. For the sake of summarizing a large body of evidence succinctly, if a meta-analysis was available, results from that analysis were presented along with any RCTs published since the meta-analysis. Because the goal of this review was to be clinically useful, studies were excluded if the drug was not available for use in the United States or was no longer in wide clinical use (eg, tacrine). Studies were also excluded if they were post hoc analyses of trials already selected for inclusion or were duplicate publications. Data on the inclusion criteria, patients, methods, results, and quality of each study were independently abstracted by 2 authors (K.M.S. and K.F.H.). Disagreements were discussed and if consensus was not reached, a third author (K.Y.) was the final arbitrator. From 78 articles that were reviewed, only 25 RCTs and 4 meta-analyses met our inclusion criteria.


EVIDENCE SYNTHESIS

Typical Antipsychotics

Two meta-analyses20-21 covering 12 RCTs and 2 additional RCTs22-23 of typical (or conventional) antipsychotics were reviewed (Table 1 and Table 2). Trials varied in length from 17 days to 16 weeks. In an early meta-analysis21 of antipsychotic drugs (including haloperidol, thioridazine, thiothixene, chlorpromazine, trifluoperazine, and acetophenazine) covering 7 RCTs, the authors concluded that "18 of 100 patients benefited from neuroleptics (beyond that of placebo)." There was no difference in efficacy among the different typical antipsychotics on neuropsychiatric symptoms. An RCT of thioridazine vs placebo23 found significant improvements in agitation, but the trial was of poor quality with no mention of effect size, methods for randomization and allocation concealment, or the number of patients who completed the trial. A recent Cochrane review20 comparing haloperidol with placebo for the treatment of agitation in dementia concluded that aggression, but not agitation, behavioral symptoms as a whole, or clinical global impression of change was improved by treatment with haloperidol. Whether the statistically significant result for the aggression subscale represents a true benefit on aggression vs a chance finding in the setting of multiple hypothesis testing is unclear. In addition, the authors reported that dropouts due to adverse events, such as extrapyramidal symptoms and somnolence, were more than twice as likely to occur among those individuals randomized to haloperidol than placebo (odds ratio [OR], 2.5; 95% confidence interval [CI], 1.2-5.2). Lastly, perphenazine was not found to be of benefit compared with placebo in a 17-day trial of hospitalized patients.22


________
Table 1. Studies of Typical and Atypical Antipsychotics: Study Characteristics
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T1
________


________
Table 2. Studies of Typical and Atypical Antipsychotics: Outcomes
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T2
________


There is no clear evidence that typical antipsychotic drugs are useful for treating neuropsychiatric symptoms defined broadly. There may be a slight benefit for haloperidol with aggression (doses of 1.2-3.5 mg/d) but it is unclear if this benefit outweighs the adverse effects, particularly extrapyramidal symptoms and sedation. There is no evidence that any one typical antipsychotic is more efficacious than another.


Atypical Antipsychotics

Atypical antipsychotics, also known as second-generation antipsychotics, include clozapine, olanzapine, risperidone, quetiapine, ziprasidone, and aripiprazole. Four of 6 RCTs of atypical antipsychotics (olanzapine and risperidone) reported benefit in the treatment of neuropsychiatric symptoms of dementia (Table 1 and Table 2). 24-29 These trials ranged from 24 hours to 12 weeks and were all conducted among nursing home residents, generally with moderate to severe dementia (mean Mini-Mental State Examination score, 5.5-13.7). The first trial of oral olanzapine reported that doses of 5 and 10 mg but not 15 mg were associated with a statistically significant decrease in the primary outcome of the sum of 3 Neuropsychiatric Inventory (NPI) core symptoms (agitation/aggression, hallucinations, and delusions).26 Only the 5-mg dose was associated with improvement in total NPI score (mean, 8.8 point improvement over placebo; range, 0-144; P = .005). Another trial comparing varying doses of olanzapine with placebo for patients with dementia-related psychosis found no significant difference between any of the doses and placebo in either of the primary outcomes.29 However, the authors show that the 7.5-mg dose was better than placebo in some secondary analyses. Finally, results from a 24-hour trial of intramuscular olanzapine (2.5 or 5.0 mg) reported a statistically significant improvement in the primary outcome at 2 hours in those individuals treated with either dose of olanzapine compared with placebo.27

In 1 fixed-dose trial of risperidone,25 45% of patients receiving 1.0 mg and 50% patients receiving 2.0 mg compared with 33% receiving placebo had at least 50% reduction in the Behavioral Pathology in Alzheimer Disease Rating Scale (BEHAVE-AD) score (P = .02 and P=.002 vs placebo, respectively). A 2.0-mg dose was not more efficacious than 1.0-mg dose and resulted in significantly higher adverse events, including extrapyramidal symptoms and somnolence. A subgroup analysis of patients who were not somnolent found similar benefit for risperidone vs placebo, suggesting that somnolence was not the mechanism for efficacy. Another trial24 of 229 patients did not report a significant benefit of risperidone (mean dose, 1.1 mg/d) in their primary outcome (≥30% reduction in BEHAVE-AD total score) but did report significant results for several secondary analyses. This trial found no difference between risperidone and haloperidol in the primary outcome, although extrapyramidal symptoms were more common with haloperidol. In the most recent trial of risperidone,28 mean doses of 0.95 mg/d were found to be more efficacious than placebo on the primary outcome (4.4-point difference on Cohen-Mansfield Agitation Inventory aggression score [scale of 84 points], P<.001) and several secondary outcomes. In this trial, 16.8% of the patients randomized to risperidone (vs 8.8% of placebo group) had serious adverse events, including 5 strokes and 1 transient ischemic attack. All of the cerebrovascular events reported in the trial occurred with risperidone. However, all of these patients also had stroke risk factors (5 of 6 patients had atrial fibrillation).

Doses of 5 to 10 mg/d of olanzapine or 1.0 mg/d of risperidone appear to be at least modestly effective for treating neuropsychiatric symptoms of dementia in patients with AD or vascular dementia. The incidence of extrapyramidal symptoms appears to be low when receiving these doses of olanzapine and risperidone, but somnolence remains a concern. To our knowledge, there have been no published RCTs of clozapine, quetiapine, ziprasidone, or aripiprazole for neuropsychiatric symptoms of dementia. Furthermore, there have been no published RCTs designed to compare the efficacy of typical and atypical agents.


Antidepressants

Of the 5 RCTs that have investigated the use of serotonergic antidepressants for the treatment of neuropsychiatric symptoms (sertraline, fluoxetine, citalopram, and trazodone),22, 30-33 only the trial of citalopram found benefit (Table 3 and Table 4). This 17-day study of hospitalized patients found a 10-point change (of 168 points) in the Neurobehavioral Rating Scale for patients randomized to citalopram compared with a 2.3-point change for placebo (P<.001).22 Of the 7 subscales examined, only agitation and lability were significantly improved with citalopram compared with placebo. The trial had a high dropout rate, with more than half of patients in each group failing to complete the study, most commonly due to lack of efficacy. Lyketsos et al31 found sertraline to be effective in the treatment of depression among patients with dementia. However, there was no significant benefit of sertraline on neuropsychiatric symptoms. The authors did report that in subgroup analyses of full responders vs nonresponders (in terms of depression symptoms), full responders had significantly greater improvement on nonmood items of the NPI than nonresponders.


________
Table 3. Studies of Antidepressants and Mood Stabilizers: Study Characteristics*
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T3
________


________
Table 4. Studies of Antidepressants and Mood Stabilizers: Outcomes
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T4
________


We conclude from these trials that although serotonergic agents are well tolerated, they do not appear to be very effective in the treatment of neuropsychiatric symptoms of dementia other than depression.39-40


Mood Stabilizers

Three RCTs have investigated valproate36-38 and 2 studies have investigated carbamazepine for neuropsychiatric symptoms34-35 (Table 3 and Table 4). Valproate does not appear to be effective for the treatment of neuropsychiatric symptoms of dementia whether in short- or long-acting preparations. In addition, valproate caused significantly more adverse events than placebo, sedation being the most common. Therefore, we do not recommend the use of valproate in the management of neuropsychiatric symptoms of dementia. Based on 2 small trials (1 positive34 and 1 negative35), there is currently not enough evidence of benefit to recommend the use of carbamazepine for treatment of neuropsychiatric symptoms, especially in light of the black box warning for hematologic toxicity and the potential drug-drug interactions between carbamazepine and other drugs commonly prescribed to elderly individuals. To our knowledge, there have been no published placebo-controlled RCTs of lithium for the treatment of neuropsychiatric symptoms of dementia.


Cholinesterase Inhibitors

Two meta-analyses41-42 and 6 additional RCTs43-48 of various cholinesterase inhibitors with neuropsychiatric symptom outcomes have been published (Table 5 and Table 6) with 5 of the 8 studies reporting statistically significant benefit. In a recent meta-analysis of cholinesterase inhibitors,42 the authors reported a small but statistically significant benefit from cholinesterase inhibitors with NPI scores (summary estimate 1.72-point improvement vs placebo on a scale of 0-120) but not Alzheimer Disease Assessment Scale, noncognitive portion scores. The statistically significant effect on NPI scores was most likely driven by 2 studies of metrifonate, which was never approved by the Food and Drug Administration for use in the United States due to toxicities.


________
Table 5. Studies of Cholinesterase Inhibitors and Memantine: Study Characteristics
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T5
________



________
Table 6. Studies of Cholinesterase Inhibitors and Memantine: Outcomes
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066T6
________


A Cochrane review of galantamine for AD reported 2 RCTs of galantamine that included the NPI as an outcome.41 In one trial, there was no benefit of either galantamine 24-mg/d or 32-mg/d dose vs placebo.51 In the second trial, the intention-to-treat analysis found only the 16-mg/d dose to be significantly better than placebo (mean difference of 2.1 points, P = .03) with no benefit for the other doses.52 One additional RCT using 24-mg/d dose of galantamine also reported a mean treatment difference in NPI scores of 2.2 points (P<.05).43 However, a 2-point difference on the NPI (range, 0-120) is unlikely to be clinically significant.

Four additional RCTs using donepezil have reported conflicting results. In a 24-week trial of 208 patients in a nursing home, there was no difference between treatment groups in the mean change in NPI scores.46 In subgroup analyses using the NPI items as categorical outcomes, only agitation/aggression was better for those individuals randomized to donepezil vs placebo (45% improved vs 28%, P = .04). A 24-week trial of patients living in the community or assisted-living facilities found a slightly larger treatment difference of 5.6 points on the NPI in favor of donepezil (P < .001), with statistically significant benefit on the apathy, depression, and anxiety subscales.44 A similar magnitude of benefit was reported in a 12-week trial of 96 patients who had significant neuropsychiatric symptoms on enrollment.48 However, in the longest trial of cholinesterase inhibitors to date, there was no significant difference in the mean changes in NPI scores between those individuals randomized to donepezil (5 or 10 mg) or placebo at any time point up to 4 years of follow-up.47

An RCT of patients with mild to moderate dementia with Lewy bodies found no difference in NPI scores between rivastigmine and placebo on either a 4-item NPI "dementia with Lewy bodies cluster" (delusions, hallucinations, apathy, and depression) or the full 10-item NPI.45 However, using a predefined cutoff of at least 30% improvement, significantly more patients receiving rivastigmine showed improvement compared with placebo (47.5% vs 27.9%, P = .03).

Although some trials of cholinesterase inhibitors have shown statistically significant differences, the magnitude of effect has been small and of questionable clinical significance. Most of the patients enrolled in the cholinesterase inhibitor trials had little neuropsychiatric symptoms with only 2 trials,46, 48 requiring significant neuropsychiatric symptoms as part of the entry criteria.


Other Drugs

Memantine, an N-methyl-D-aspartate receptor antagonist, was recently approved in the United States for the treatment of moderate to severe AD. Two RCTs of community-dwelling patients with moderate to severe AD have included neuropsychiatric symptoms as secondary outcomes (Table 5 and Table 6).49-50 In 1 trial,49 NPI scores were not significantly different between groups. In the other trial,50 there was a statistically significant difference in NPI change scores, largely because those individuals randomized to placebo got worse. Patients receiving memantine improved their NPI score by an average of 0.1 points, whereas patients receiving placebo declined 3.7 points (P = .002). This difference is of unclear clinical significance. We conclude that although memantine may be of benefit in cognitive and functional domains, there does not appear to be a clinically significant benefit in the treatment of neuropsychiatric symptoms for patients with moderate to severe AD.

Only 1 placebo-controlled RCT has been published on the use of benzodiazepines for the management of neuropsychiatric symptoms of dementia.27 This 24-hour trial of intramuscular olanzapine (2.5 mg or 5 mg) vs intramuscular lorazepam (1 mg) vs placebo is described in the section on atypical antipsychotics in Table 2. To our knowledge, there have been no published placebo-controlled RCTs on buspirone for the management of neuropsychiatric symptoms of dementia.


CONTROVERSIES

Interpretation of Data

There are several important methodological issues in neuropsychiatric symptom trials that limit the interpretation of the data and generate controversies on pharmacological management of neuropsychiatric symptoms of dementia. One controversy is how to define clinically significant improvement in neuropsychiatric symptoms. A particularly good example of this problem is highlighted with the cholinesterase inhibitor trials in which several trials reported very small but statistically significant changes in the NPI scores. Because there are no gold standard outcomes for neuropsychiatric symptoms, it is difficult to interpret small changes in scale scores and also to compare results across trials using different scales to measure neuropsychiatric symptoms (Box). To address this, we attempted to calculate standardized response means,53 but the vast majority of trials did not present the data needed to calculate such a measure. Clinically useful outcomes such as nursing home placement, quality of life, and caregiver burden and depression would enhance a clinician’s ability to interpret trial results and counsel patients and families regarding risks and benefits of treatment.


________
Box. Neuropsychiatric Symptom Rating Scales

- Agitated Behavior Inventory for Dementia (ABID)
- Agitation-Calmness Evaluation Scale (ACES)
- Alzheimer Disease Assessment Scale, noncognitive portion (ADAS-noncog)
- Bech-Rafaelsen Mania Scale (BRMS)
- Behavior Observation Scale for Intramural Psychogeriatric Patients (GIP)
- Behavior Rating Scale for Dementia (by the Consortium to Establish a Registry for Dementia) (BRSD)
- Behavioral Pathology in Alzheimer Disease Rating Scale (BEHAVE-AD)
- Brief Psychiatric Rating Scale (BPRS)
- Caregiver Burden Questionnaire (CBQ)
- Clinical Global Impression of Change (1 = very much improved to 7 = very much worse) (CGIC)
- Clinical Global Impression Scale (CGIS)
- Clinicians Interview Based Impression of Change plus caregiver input (CIBIC-plus)
- Cohen-Mansfield Agitation Inventory (CMAI)
- Hamilton Rating Scale for Depression (Ham-D)
- Iowa Caregiver Stress Inventory (CSI)
- Neurobehavioral Rating Scale (0 = not present to 7 = extremely severe), derived from the Brief Psychiatric Rating Scale (NRS)
- Neuropsychiatric Inventory (usually 10 items, 120 points) (NPI)
- Neuropsychiatric Inventory-Nursing Home version (12 items, 144 points) (NPI-NH)
- Neuropsychiatric Inventory minus 5 "mood" items (NPI-NM)
- Overt Aggression Scale (OAS)
- Positive and Negative Syndrome Scale-Excited Component (PANSS-EC)
- Revised Memory and Behavior Problem Checklist (RMBPC)
- Screen for Caregiver Burden (SCB)
- Social Dysfunction and Aggression Scale (SDAS-9)
________



Another problem with the current literature is that most trials report on multiple outcomes from several different scales and subscales. What does it mean if the score was significantly improved on 1 scale but not on 4 others? This multiple comparison testing raises the concern for type I error. In addition, when reporting their results, many of the studies downplay the negative primary outcome while emphasizing positive secondary outcomes, especially in the abstract and discussion sections. The majority of these trials have been funded by the pharmaceutical industry. In this review, we have focused on the primary outcome as specified by the authors.


Clinical Dilemmas

The current evidence appears to suggest that, if behavioral interventions have failed, neuropsychiatric symptoms of dementia are best treated with the atypical antipsychotic agents (risperidone and olanzapine). However, the product label warning for cerebrovascular events (strokes and transient ischemic attacks) for these drugs creates a clinical dilemma. The pooled incidence of cerebrovascular events across 6 RCTs of risperidone in the patients with dementia (N = 1721) was reported to be 3.3% for risperidone vs 1.1% for placebo (P = .03).54 Similarly, combining data from 5 RCTs of olanzapine in 1656 patients with dementia-related psychosis revealed the incidence of cerebrovascular events in patients treated with olanzapine was significantly higher than in the placebo group (1.3% vs 0.4%, P = .02), even after adjustment for age, sex, and type of dementia.55 Therefore, physicians considering the prescription of risperidone or olanzapine should discuss the potential risks and benefits of such treatment with patients and their surrogate decision makers, especially for patients with risk factors for cerebrovascular disease.

Another area of clinical uncertainty pertains to the treatment of neuropsychiatric symptoms in patients with dementia with Lewy bodies, increasingly recognized as a common form of dementia.2, 56-57 Very few trials regarding the treatment of neuropsychiatric symptoms have included patients with dementia with Lewy bodies. Therefore, no conclusions can be drawn regarding the efficacy of drug treatment for neuropsychiatric symptoms occurring in this setting. However, antipsychotics should be used cautiously in patients suspected to have dementia with Lewy bodies as these patients have been reported to have marked sensitivity, including life-threatening neuroleptic malignant syndrome, to typical and atypical antipsychotics.57-61 Although dementia with Lewy bodies is characterized by fluctuating levels of impairment, hallucinations (often visual), and parkinsonism, the diagnosis may be overlooked, especially in later stages of the illness, until the patient experiences significant extrapyramidal symptoms while receiving an antipsychotic. If this occurs, the drug should be discontinued and a diagnosis of dementia with Lewy bodies entertained.


CONCLUSIONS AND PERSPECTIVES

Among the many drugs in use for the treatment of neuropsychiatric symptoms, we found only the atypical antipsychotics, risperidone and olanzapine, to have convincing evidence of efficacy for neuropsychiatric symptoms. In addition, trials of cholinesterase inhibitors have had remarkably consistent, albeit small positive effects on neuropsychiatric symptoms. However, it is clear that none of the drugs in use for neuropsychiatric symptoms offer a "magic pill." The effect sizes have been modest at best, with treatment differences on the NPI scale of about 2 points for cholinesterase inhibitors and up to 8 points for olanzapine.


Potential Treatment Strategies

The management of neuropsychiatric symptoms in dementia should always begin with an assessment of the patient for medical (eg, pain and delirium) and environmental causes of the behavior. If the problem persists after these have been addressed, nonpharmacological interventions should be attempted before moving to drug therapy. Although a comprehensive review of nonpharmacological interventions is outside the scope of this article, several interventions have been shown in small studies to have varying degrees of success including but not limited to music therapy,62-63 aromatherapy,64-65 and pet therapy.66 Caregiver (either formal or informal) education is also an integral part of the management.67 Larger, well-designed, controlled trials of nonpharmacological interventions are needed.

Until there are better answers, if drug therapy is to be instituted, there are 2 reasonable approaches to the management of neuropsychiatric symptoms, each with its merits and pitfalls. One approach would be to identify the target symptom and choose a drug that is known to treat a symptom most closely related to the one the patient is exhibiting. For example, one might use an antipsychotic for psychotic symptoms or an antidepressant for anxiety symptoms, such as repetitive vocalizations or pacing. Although this approach is intuitive, RCTs have not been designed to confirm that this approach is effective and secondary analyses suggest it might not be.68

An alternative approach is one guided by the current state of evidence in combination with the goal of minimizing adverse effects (Figure). For example, although the evidence for cholinesterase inhibitors as effective treatments for neuropsychiatric symptoms is not as convincing as that for risperidone or olanzapine, we recommend beginning with a cholinesterase inhibitor if the patient is not already receiving one because they are well tolerated and may benefit cognition and function, even if they are not beneficial for neuropsychiatric symptoms.67 Additionally, typical antipsychotics do not appear in the suggested algorithm because there is less evidence of benefit and more adverse effects compared with the atypical antipsychotics.


________
Figure. Recommended Algorithm for Management of Neuropsychiatric Symptoms of Dementia
http://jama.ama-assn.org/cgi/content-nw/full/293/5/596/JCR40066F1
________


Benzodiazepines are not part of the recommended management of neuropsychiatric symptoms and should be avoided, especially for long-term management. Although the only RCT using a benzodiazepine for acutely agitated patients with dementia (Table 2)27 did not report a significant difference in adverse events in the 24 hours after intramuscular injection, case reports and anecdotal evidence suggest that benzodiazepines lead to increased confusion, falls, and may paradoxically increase agitation in patients with dementia.69-70 Consistent with this information, the report published by the Expert Consensus Panel for Agitation in Dementia generally recommended against the use of benzodiazepines except for short-term or occasional use for anxiety symptoms.71 In addition, no psychoactive medication prescribed to treat neuropsychiatric symptoms of dementia should be continued indefinitely and attempts at drug withdrawal should be made regularly.72 Many patients who are prescribed antipsychotics for neuropsychiatric symptoms will no longer need them when the drug is later discontinued.73 Physical restraints should be avoided as they are associated with injury, not protection, of patients who are confused.74


Directions for Future Research

Because there is no "magic pill" for neuropsychiatric symptoms of dementia, it is especially important to continue efforts to better understand the pathophysiology of the symptoms and whether they vary by dementia type; perform high-quality trials of nonpharmacological treatments, especially in combination with drug therapy; include nursing home placement and caregiver outcomes in trials47 ; and support non–industry-funded trials aimed specifically at treating patients with neuropsychiatric symptoms. The results from the National Institute of Mental Health–funded Clinical Antipsychotic Trials of Intervention Effectiveness trial75 will be particularly valuable. This trial is a multicenter RCT comparing risperidone, olanzapine, quetiapine, citalopram, and placebo for up to 36 weeks for the treatment of psychosis and agitation in patients with AD.75 This will be the first and longest placebo-controlled, head-to-head trial of atypical antipsychotic drugs.



AUTHOR INFORMATION

Corresponding Author: Kaycee M. Sink, MD, Sticht Center on Aging, Wake Forest University School of Medicine, One Medical Center Boulevard, Winston- Salem, NC 27157 (kmsink@wfubmc.edu).

Author Contributions: Drs Sink, Holden, and Yaffe had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Study concept and design: Sink, Yaffe.

Acquisition of data: Sink, Holden.

Analysis and interpretation of data: Sink, Holden, Yaffe.

Drafting of the manuscript: Sink, Holden.

Critical revision of the manuscript for important intellectual content: Sink, Yaffe.

Obtained funding: Sink, Yaffe.

Administrative, technical, or material support: Yaffe.

Study supervision: Yaffe.

Financial Disclosure: Dr Yaffe has received grant support through University of California, San Francisco from Pfizer and Eli Lilly, and has served as a consultant for Novartis. Drs Sink and Holden reported no financial disclosures.

Funding/Support: This work was supported in part by a National Institute on Aging T32 training grant at the University of California, San Francisco, and the Roena B. Kulynych Research Center, Wake Forest University, Winston-Salem, NC (Dr Sink); and by the Paul Beeson Faculty Scholars in Aging Research and grant NIA-R01 AG021918-01 (Dr Yaffe).

Role of the Sponsors: The sponsors had no role in the design or conduct of the study, the data collection, interpretation of results, or preparation and approval of the manuscript.

Acknowledgment: We thank Jay Luxenberg, MD, for his thoughtful review of the manuscript.

Author Affiliations: Sticht Center on Aging, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (Dr Sink); Division of Geriatrics (Dr Sink), Department of Psychiatry (Dr Holden), and Departments of Psychiatry, Neurology, and Epidemiology (Dr Yaffe), University of California; and San Francisco Veterans Affairs Medical Center (Dr Yaffe), San Francisco.


REFERENCES

1. Small GW, Rabins PV, Barry PP, et al. Diagnosis and treatment of Alzheimer disease and related disorders: consensus statement of the American Association for Geriatric Psychiatry, the Alzheimer's Association, and the American Geriatrics Society. JAMA. 1997;278:1363-1371. FREE FULL TEXT

2. Rahkonen T, Eloniemi-Sulkava U, Rissanen S, Vatanen A, Viramo P, Sulkava R. Dementia with Lewy bodies according to the consensus criteria in a general population aged 75 years or older. J Neurol Neurosurg Psychiatry. 2003;74:720-724. FREE FULL TEXT

3. Lyketsos CG, Steinberg M, Tschanz JT, Norton MC, Steffens DC, Breitner JC. Mental and behavioral disturbances in dementia: findings from the Cache County Study on Memory in Aging. Am J Psychiatry. 2000;157:708-714. FREE FULL TEXT

4. Finkel SI. Behavioral and psychological symptoms of dementia: a current focus for clinicians, researchers, and caregivers. J Clin Psychiatry. 2001;62(suppl 21):3-6. PUBMED

5. Lyketsos CG, Lopez O, Jones B, Fitzpatrick AL, Breitner J, DeKosky S. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the Cardiovascular Health Study. JAMA. 2002;288:1475-1483. FREE FULL TEXT

6. Mega MS, Cummings JL, Fiorello T, Gornbein J. The spectrum of behavioral changes in Alzheimer's disease. Neurology. 1996;46:130-135. FREE FULL TEXT

7. Sink KM, Covinsky KE, Newcomer R, Yaffe K. Ethnic differences in the prevalence and pattern of dementia-related behaviors. J Am Geriatr Soc. 2004;52:1277-1283. FULL TEXT | ISI | PUBMED

8. Borson S, Raskind MA. Clinical features and pharmacologic treatment of behavioral symptoms of Alzheimer's disease. Neurology. 1997;48(5 suppl 6):S17-S24. ABSTRACT

9. Clyburn LD, Stones MJ, Hadjistavropoulos T, Tuokko H. Predicting caregiver burden and depression in Alzheimer's disease. J Gerontol B Psychol Sci Soc Sci. 2000;55:S2-S13. ABSTRACT

10. Coen RF, Swanwick GR, O'Boyle CA, Coakley D. Behaviour disturbance and other predictors of carer burden in Alzheimer's disease. Int J Geriatr Psychiatry. 1997;12:331-336. FULL TEXT | ISI | PUBMED

11. Covinsky KE, Eng C, Lui LY, et al. Reduced employment in caregivers of frail elders: impact of ethnicity, patient clinical characteristics, and caregiver characteristics. J Gerontol A Biol Sci Med Sci. 2001;56:M707-M713. FREE FULL TEXT

12. Donaldson C, Tarrier N, Burns A. Determinants of carer stress in Alzheimer's disease. Int J Geriatr Psychiatry. 1998;13:248-256. FULL TEXT | ISI | PUBMED

13. Nagaratnam N, Lewis-Jones M, Scott D, Palazzi L. Behavioral and psychiatric manifestations in dementia patients in a community: caregiver burden and outcome. Alzheimer Dis Assoc Disord. 1998;12:330-334. ISI | PUBMED

14. Wancata J, Windhaber J, Krautgartner M, Alexandrowicz R. The consequences of non-cognitive symptoms of dementia in medical hospital departments. Int J Psychiatry Med. 2003;33:257-271. FULL TEXT | ISI | PUBMED

15. Steele C, Rovner B, Chase GA, Folstein M. Psychiatric symptoms and nursing home placement of patients with Alzheimer's disease. Am J Psychiatry. 1990;147:1049-1051. FREE FULL TEXT

16. Stern Y, Tang MX, Albert MS, et al. Predicting time to nursing home care and death in individuals with Alzheimer disease. JAMA. 1997;277:806-812. FREE FULL TEXT

17. Yaffe K, Fox P, Newcomer R, et al. Patient and caregiver characteristics and nursing home placement in patients with dementia. JAMA. 2002;287:2090-2097. FREE FULL TEXT

18. Taylor DH Jr, Sloan FA, Doraiswamy PM. Marked increase in Alzheimer's disease identified in medicare claims records between 1991 and 1999. J Gerontol A Biol Sci Med Sci. 2004;59:762-766. FREE FULL TEXT

19. Beeri MS, Werner P, Davidson M, Noy S. The cost of behavioral and psychological symptoms of dementia (BPSD) in community dwelling Alzheimer's disease patients. Int J Geriatr Psychiatry. 2002;17:403-408. FULL TEXT | ISI | PUBMED

20. Lonergan E, Luxenberg J, Colford J. Haloperidol for agitation in dementia. Cochrane Database Syst Rev. 2002;2:CD002852. PUBMED

21. Schneider LS, Pollock VE, Lyness SA. A meta-analysis of controlled trials of neuroleptic treatment in dementia. J Am Geriatr Soc. 1990;38:553-563. ISI | PUBMED

22. Pollock BG, Mulsant BH, Rosen J, et al. Comparison of citalopram, perphenazine, and placebo for the acute treatment of psychosis and behavioral disturbances in hospitalized, demented patients. Am J Psychiatry. 2002;159:460-465. FREE FULL TEXT

23. Stotsky B. Multicenter study comparing thioridazine with diazepam and placebo in elderly, nonpsychotic patients with emotional and behavioral disorders. Clin Ther. 1984;6:546-559. ISI | PUBMED

24. De Deyn PP, Rabheru K, Rasmussen A, et al. A randomized trial of risperidone, placebo, and haloperidol for behavioral symptoms of dementia. Neurology. 1999;53:946-955. FREE FULL TEXT

25. Katz IR, Jeste DV, Mintzer JE, Clyde C, Napolitano J, Brecher M. Comparison of risperidone and placebo for psychosis and behavioral disturbances associated with dementia: a randomized, double-blind trial: Risperidone Study Group. J Clin Psychiatry. 1999;60:107-115. ISI | PUBMED

26. Street JS, Clark WS, Gannon KS, et al. Olanzapine treatment of psychotic and behavioral symptoms in patients with Alzheimer disease in nursing care facilities: a double-blind, randomized, placebo-controlled trial: the HGEU Study Group. Arch Gen Psychiatry. 2000;57:968-976. FREE FULL TEXT

27. Meehan KM, Wang H, David SR, et al. Comparison of rapidly acting intramuscular olanzapine, lorazepam, and placebo: a double-blind, randomized study in acutely agitated patients with dementia. Neuropsychopharmacology. 2002;26:494-504. FULL TEXT | ISI | PUBMED

28. Brodaty H, Ames D, Snowdon J, et al. A randomized placebo-controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementia. J Clin Psychiatry. 2003;64:134-143. ISI | PUBMED

29. De Deyn PP, Carrasco MM, Deberdt W, et al. Olanzapine versus placebo in the treatment of psychosis with or without associated behavioral disturbances in patients with Alzheimer's disease. Int J Geriatr Psychiatry. 2004;19:115-126. FULL TEXT | ISI | PUBMED

30. Finkel SI, Mintzer JE, Dysken M, Krishnan KR, Burt T, McRae T. A randomized, placebo-controlled study of the efficacy and safety of sertraline in the treatment of the behavioral manifestations of Alzheimer's disease in outpatients treated with donepezil. Int J Geriatr Psychiatry. 2004;19:9-18. FULL TEXT | ISI | PUBMED

31. Lyketsos CG, DelCampo L, Steinberg M, et al. Treating depression in Alzheimer disease: efficacy and safety of sertraline therapy, and the benefits of depression reduction: the DIADS. Arch Gen Psychiatry. 2003;60:737-746. FREE FULL TEXT

32. Auchus AP, Bissey-Black C. Pilot study of haloperidol, fluoxetine, and placebo for agitation in Alzheimer's disease. J Neuropsychiatry Clin Neurosci. 1997;9:591-593. FREE FULL TEXT

33. Teri L, Logsdon RG, Peskind E, et al. Treatment of agitation in AD: a randomized, placebo-controlled clinical trial. Neurology. 2000;55:1271-1278. FREE FULL TEXT

34. Tariot PN, Erb R, Podgorski CA, et al. Efficacy and tolerability of carbamazepine for agitation and aggression in dementia. Am J Psychiatry. 1998;155:54-61. FREE FULL TEXT

35. Olin JT, Fox LS, Pawluczyk S, Taggart NA, Schneider LS. A pilot randomized trial of carbamazepine for behavioral symptoms in treatment-resistant outpatients with Alzheimer disease. Am J Geriatr Psychiatry. 2001;9:400-405. FULL TEXT | ISI | PUBMED

36. Porsteinsson AP, Tariot PN, Erb R, et al. Placebo-controlled study of divalproex sodium for agitation in dementia. Am J Geriatr Psychiatry. 2001;9:58-66. FULL TEXT | ISI | PUBMED

37. Tariot PN, Schneider LS, Mintzer J, et al. Safety and tolerability of divalproex sodium in the treatment of signs and symptoms of mania in elderly patients with dementia: results of a double-blind, placebo-controlled trial. Curr Ther Res Clin Exp. 2001;62:51-67. FULL TEXT

38. Sival RC, Haffmans PM, Jansen PA, Duursma SA, Eikelenboom P. Sodium valproate in the treatment of aggressive behavior in patients with dementia: a randomized placebo controlled clinical trial. Int J Geriatr Psychiatry. 2002;17:579-585. FULL TEXT | ISI | PUBMED

39. Lyketsos CG, Lee HB. Diagnosis and treatment of depression in Alzheimer's disease: a practical update for the clinician. Dement Geriatr Cogn Disord. 2004;17:55-64. FULL TEXT | ISI | PUBMED

40. Bains J, Birks JS, Dening TR. The efficacy of antidepressants in the treatment of depression in dementia. Cochrane Database Syst Rev. 2002;4:CD003944. PUBMED

41. Olin J, Schneider L. Galantamine for Alzheimer's disease. Cochrane Database Syst Rev. 2002;3:CD001747. PUBMED

42. Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase inhibitors in the treatment of neuropsychiatric symptoms and functional impairment in Alzheimer disease: a meta-analysis. JAMA. 2003;289:210-216. FREE FULL TEXT

43. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359:1283-1290. FULL TEXT | ISI | PUBMED

44. Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E. A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer's disease. Neurology. 2001;57:613-620. FREE FULL TEXT

45. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356:2031-2036. FULL TEXT | ISI | PUBMED

46. Tariot PN, Cummings JL, Katz IR, et al. A randomized, double-blind, placebo-controlled study of the efficacy and safety of donepezil in patients with Alzheimer's disease in the nursing home setting. J Am Geriatr Soc. 2001;49:1590-1599. FULL TEXT | ISI | PUBMED

47. Courtney C, Farrell D, Gray R, et al. Long-term donepezil treatment in 565 patients with Alzheimer's disease (AD2000): randomised double-blind trial. Lancet. 2004;363:2105-2115. FULL TEXT | ISI | PUBMED

48. Holmes C, Wilkinson D, Dean C, et al. The efficacy of donepezil in the treatment of neuropsychiatric symptoms in Alzheimer disease. Neurology. 2004;63:214-219. FREE FULL TEXT

49. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. Memantine in moderate-to-severe Alzheimer's disease. N Engl J Med. 2003;348:1333-1341. FREE FULL TEXT

50. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291:317-324. FREE FULL TEXT

51. Rockwood K, Mintzer J, Truyen L, Wessel T, Wilkinson D. Effects of a flexible galantamine dose in Alzheimer's disease: a randomised, controlled trial. J Neurol Neurosurg Psychiatry. 2001;71:589-595. FREE FULL TEXT

52. Tariot PN, Solomon PR, Morris JC, Kershaw P, Lilienfeld S, Ding C. A 5-month, randomized, placebo-controlled trial of galantamine in AD: the Galantamine USA-10 Study Group. Neurology. 2000;54:2269-2276. FREE FULL TEXT

53. Rockwood K, MacKnight C. Assessing the clinical importance of statistically significant improvement in anti-dementia drug trials. Neuroepidemiology. 2001;20:51-56. FULL TEXT | ISI | PUBMED

54. Greenspan A, Eerdekens M, Mahmoud R. Is there an increased rate of cerebrovascular events among dementia patients? Poster presented at: 24th Congress of the Collegium Internationale Neuro-Psychopharmacologicum (CINP); June 20-24, 2004; Paris, France.

55. Cavazzoni P, Young C, Polzer J, et al. Incidence of cerebrovascular adverse events and mortality during antipsychotic clinical trials of elderly patients with dementia. Poster presented at: 44th Annual New Clinical Drug Evaluation Unit; June 1-4, 2004; Phoenix, Ariz.

56. Wilcock GK. Dementia with Lewy bodies. Lancet. 2003;362:1689-1690. FULL TEXT | ISI | PUBMED
57. Baskys A. Lewy body dementia: the litmus test for neuroleptic sensitivity and extrapyramidal symptoms. J Clin Psychiatry. 2004;65(suppl 11):16-22. PUBMED

58. Ballard C, Grace J, McKeith I, Holmes C. Neuroleptic sensitivity in dementia with Lewy bodies and Alzheimer's disease. Lancet. 1998;351:1032-1033. FULL TEXT | ISI | PUBMED

59. McKeith IG, Ballard CG, Harrison RW. Neuroleptic sensitivity to risperidone in Lewy body dementia. Lancet. 1995;346:699. FULL TEXT | ISI | PUBMED

60. Sechi G, Agnetti V, Masuri R, et al. Risperidone, neuroleptic malignant syndrome and probable dementia with Lewy bodies. Prog Neuropsychopharmacol Biol Psychiatry. 2000;24:1043-1051. FULL TEXT | PUBMED

61. Walker Z, Grace J, Overshot R, et al. Olanzapine in dementia with Lewy bodies: a clinical study. Int J Geriatr Psychiatry. 1999;14:459-466. FULL TEXT | ISI | PUBMED

62. Clark ME, Lipe AW, Bilbrey M. Use of music to decrease aggressive behaviors in people with dementia. J Gerontol Nurs. 1998;24:10-17. PUBMED

63. Gerdner LA. Effects of individualized versus classical "relaxation" music on the frequency of agitation in elderly persons with Alzheimer's disease and related disorders. Int Psychogeriatr. 2000;12:49-65. FULL TEXT | ISI | PUBMED

64. Ballard CG, O'Brien JT, Reichelt K, Perry EK. Aromatherapy as a safe and effective treatment for the management of agitation in severe dementia: the results of a double-blind, placebo-controlled trial with Melissa. J Clin Psychiatry. 2002;63:553-558. ISI | PUBMED

65. Holmes C, Hopkins V, Hensford C, MacLaughlin V, Wilkinson D, Rosenvinge H. Lavender oil as a treatment for agitated behaviour in severe dementia: a placebo controlled study. Int J Geriatr Psychiatry. 2002;17:305-308. FULL TEXT | ISI | PUBMED

66. Cohen-Mansfield J. Nonpharmacologic interventions for inappropriate behaviors in dementia: a review, summary, and critique. Am J Geriatr Psychiatry. 2001;9:361-381. FULL TEXT | ISI | PUBMED

67. Doody RS, Stevens JC, Beck C, et al. Practice parameter: management of dementia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2001;56:1154-1166. FREE FULL TEXT

68. Sultzer DL, Gray KF, Gunay I, Wheatley MV, Mahler ME. Does behavioral improvement with haloperidol or trazodone treatment depend on psychosis or mood symptoms in patients with dementia? J Am Geriatr Soc. 2001;49:1294-1300. FULL TEXT | ISI | PUBMED

69. Wagner AK, Zhang F, Soumerai SB, et al. Benzodiazepine use and hip fractures in the elderly: who is at greatest risk? Arch Intern Med. 2004;164:1567-1572. FREE FULL TEXT

70. Hogan DB, Maxwell CJ, Fung TS, Ebly EM. Prevalence and potential consequences of benzodiazepine use in senior citizens: results from the Canadian Study of Health and Aging. Can J Clin Pharmacol. 2003;10:72-77. PUBMED

71. Treatment of agitation in older persons with dementia: the Expert Concensus Panel for Agitation in Dementia. Postgrad Med. 1998;Spec No:1-88. PUBMED

72. Stoudemire A, Smith DA. OBRA regulations and the use of psychotropic drugs in long-term care facilities: impact and implications for geropsychiatric care. Gen Hosp Psychiatry. 1996;18:77-94. FULL TEXT | ISI | PUBMED

73. Ruths S, Straand J, Nygaard HA, Bjorvatn B, Pallesen S. Effect of antipsychotic withdrawal on behavior and sleep/wake activity in nursing home residents with dementia: a randomized, placebo-controlled, double-blinded study: the Bergen District Nursing Home Study. J Am Geriatr Soc. 2004;52:1737-1743. FULL TEXT | ISI | PUBMED

74. Miles SH, Irvine P. Deaths caused by physical restraints. Gerontologist. 1992;32:762-766. ABSTRACT

75. Schneider LS, Tariot PN, Lyketsos CG, et al. National Institute of Mental Health Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE): Alzheimer disease trial methodology. Am J Geriatr Psychiatry. 2001;9:346-360. FULL TEXT | ISI | PUBMED

___

Clinical Review Section Editor: Michael S. Lauer, MD. We encourage authors to submit papers for consideration as a "Clinical Review." Please contact Michael S. Lauer, MD, at lauerm@ccf.org.
Comments: 0
Votes:23