CIA_1698_Hersch.indd © 2007 Dove Medical Press Limited. All rights reserved Clinical Interventions in Aging 2007:2(4) 611–621 611 R E V I E W Management of the behavioral and psychological symptoms of dementia Elizabeth C Hersch Sharon Falzgraf VA Puget Sound Health Care System, Tacoma, Washington, USA Correspondence: Elizabeth C Hersch Geriatrics and Extended Care (A-182GEC), VA Puget Sound Health Care System, Bldg 2, Room 344, 9600 Veterans Drive SW, Tacoma, WA 98493, USA Tel +1 253 582 8440 ext 72085 Fax +1 253 589 4105 Email herschec@yahoo.com Abstract: More than 50% of people with dementia experience behavioral and psychological symptoms of dementia (BPSD). BPSD are distressing for patients and their caregivers, and are often the reason for placement into residential care. The development of BPSD is associated with a more rapid rate of cognitive decline, greater impairment in activities of daily living, and diminished quality of life (QOL). Evaluation of BPSD includes a thorough diagnostic investiga- tion, consideration of the etiology of the dementia, and the exclusion of other causes, such as drug-induced delirium, pain, or infection. Care of patients with BPSD involves psychosocial treatments for both the patient and family. BPSD may respond to those environmental and psychosocial interventions, however, drug therapy is often required for more severe presentations. There are multiple classes of drugs used for BPSD, including antipsychotics, anticonvulsants, antidepressants, anxiolytics, cholinesterase inhibitors and NMDA modulators, but the evidence base for pharmacological management is poor, there is no clear standard of care, and treatment is often based on local pharmacotherapy customs. Clinicians should discuss the potential risks and benefi ts of treatment with patients and their surrogate decision makers, and must ensure a balance between side effects and tolerability compared with clinical benefi t and QOL. Keywords: dementia, management, behavioral symptoms, psychological symptoms Typical behavioral and psychological symptoms of dementia Eighty to 94% of residents of long term care facilities have a major psychiatric ill- ness. Dementia is the most prevalent, observed in 47%–78% of residents (Rovner et al 1990; DeVane and Mintzer 2003). Regardless of its etiology, dementia is a clinical syndrome that expresses itself in three areas: cognitive defi cits, psychiatric and behavioral disturbances, and diffi culties in carrying out daily functions (De Dyn et al 2005). Alois Alzheimer, in his 1906 description of dementia, noted behavioral and psychological symptoms of dementia (BPSD) are prominent manifestations of the illness, including paranoia, delusions of sexual abuse, hallucinations and screaming (Kozman et al 2006). In 1996, the International Psychogeriatric Association convened a consensus conference on the behavioral disturbances in dementia. The consensus group made this statement: “The term behavioral disturbances should be replaced by the term BPSD, defi ned as symptoms of disturbed perception, thought content, mood or behavior that frequently occur in patients with dementia (Kozman et al 2006, p 1).” BPSD is not a diagnostic entity but is instead a term that describes a clinical dimension of dementia (Lawlor 2004). The multiple cognitive impairments of dementia are often associated with mood disorders and sleep disturbances. BPSD includes disinhibited behavior, delusions and hallucinations, verbal and physical aggression, agitation, anxiety and depression (Carson et al 2006). BPSD can cause tremendous distress for both the patient and the caregiver, and is often the trigger for referral of these patients to primary care and specialist services and placement in residential or nursing home Clinical Interventions in Aging 2007:2(4)612 Hersch and Falzgraf care (Steele et al 1990; Ballard et al 2006). The development of BPSD is also associated with a poorer prognosis, a more rapid rate of cognitive decline, illness progression (Stern et al 1987; Paulsen et al 2000), greater impairment in activities of daily living (ADLs) (Lyketsos et al 1997) and diminished quality of life (QOL) (Gonzales-Salvador et al 2000), and it adds signifi cantly to the direct and indirect costs of care (O’Brien and Caro 2001). At least half of patients attending outpatient dementia clinics, and more than 75% of patients in nursing homes have some sort of BPSD (Zaudig 2000). The prevalence of BPSD in these 24 hour care settings has been reported to be as high as 90%, with individual behaviors including delusions (20%–73%), depression (up to 80%), and aggression and hostility (20%–50%). As many as 80% of Alzheimer’s dementia (AD) patients will develop symptoms of BPSD during the course of their illness, often with the onset of cognitive impairment (Lyketsos et al 2002). Patients with mixed AD and vascular dementia have the highest level of psychiatric disturbances (Zaudig 2000; Kindermann et al 2002; Kozman et al 2006). While the origin of BPSD remains unclear; it is presumed there are multiple etiologies for these symptoms. There are neurobiological, psychological (premorbid personality features and responses to stress), and social (environmental change and caregiver factors) aspects (Zaudig 2000). The neurobiology of behavioral disturbances involves correlations between memory defi cits and decreasing cholinergic function, and between serotonin and noradrenaline depletion and a his- tory of depression or aggression. Dysregulations in GABA (gamma-aminobutyrate)-ergic, serotonergic and noradrener- gic neurotransmitter systems that have been associated with increased aggressiveness and disturbances are also found in dementia patients (Eichelman 1987; Stoppe et al 1999). BPSD are now accepted as an important therapeutic target in dementia. Mild forms of BPSD may respond to simple environmental and psychosocial interventions. Although non-pharmacologic interventions should be the fi rst line of treatment, drug therapy is often required for the more severe psychotic, aggressive, and agitated presentations (Lawlor 2004; Sink et al 2005). Methods of measuring BPSD and the associated challenges It is important to remember that behavior is merely a form of communication (Kozman et al 2006). BPSD can be diffi cult to diagnose, given the variety of symptoms. Evaluation of BPSD includes a thorough diagnostic investigation, careful consideration of the etiology of the dementia, and the exclusion of other causes, such as drug-induced delirium or adverse effects of treatments for comorbid conditions (Finkel et al 1996; Zaudig 2000). Undiagnosed medical problems such as pain, depression, dehydration, sleeping diffi culty, anxiety and delirium can all lead to agitation. Pain is often underdiagnosed in patients with demen- tia, and can manifest itself by behavior changes (such as agitation and increased confusion) and decreased mobility (Pautex et al 2006). Language diffi culties associated with dementia interfere with the patient’s ability to express pain. In addition, the autonomic activation in response to pain may be blunted in AD patients. A number of pain scales have been developed to evaluate pain in patients with dementia. Some are self-report (for milder dementia); others, such as the PAIN-AD-Pain Assessment in Advanced Dementia (Lane 2003), measure non-verbal signs such as breathing, vocalization, facial expression, and body language. A small study (Douzjian 1998) found that empiric pain medication reduced troublesome behaviors, and allowed for a reduction in psychoactive medication. Positioning and physical therapy may able be helpful. Sleep disturbances may be associated with and part of BPSD. Circadian rhythms may be altered in AD. Patients with Lewy Body dementia have a high incidence of REM sleep disorders, acting out their dreams. Clinicians should evaluate medications that may disrupt sleep. Other common medical causes of confusion and agitation in the elderly include infections, endocrine disorders, fl uid and electrolyte imbalances, and constipation (Daniel 2000). Emotional and interpersonal issues (such as dislike for certain foods or reactions of other people to the patient’s behavior) can be a signifi cant factor in agitation. Environmental factors, such as enclosed spaces, isolation, and visual and auditory sensory deprivation may all contribute to or cause a problem. The diagnostic evaluation and treatment of BPSD require special considerations. Institutionalized, demented and acutely ill elders, particularly those taking multiple medications, are at risk for BPSD. The aging brain provides a different substrate for both the therapeutic and potentially toxic effects of medications. In elderly patients with degen- erative brain disorders, the normal redundancy, interdepen- dence, and checks and balances of neuronal networks and neurotransmitters may be disturbed or defi cient. For example, delirium may be reversible if the underlying medical causes are addressed promptly, or fatal if overlooked or untreated. A careful medication review should be performed, paying particular attention to any recently introduced medications. Clinical Interventions in Aging 2007:2(4) 613 Management of BPSD Elderly patients may be more vulnerable to the cognitive effects of drug interactions or to what may be considered therapeutic blood drug levels in younger patients (Stoppe et al 1999; Daniel 2000). Many medications can cause delirium or dementia-like symptoms (Tariot, Profenno et al 2004). While drugs with anticholinergic properties are often associated with delirium or behavior changes, many others such as psychoactive and cardiovascular drugs may also be implicated. More than 30 scales are available for measuring the behavioral manifestations of dementia (Weiner et al 1996; Stoppe 1999). A number of instruments have been developed to assess the range and severity of BPSD (see Table 1). The most useful, in terms of outcomes assessment, are the Cohen- Mansfi eld Agitation Inventory (CMAI), neuropsychiatric inventory: nursing home version (NPI-NH), and behavioral pathology in AD (BEHAVE-AD) scales. They are particu- larly useful because of their specifi city, reliability and validity in BPSD (Zaudig 2000; De Deyn et al 2001). It is not clear how much of a percentage change in each assessment tool represents a clinically signifi cant response, however (Lee et al 2004). Recognition of BPSD is the fi rst step in developing a management plan, and care must be taken to establish its presence. The goal of treatment should be to detect and manage BPSD before caregiver burnout and irreversible damage to the support environment occurs. The plan should consider the severity and intrusiveness of the behavior and whether non-pharmacologic intervention is suffi cient or the behavior is signifi cant enough to require both pharmacologic and psychological interventions (Lawlor 2004). Identify- ing target syndromes (such as psychotic syndrome versus psychomotor agitation or a sleep disturbance) is useful in development of the management plan. Useful clinical out- comes such as nursing home placement, QOL, and caregiver burden improve a clinician’s ability to interpret test results and inform patients and families about the risks and benefi ts of treatment (Sink et al 2005). Non-pharmacologic therapy and its evidence Care of patients with BPSD involves a broad range of psycho- social treatments for both the patient and family. Caregiver education, support and behavioral training are integral parts of the intervention for these patients (Lawlor 2004; Sink et al 2005). Interventions need to be approached in a systematic manner that includes management of the patient’s physical health, psychiatric symptoms, and environmental factors (Kozman et al 2006). In a study by Palmer et al (1999), the use of hearing aids improved scores on the BEHAVE-AD. Environmental adjustments, such as lifestyle support are generally fi rst line interventions; however, many cases of aggression, agitation and psychotic symptoms may require pharmacotherapy (Zaudig 2000). Individualized music therapy, bright light treatment (BLT) and aromatherapy have been found to improve certain prob- lematic behavioral symptoms (Lawlor 2004), as has pet therapy (Sink et al 2005). In a randomized controlled trial (RCT) by Smallwood et al (2000), a combination of aromatherapy and massage mid-afternoon resulted in a reduction in the fre- quency of behavioral disturbances. In a larger, double-blind, placebo-controlled trial, aromatherapy was found to be a safe and effective treatment for clinically signifi cant agitation in patients with severe dementia (Ballard et al 2002). Lovell et al (1995) studied the effect of BLT on levels of agitation in moderately to severely demented persons. Agitation scores were signifi cantly reduced during therapy compared with controls (Lovell et al 1995). Good sleep hygiene, avoidance of Table 1 Common scales used for measuring the behavioral manifestations of dementia Test name Test description Cohen-Mansfi eld Developed in 1986. It examines 29 Agitation Inventory types of agitated behavior, including (CMAI) pacing, verbal or physical aggression, repetitious mannerisms, screaming and general restlessness.a Neuropsychiatric Inventory: Assesses 12 behavioral disturbances: Nursing Home delusions, hallucinations, agitation, version (NPI-NH) dysphoria, anxiety, apathy, irritability, euphoria, disinhibition, aberrant motor behavior, nighttime behavior disturbances and appetite and eating abnormalities.b Behavioral Pathology in Developed in 1987. It is a structured AD (BEHAVE-AD) psychiatric interview, assessing 25 behaviors in 7 areas: paranoid and delusional ideation, hallucinations, activity disturbances, aggressiveness, diurnal rhythm disturbances, affective disturbance and anxieties and phobia.c Clinical Global Impression Establishes a global rating of all aspects of Change (CGI-C) of the patient’s condition. Functional Assessment Measures the levels of basic activities Staging scale (FAST) such as bathing and toileting, and rates patients from independent to totally dependent. aBallard et al 2006; Kozman et al 2006. bCarson et al 2006. cCarson et al 2006; Kozman et al 2006. Clinical Interventions in Aging 2007:2(4)614 Hersch and Falzgraf caffeine and alcohol, and adequate daytime physical activity can be benefi cial, particularly for patients who have sleep disturbances and depression. In a randomized controlled trial by Teri et al (2003), a combination of exercise training and caregiver education on behavioral management techniques resulted in improvements in depression and a trend toward less institutionalization. Teaching caregivers techniques to minimize behavior problems can make the home environ- ment less stressful for both the family and the patient (see Table 2). Sloane et al (2004) performed an RCT which mea- sured agitation and aggression, comparing usual hygiene with person-centered showering and towel baths. Results showed that the latter techniques were effective methods of reducing BPSD during bathing persons with dementia. Finally, physi- cal restraints should be avoided, for they are associated with injury, not protection, of confused or demented patients (Miles et al 1992; Sink et al 2005). Pharmacologic management and its evidence The evidence base for drug treatment of the behavioral and psychological symptoms of dementia is poor, considering the size of the problem and the distress these symptoms cause. Over the years, drug prescribing for BPSD has evolved in a haphazard and anecdotal way. Although there are multiple classes of drugs in use for neuropsychological symptoms, including antipsychotics, anticonvulsants, antidepressants, anxiolytics, cholinesterase inhibitors and NMDA modula- tors, there is no consensus nor clear standard of care, and treatment is often based on local pharmacotherapy customs (Sink et al 2005). A balance needs to be made between side effects and tolerability and safety issues compared with clini- cal benefi t and QOL (Kozman et al 2006). In elderly patients, it is possible that any medication could help and/or harm, and the safety of a drug must be considered in the context of its known effi cacy (Schneider et al 2005). Since the neurobiology of BPSD is still unclear, it seems likely that the symptoms may involve different neurotransmitter systems and may therefore respond to different therapies (Kozman et al 2006). Drug-responsive symptoms include anxiety, verbal and physical agitation, hallucinations, delusions, and hostility, whereas wandering, hoarding, unsociability, poor self-care, screaming and other stereotypical behavior seem to be unresponsive to all drugs (Maletta 1990; Stoppe et al 1999). Pharmacologic interven- tion is often necessary and includes use of antidepressants for mood disorders, anticonvulsants for nonpsychotic agitation and antipsychotics for aggression, agitation and psychotic symptoms. Antidepressants, anxiolytics and hypnotics should only be used in patients with marked and persistent symptoms and drug treatment should be targeted to specifi c syndromes that are clinically signifi cant because of their frequency, pervasiveness, or impact (Lawlor 2004). If drug therapy is to be instituted, Sink et al (2005) recommend two approaches to the management. One is to identify the target symptom and choose a drug that is known to treat symptoms most closely related to the one the patient is exhibiting (as mentioned above). An alternative approach is one guided by current evidence in combination with the goal of minimizing side effects. They recommend beginning with a cholinesterase inhibitor if the patient is not already on one, because they are well tolerated and may benefi t cognition and function. It is important to remember that titration speed and target dosage of psychoactive drugs are substantially reduced in the elderly (Daniel 2000). Benzodiazepines should be avoided, especially for long term management, as their use can lead to increased confusion, falls and may paradoxically increase agitation in patients with dementia. No psychoactive medication prescribed to treat neuropsychiatric symptoms of dementia should be continued indefi nitely and attempts at Table 2 Behavioral management interventions Activity associated Intervention with potential BPSD Bathing Make a safe bathroom. Be prepared, don’t rush. Ensure room and water temperature are comfortable. Wash hair last. A recent study found benefi t of person-centered bathing and towel bath in decreasing agitation and discomfort.a Dressing Limit choices. Prepare clothing. Give specifi c cues. Provider larger clothing and soft stretchy fabrics. Provide duplicate outfi ts and comfortable shoes with Velcro. Give positive reinforcement. Eating Maintain a regular mealtime. Avoid distraction at meals. Check the food temperature. Honor preferences when possible, and offer fi nger foods. Wandering Provide adequate daily physical activity. Create a safe environment and safe wandering paths. Remove reminders of leaving (coats, umbrella). Have alarms or bells at exit doors. ID bracelet and “Safe Return” programs are available. Incontinence Scheduled voiding. Be attentive to nonverbal cues (such as pacing). Simplify clothing and clear obstacles. Put signs (including pictures) at the bathroom door. Give positive reinforcement. aSloane et al 2004. Clinical Interventions in Aging 2007:2(4) 615 Management of BPSD drug withdrawal should be made regularly. Because of the instability of the symptoms of BPSD, many patients who are prescribed antipsychotics for neuropsychiatric symptoms will no longer need them when the drug is later discontinued (Stoppe et al 1999). Medications commonly used to treat BPSD In psychotic, behaviorally disturbed elders, an ideal medication should have rapid onset, sustained action and minimal somatic and cognitive side effects (Daniel 2000). Conventional anti- psychotics, such as haloperidol, have been used effectively to control the behavioral and psychological symptoms of demen- tia. Other drugs, such as valproate and carbamazepine, have shown some effi cacy in controlling behavioral symptoms in elderly patients (Mellow et al 1993; Tariot et al 1994; DeVane and Mintzer 2003). However, only the atypical antipsychotics risperidone and olanzapine currently have the best evidence of effi cacy in treating neuropsychiatric symptoms. Trials of cholinesterase inhibitors have had consistent yet small positive effects as well (Sink et al 2005). Antipsychotics Antipsychotic medications have been the mainstay of psycho- pharmacological treatment for BPSD during the last several decades despite their overuse in the 1980s and the federal regulations implemented in the early 1990s (Schneider et al 2005). Up until the mid 1990s, conventional neuroleptics such as haloperidol were the primary pharmacologic treat- ments for BPSD. Antipsychotics are the drugs of choice in the treatment of intrusive delusions and hallucinations. Coexisting nonpsychotic symptoms including sleeplessness, excitability, hostility, belligerence, emotional liability, rest- lessness, agitation, aggression and irritability may also show improvement with antipsychotics. Other symptoms such as hypersexuality, apathy and withdrawal do not generally improve. Since antipsychotics have such a narrow therapeutic window, they should be prescribed and dosage adjusted with the expectation of clinical improvement within a certain timeframe (Kindermann et al 2002). If improvement is not observed, the medication could be discontinued or switched after two to four weeks (Schneider et al 2005; Schneider, Tariot et al 2006). Low dosages and careful dose titration are needed when prescribing them to the elderly with dementia. However, these low dosages may also lead to limited effi cacy (Stoppe et al 1999). In the past, antipsychotic use in dementia has been excessive, possibly inappropriate, and poorly monitored. The goal of antipsychotic therapy must be the improvement in a specifi c target behavioral syndrome without impairing other aspect of dementia such as cognition, function, and quality of life (Lawlor 2004). Antipsychotics are more effective than placebo, but the effect is modest and they are frequently associated with adverse effects, includ- ing increased risk of falls and drowsiness, parkinsonism, akathisia, tardive dyskinesa (TD), social withdrawal, accelerated cognitive decline, QT prolongation, stroke, and sensitivity reactions (Ballard et al 2006). Both conventional antipsychotics and just the presence of psychosis have been associated with more rapid cognitive decline in dementia patients (Schneider, Dagerman et al 2006). Patients with Lewy Body dementia have been reported to have marked sensitivity, including neuroleptic malignant syndrome, to typical and atypical antipsychotics, especially risperidone (Ballard et al 1998; Sink et al 2005). The newer atypical antipsychotics are associated with fewer extrapyramidal symptoms (EPS) (Jeste et al 1999; De Dyn et al 2005). The perceived safety advantages of the atypi- cals include less of the following: sedation, cardiovascular (CV) adverse effects, postural instability, falls and movement disorders. The FDA added warnings of increased CV adverse events to the US prescribing information for some atypical antipsychotics in April 2003, January 2004 and February 2005. In April 2005, the FDA issued a health advisory for increased risk for death with use of atypicals in patients with dementia (Kozman 2006; Schneider, Dagerman et al 2006). Atypical antipsychotics The American Academy of Neurology recommends the use of antipsychotics to treat agitation and psychosis in patients with dementia where environmental manipulation fails, and guidelines state that atypical antipsychotics may be better tolerated than older conventional antipsychotics (Doody et al 2001; Carson et al 2006). Clozapine, the prototypical atypical antipsychotic, was approved in 1989. Since then, risperidone, olanzapine, quetiapine, ziprasidone and aripip- razole have been introduced (Carson et al 2006). Risperidone was the fi rst agent to be proven effective for the behavioral and psychological diagnoses of dementia, and it has the largest database of double-blind controlled trials to support its effi cacy (De Dyn et al 2005). It is effective in controlling aggression, agitation and psychotic symptoms in patients with many different forms of dementia. It is well tolerated and does not further impair the daily function of elderly patients with dementia (Zaudig 2000). Atypical antipsychotic drugs in general are widely used to treat psychosis, aggression and Clinical Interventions in Aging 2007:2(4)616 Hersch and Falzgraf agitation in patients with AD, however, their benefi ts are uncertain and concerns about safety have emerged. In a double-blind, placebo-controlled trial (Schneider, Tariot et al 2006), 421 outpatients with AD and psychosis, aggression or agitation were randomly assigned to receive risperidone, olanzapine, quetiapine, or placebo. No signifi - cant differences were noted with regard to improvement on the CGIC scale. Adverse effects offset advantages in the effi cacy of atypicals for the treatment of BPSD (Schneider, Tariot et al 2006). A 2006 Cochrane Review (Ballard et al 2006) evaluated 16 placebo-controlled studies with atypicals. Only 9 had suffi cient data for a meta-analysis. The review looked at outpatients or people living in care facilities and found (Ballard et al 2006, p 5): 1. A signifi cant improvement in aggression with risperidone and olanzapine compared to placebo. 2. A s i g n i f i c a n t i m p r o v e m e n t i n p s y c h o s i s w i t h risperidone. 3. Risperidone and olanzapine patients had a signifi cantly higher incidence of serious adverse CV events and EPS (especially with risperidone doses greater than 1 milligram). 4. A signifi cant increase in drop-outs with risperidone and olanzapine. 5. Data was insufficient to comment on cognitive function. These reviewers and numerous other studies throughout the literature have come to similar conclusions that, over- all, the evidence for olanzapine and risperidone supports their effi cacy compared with placebo, but their potential for increased risk of CV events and mortality is a serious concern and limits their overall effectiveness (Ballard et al 2006; Carson et al 2006; Schneider, Tariot et al 2006). Like- wise, the Committee on the Safety of Medications reported a three-fold increase in the risk of CV adverse events with the atypicals, compared with placebo. Both olanzapine and risperidone have good evidence base and they appear to be well tolerated otherwise. Considering the consistency of the risks among the various studies reviewed, it is likely that there is increased risk from any of the atypicals and not from a particular one (Schneider et al 2005). The modest effi cacy and uncertain response rates combined with the risks detailed suggest that antipsychotics should be used within the context of medical need and the effi cacy and safety of alternatives (Schneider, Dagerman et al 2006). Their use should be targeted towards the treatment of those patients in whom BPSD are prominent and associated with signifi cant distress, functional impairment or danger to the patient (De Dyn et al 2005). The use of lower doses might be prudent and effective (Schneider, Dagerman et al 2006). The adverse effects associated with atypical antipsychotics seem to be dose related, supporting the practice of starting with a low dose and increasing slowly as the drug is tolerated. In addition to the adverse CV effects discussed above, there is increasing evidence that the treatment with antipsychotics may be associated with metabolic disturbances such as impaired glucose metabolism, hyperlipidemia, and weight gain, all of which may adversely affect QOL (Lee et al 2004; Jones et al 2006). Neutropenia and agranulocytosis may also occur with olanzapine (Benedetti et al 1999; Zaudig 2000). There were no clinically relevant changes in blood pressure or heart rate in phase III studies with risperidone, nor were there any occurrences of EKG abnormalities (Katz et al 1999; Zaudig 2000). There has been no evidence for increased injury, fall or syncope; however the risks of somnolence and urinary tract infections (or incontinence) are increased with the atypicals (Schneider, Dagerman et al 2006). Sedation is particularly likely with olanzapine, which can also be associated with increased confusion (Schneider, Tariot et al 2006). Conventional antipsychotics Conventional (or typical) antipsychotics have been exten- sively studied in elderly demented patients with disappointing results. Effect size and response rates have been modest, with no consistent evidence that any conventional antipsychotic is more effective than another. Potentially serious adverse effects occur frequently, especially motor side effects, sedation, cognitive impairment, orthostatic hypotension, constipation, and urinary hesitancy (Daniel 2000; Lee et al 2004). In the US, concerns about overuse of antipsychotics led to the introduction of legislation (the Omnibus Budget Reconciliation Act of 1987) that attempted to restrict pre- scribing of antipsychotics to residents of nursing homes (Streim 1995; Stoppe et al 1999). Before the introduction of this act, up to 55% of nursing home residents were treated with antipsychotics (Lee et al 2004). Haloperidol is the most widely studied typical antipsy- chotic and modest improvement in psychosis and agitation has been reported in investigations with it (Kindermann et al 2002). A Cochrane review of haloperidol compared with placebo con- cluded that haloperidol had no effect on agitation, behavioral symptoms as a whole, or CGIC scores, but appeared to reduce aggression (Sink et al 2005; Kozman et al 2006). In two other meta-analyses there was no difference in effi cacy among the different typical antipsychotics on neuropsychiatric symptoms (these studies included haloperidol, thioridazine, thiothixene, Clinical Interventions in Aging 2007:2(4) 617 Management of BPSD chlorpromazine, trifl uoperazine and acetophenazine). There is no evidence that any one typical antipsychotic is more effec- tive than another (Sink et al 2005). In addition, conventional antipsychotics were associated with a signifi cantly higher adjusted risk of death than were atypicals in all subgroups defi ned according to the presence or absence of dementia or nursing home residency (Wang et al 2005). The greatest increases in risk occurred soon after therapy was initiated and with higher dosages. Conventional antipsychotics are at least as likely as atypicals to increase the risk of death among elderly persons and should not be used to replace the atypicals that were discontinued in response to the 2005 FDA warning (Wang et al 2005). Elderly patients are sensitive to EPS, especially parkin- sonism, akathisia, and TD. Only one third of patients with dementia show behavioral improvement with conventional antipsychotic treatment, while the majority of patients treated with these will experience side effects, especially the anticho- linergic effects (Tariot, Profenno et al 2004). The presence of EPS can lead to medication intolerance, falls and other adverse effects. In addition, typical antipsychotics have seri- ous, potentially fatal, consequences in patients with Lewy Body dementia (Ballard et al 1998; Zaudig 2000). In one study, patients started on very low dosages of haloperidol or thioridazine developed parkinsonism within nine months of antipsychotic therapy (Kindermann et al 2002). The lower incidence of EPS and TD seen with atypicals represents a signifi cant benefi t to patients. Choosing antipsychotic agents with fewer anticholinergic properties is an important consid- eration in elderly patients. Anticonvulsants Anecdotal reports have suggested that anticonvulsants such as carbamazepine, valproic acid and gabapentin may be effective in the treatment of BPSD. Gabapentin has shown some benefi t when treating aggressive behavior in patients with dementia, but it has not been well studied (Hawkins et al 2000; Kozman et al 2006). Carbamazepine has been investigated in several trials and was found to reduce agita- tion, restlessness and anxiety (Tariot et al 1994; Stoppe et al 1999), however the effi cacy and tolerability of long term use of this drug is yet to be established (Kozman et al 2006). Ataxia can occur in elderly patients treated with carbamaze- pine (Kindermann et al 2002) and in the United States there is an FDA “black box” warning for hematologic toxicity and the potential for drug-drug interaction with its use (Sink et al 2005). Valproic acid had been reported to show some positive effects, with a benign adverse effect profi le (Mellow et al 1993; Stoppe et al 1999); however, in more recent studies it does not appear to be effective for the treatment of neuropsychiatric symptoms of dementia, whether in short or long acting preparations. It also caused signifi cantly more adverse effects than placebo, especially sedation. Therefore, valproic acid is not routinely recommended for the treatment of BPSD (Sink et al 2005). Antidepressants Depression is common in patients with dementia. As many as 40% of patients with dementia have signifi cant depres- sive symptoms at some stage. Reducing symptoms such as irritability may aid in the treatment of BPSD (Kozman et al 2006). Biochemical data have suggested that serotonergic defi cits in AD contribute to aggressive verbal and physi- cal outbursts, sleep disturbance, depression and psychosis (Polluck et al 2002). Some antidepressants have signifi cant side effects, and not all studies have shown effi cacy in treat- ment of neuropsychiatric symptoms of dementia other than depression (Sink et al 2005; Kozman et al 2006). Selective serotonin reuptake inhibitors (SSRI’s) may have “neuroleptic” effects by reducing dopaminergic outfl ow, and dysregulation in serotonergic neurotransmission may play an important role in the psychotic symptoms of dementia patients (Polluck et al 2002). Citalopram is the most selec- tive, with moderate potency and high bioavailability. An open pilot study using citalopram demonstrated that it was well tolerated and the patients experienced a signifi cant reduction in agitation, hostility and suspicion (Polluck et al 1997). One review found that citalopram signifi cantly improved emotional bluntness, confusion, irritability, anxi- ety, fear, depressed mood, and restlessness (Nyth et al 1990; Kozman et al 2006). In AD patients, psychotic and nonpsy- chotic behavioral disturbances improved acutely with both citalopram and perphenazine. However, only citalopram demonstrated acute effi cacy superior to placebo (Polluck et al 2002). Sertraline, in conjunction with donepezil, showed statistically signifi cant improvement in CGIC scores (Finkel et al 2004; Kozman et al 2006), but in another trial, there was no signifi cant benefi t of sertraline on neuropsychiatric symptoms other than depression (Lyketsos et al 2003; Sink et al 2005). Trazodone is widely used for agitation, sleep disorders, and disruptive behavior because of its sedative effect and negligible anticholinergic activity. A comparison of trazodone with haloperidol for treatment of agitation in 28 patients with dementia showed similar overall effi cacy of both drugs and a lower rate of adverse effects in the trazodone group (Sultzer et al 1997; Stoppe et al 1999). Clinical Interventions in Aging 2007:2(4)618 Hersch and Falzgraf Cholinesterase inhibitors Cholinesterase inhibitors are licensed for the treatment of mild to moderate AD (Kozman et al 2006). In some studies, donepezil had no effect on neuropsychiatric symptoms while in one study, anxiety, depression/dysphoria and apathy were signifi cantly improved compared with placebo (Feldman et al 2001; Kozman et al 2006). In Lewy Body dementia, donepezil has shown signifi cant improvement over time in behavioral symptoms (Lanctot and Herrmann 2000; Kozman et al 2006). Galantamine has demonstrated effectiveness, and has shown a signifi cant reduction in behavioral disturbances and improvement in the total NPI caregiver burden. A study using rivastigmine showed that long term therapy with it can slow the progression of BPSD symptoms, including aggressiveness and activity disturbances (Rosler et al 1999; Kozman et al 2006). In another study, rivastigmine signifi - cantly improved neuropsychiatric and behavioral symptoms compared with baseline (Aupperle et al 2004; Kozman et al 2006). Although some of these trials have shown statistically signifi cant differences, most data reporting the benefi ts on behavioral and psychological disturbances are from second- ary outcome measures and the magnitude of effect has been small and of questionable clinical signifi cance (Sink et al 2005). Further research is needed. Other medications Medications such as memantine, buspirone, beta blockers, benzodiazepines, and thiothixene (Finkel et al 1995) have been evaluated for their use in treating BPSD. Adding memantine to donepezil resulted in better outcomes (than placebo) for dementia patients on measures of cognition, ADLs, global outcome, and behavior. It showed a signifi - cantly benefi cial effect compared with placebo in relation to agitation and aggression (Tariot, Farlow et al 2004; Kozman et al 2006). However, there does not appear to be a clini- cally signifi cant benefi t using memantine in the treatment of neuropsychiatric symptoms of patients with moderate to severe AD (Sink et al 2005). A case report noted the addition of buspirone to antidepressants and olanzapine after 2 weeks resulted in signifi cant improvements (Cooper 2003; Kozman et al 2006). Its benign adverse effect profi le also makes buspirone a useful alternative in mild agitation. However, it showed less effect on agitation compared with trazodone or placebo (Lawlor et al 1994; Stoppe et al 1999). Low dose propranolol was effective in reducing disruptive, aggressive behavior in the majority of outpatients in one study; however, there are no controlled trials on use of beta blockers in BPSD (Shankle et al 1995; Kozman et al 2006). Benzodiazepines show signifi cantly more improvement in BPSD symptoms when compared with placebo; however, they should be used with caution because of their adverse effects (Stoppe et al 1999; Zaudig 2000; Kindermann et al 2002). Risks associated with pharmacologic treatment of BPSD The elderly are at particular risk for drug-related adverse events. Older patients in general, and patients with dementia in particular, are more sensitive to medication adverse effects, including anticholinergic effects, orthostatic hypotension, sedation, parkinsonism, tardive dyskinesia (TD) and cog- nitive impairment than younger patients with dementia or individuals without dementia. They are also especially prone to falls, and drugs that cause sedation, postural hypotension or extrapyramidal symptoms (EPS) have been found to cause an increased incidence of falls (Zaudig 2000). The aging body undergoes physiological changes that affect both pharmacokinetics and pharmacodynamics (DeVane and Mintzer 2003). Aging is characterized by a progressive loss of the functional capacities of all the vital organs, including the brain. There is an alteration in the body’s composition of lean and fat body mass and a decrease in the excretory capacity of the kidney, causing many drugs to be eliminated from the body more slowly. Changes in drug distribution are also important. Age related reductions in liver mass, hepatic blood fl ow and hepatocyte function result in delayed clearance of drugs that are metabolized in the liver. Lower starting doses, smaller dose increments and longer dose escalation periods must be used to avoid potentially toxic drug accumulation. Concurrent illnesses and associated polypharmacy are extremely common in elderly patients. Potential drug-drug interactions must be considered (Zaudig 2000). Central and peripheral anticholinergic adverse effects, such as constipation, urinary retention, dry mouth, blurred vision and cognitive defi cits, are particularly troublesome for elderly patients (Kindermann et al 2002). Peripheral effects include urinary retention, constipation, blurred vision, and dry mouth. Central anticholinergic effects include sedation, confusion, delirium, and cognitive decline. Elders are also more sensitive to the adverse effects related to blockade of adrenergic and histaminic receptors. Histaminic blockade causes sedation and adrenergic receptor blockade can lead to orthostatic hypotension, dizziness and syncope (which can contribute to falls) (DeVane and Mintzer 2003). Evidence also suggests that the relative affi nity at muscarinic and adrenergic receptors may affect multiple aspects of cognition, including memory and executive functions (Daniel 2000). Clinical Interventions in Aging 2007:2(4) 619 Management of BPSD An approach to management of BPSD The initial approach to the management of BPSD should always include the non-pharmacologic therapies discussed earlier. Often, pharmacologic intervention is needed. As mentioned previously, there is no consensus nor clear stan- dard of care when it comes to pharmacologic management of BPSD. We would like to offer, instead, general guidance for fi rst and second line treatment of some of the behavioral and psychological symptoms associated with dementia. Most patients with dementia present with more than one of these symptoms, and a single medication may be able to treat multiple symptoms. First, when the diagnosis of AD has been made, a cholinesterase inhibitor could be initiated, with or without memantine (in mild to moderate AD). For second line therapies, providers should use target symptoms to guide their treatment. The SSRI antidepressants citalopram and sertraline have the best evidence for their use in reducing symptoms such as irritability, sleep disturbances, and some aggression. Trazodone may also be helpful, particularly with sleep disturbances and agitation. Benzodiazepines can be effective but must be used with caution in the elderly. Finally, the atypical antipsychotics such as risperidone and olanzap- ine may be used for controlling aggression, agitation and psychotic symptoms in many different forms of dementia. We caution, however, that close monitoring be undertaken, and the medications be carefully titrated for effect. If they are not effective in controlling target symptoms, then the medications should be discontinued. Conclusion The increasing life expectancy in most Western societies along with other factors such as improved detection of dementia, administration of disease modifying agents, and improved healthcare techniques, has led to a rapidly growing number of elderly people with dementia and longer survival of these patients. Behavioral and psychological disorders occur in most dementing conditions, usually in later stages. One half of these patients experience psychotic symptoms, such as delusions and hallucinations, which in turn makes them more vulnerable to severe agitation. Treatment of these disorders in dementia should reduce the patients’ and the care- givers’ burden, resulting in lower rates of institutionalization and less psychophysical morbidity in the family (Stoppe et al 1999; Kindermann et al 2002). Caregiver education, support and behavioral training, and environmental modifi cations are important components of the management of BPSD, and should be the fi rst step in approaching the dementia patient with these symptoms. However, pharmacologic management is often needed. There are many classes of medications to choose from for treating these neuropsychiatric symptoms, but the evidence behind treatment is varied and confusing. Clinicians considering pharmacologic therapy, especially the antipsychotics, should discuss the potential risks and benefi ts of such therapy with patients and their surrogate decision makers, noting any risk factors those patients may have (for CV disease, for example). Good clinical practice dictates that patients receive individual- ized pharmacotherapeutic dosing regimens initiated and modi- fi ed relative to clinical effi cacy and tolerability and targeted to specifi c neuropsychiatric symptoms (Tariot, Profenno et al 2004). “The art of drug treatment is to use the right drug for the right symptoms at the proper stage of the disease starting low and going slow (Gauthier 2005, p 857).” References Aupperle P, Koumaras B, Chen M, et al. 2004. Long term effects of riv- astigmine treatment on neuropsychiatric and behavioral disturbances in nursing home residents with moderate to severe Alzheimer’s Disease: Results of a 52-week open-label study. Curr Med Res Opin, 20:1605–12. Ballard C, Waite J, Birks J. 2006. Atypical antipsychotics for aggression and psychosis in Alzheimer’s Disease. Cochrane Database of Systematic Reviews, (1):CD003476. Ballard C, O’Brien J, Reichelt K, et al. 2002. 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, 63:553–8. Ballard C, Grace J, McKeith I, et al. 1998. Neuroleptic sensitivity in demen- tia with Lewy bodies and Alzheimer’s disease. Lancet, 351:1032–3. Benedetti R, Cavallaro R, Smeraldi E. 1999. Olanzapine-induced neutrope- nia after clozapine-induced neutropenia. Lancet, 354:566–7. Carson S, McDonagh M, Peterson K. 2006. A systematic review of the effi cacy and safety of atypical antipsychotics in patients with psychological and behavioral symptoms of dementia. J Amer Geri Soc, 54:354–61. Cooper J. 2003. Buspirone for anxiety and agitation in dementia. J Psych Neurosci, 28:496. Daniel D. 2000. Antipsychotic treatment of psychosis and agitation in the elderly. J Clin Psych, 61(Suppl 14):49–52. De Deyn P, Katz I, Brodaty H, et al. 2005. Management of agitation, aggression, and psychosis associated with dementia: A pooled analysis including three randomized, placebo-controlled double-blind trials in nursing home residents treated with risperidone. Clin Neurol and Neurosurg, 107:497–508. De Deyn P, Wirshing W. 2001. Scales to assess effi cacy and safety of pharmacologic agents in the treatment of behavioral and psychological symptoms of dementia. J Clin Psych, 62:19–22. DeVane C, Mintzer J. 2003. Risperidone in the management of psychiatric and neurodegenerative disease in the elderly: An update. Psychopharm Bulletin, 37:116–32. Doody R, Stevens J, Beck C, et al. 2001. Practice parameter: Manage- ment of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology, 56:1154–66. Douzjian M, Wilson C, Shultz M, et al. 1998. A program to use pain control medication to reduce psychotropic drug use in residents with diffi cult behavior. Annals of Long Term Care, 6:174–9. Clinical Interventions in Aging 2007:2(4)620 Hersch and Falzgraf Eichelman B. 1987. Neurochemical basis of aggressive behavior. Psychiatric Ann, 17:371–4. Feldman H, Gauthier S, Hecker J, et al. 2001. A 24-week randomized, double-blind study of Donepezil in moderate to severe Alzheimer’s Disease. Neurology, 53:946–55. F i n k e l S , M i n t z e r J , D y s k e n M , e t a l . 2 0 0 4 . A r a n d o m i z e d , placebo-controlled study of the effi cacy and safety of sertraline in the treatment of the behavioral manifestations of Alzheimer’s Disease in outpatients treated with Donepezil. Int J Geriatr Psych, 19:9–18. Finkel S, Costa E, Silva J, et al. 1996. Behavioral and psychological signs and symptoms of dementia: A consensus statement on current knowl- edge and implications for research and treatment. Int Psychogeriatr, 8(Suppl 3):S497–500. Finkel S, Lyons J, Anderson R, et al. 1995. A randomized, placebo- controlled trial of thiothixene in agitated, demented nursing home patients. Intl Jnl Geri Psych, 10:129–36. Gauthier S. 2005. Drugs for Alzheimer’s Disease and related dementias. BMJ, 330:857–8. Gonzalez-Salvador T, Lyketsos C, Baker A, et al. 2000. Quality of life in dementia patients in long-term care. Int J Geriatr Psych, 15:181–9. Hawkins J, Tinklenberg J, Sheikh J, et al. 2000. A retrospective chart review of Gabapentin for the treatment of aggressive and agitated behavior in patients with dementias. Am J Geriatr Psychiatry, 8:221–5. Jeste D, Rockwell E, Harris M, et al. 1999. Conventional versus newer antipsychotics in elderly patients. Am J Geriatr Psych, 7:70–6. Jones P, Barnes T, Davies L, et al. 2006. Randomized controlled trial of the effect on quality of life of second vs fi rst generation antipsychotic drugs in schizophrenia. Arch Gen Psychiatry, 63:1079–87. Katz I, Jeste D, Mintzer J, et al. 1999. Comparison of risperidone and placebo for psychosis and behavioral disturbances associated with dementia: A randomized, double-blind trial. J Clin Psych, 60:107–15. Kindermann S, Dolder C, Bailey A, et al. 2002. Pharmacological treatment of psychosis and agitation in elderly patients with dementia. Drugs Aging, 19:257–76. Kozman M, Wattis J, Curran S. 2006. Pharmacological management of behavioral and psychological disturbance in dementia. Hum Psychopharmacol Clin Exp, 21:1–12. Lanctot K, Herrmann N. 2000. Donepezil for behavioral disorders associated with Lewy bodies: a case series. Int J Geriatr Psych, 15:338–45. Lane P. 2003. A pain assessment tool for people with advanced Alzheimer’s and other progressive dementias. Home Healthcare Nurse, 21:32–7. Lawlor B. 2004. Behavioral and psychological symptoms in dementia: the role of atypical antipsychotics. J Clin Psychiatry, 65(Suppl 11):5–10. Lawlor B, Radcliffe J, Molchan S, et al. 1994. A pilot placebo controlled study of trazodone and buspirone in Alzheimer’s Disease. Int J Geriatr Psych, 9:55–9. Lee P, Gill S, Freedman M, et al. 2004. Atypical antipsychotic drugs in the treatment of behavioral and psychological symptoms of dementia: systematic review. BMJ, 329:75. Lovell B, Ancoli-Israel S, Gevirtz R. 1995. Effect of bright light treatment on agitated behavior in institutionalized elderly subjects. Psychiatry Res, 57:7–12. Lyketsos C, DelCampo L, Steinberg M, et al. 2003. Treating depression in Alzheimer Disease: Effi cacy and safety of sertraline therapy, and the benefi ts of depression reduction: the DIADS. Arch Gen Psych, 60:737–46. Lyketsos C, Lopez O, Jones B, et al. 2002. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: Results from the Cardiovascular Health Study. JAMA, 288:1475–83. Lyketsos C, Steele C, Baker L, et al. 1997. Major and minor depression in Alzheimer’s Disease: Prevalence and impact. J Neuropsychiatry Clin Neurosci, 9:556–61. Maletta G. 1990. Pharmacologic treatment and management of the aggres- sive demented patient. Psychiatric Ann, 20:446–55. Mellow A, Solano-Lopez C, Davis S. 1993. Sodium valproate in the treatment of behavioral disturbance in dementia. J Geriatr Psych Neurol, 6:205–9. Miles S, Irvine P. 1992. Deaths caused by physical restraints. Gerontologist, 32:762–6. Nyth A, Gottfries C. 1990. The clinical effi cacy of citalopram in treatment of emotional disturbances in dementia disorders: A Nordic multicenter study. Br J Psych, 57:894–901. O’Brien J, Caro J. 2001. Alzheimer’s Disease and other dementia in nursing homes: levels of management and cost. Int Psychogeriatr, 13:347–58. P a l m e r C , A d a m s S , B o u r g e o i s M , e t a l . 1 9 9 9 . R e d u c t i o n i n caregiver-identifi ed problem behaviors in patients with Alzheimer Disease post-hearing aid fi tting. J Speech Lang Hear Res, 42:312–28. Paulsen J, Salmon D, Thal L, et al. 2000. Incidence of and risk factors for hallucinations and delusions in patients with probable AD. Neurology, 54:1965–71. Pautex S, Michon A, Guedira M, et al. 2006. Pain in severe dementia: self- assessment of observational scales? JAGS, 54:1040–5. Polluck B, Mulsant B, Rosen J, et al. 2002. Comparison of citalopram, perphenazine, and placebo for the acute treatment of psychosis and behavioral disturbances in hospitalized, demented patients. Am J Psychiatry, 159:460–5. Polluck B, Muylsant B, Sweet R, et al. 1997. An open pilot study of citalopram for behavioral disturbances of dementia. Am J Geriatr Psych, 5:70–8. Rosler M, Retz W, Retz-Junginger P, et al. 1999. Effects of two-year treatment with the cholinesterase inhibitor rivastigmine on behavioral symptoms in Alzheimer’s Disease. Behav Neurol, 11:211–16. Rovner B, German P, Broadhead J, et al. 1990. The prevalence and manage- ment of dementia and other psychiatric disorders in nursing homes. Int Psychogeriatr, 2:13–24. Schneider L, Dagerman K, Insel P. 2005. Risk of death with atypical antipsy- chotic drug treatment for dementia. JAMA, 294:1934–43. Schneider L, Dagerman K, Insel P. 2006. Effi cacy and adverse effects of atypical antipsychotics for dementia: Meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psych, 14:191–210. Schneider L, Tariot P, Dagerman K, et al. 2006. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s Disease. NEJM, 355:1525–38. Shankle W, Nielson K, Cotman C. 1995. Low dose propranolol reduces aggression and agitation resembling that associated with orbitofron- tal dysfunction in elderly demented. Alzheimer Dis Assoc Disord, 9:233–7. Sink K, Holden K, Yaffe K. 2005. Pharmacological treatment of neuropsy- chiatric symptoms of dementia. JAMA, 293:596–608. Sloane P, Hoeffer B, Mitchell C, et al. 2004. Effect of person-centered show- ering and the towel bath on bathing-associated aggression, agitation, and discomfort in nursing home residents with dementia: a randomized, controlled trial. J Amer Geri Soc, 52:1795–804. Smallwood J, Brown R, Coulter F, et al. 2001. Aromatherapy and behavior disturbances in dementia: A randomized controlled trial. Int J Geriatr Psych, 16:1010–13. Steele C, Rovner B, Chase B, et al. 1990. Psychiatric symptoms and nur- sing home placement of patients with Alzheimer’s Disease. Amer Jnl Psych, 147:1049–51. Stern Y, Mayeux R, Sano M, et al. 1987. Predictors of disease course in patients with probable Alzheimer’s Disease. Neurology, 37:1649–53. Stoppe G, Brandt C, Staedt J. 1999. Behavioral problems associated with dementia. The role of newer antipsychotics. Drugs and Aging, 14:41–54. Streim J. 1995. OBRA regulations and psychiatric care in the nursing home. Psychiatric Ann, 25:413–18. Sultzer D, Gray K, Gunay I, et al. 1997. A double-blind comparison of trazodone and haloperidol for treatment of agitation in patients with dementia. Am J Geriatr Psych, 5:60–9. Clinical Interventions in Aging 2007:2(4) 621 Management of BPSD Tariot P, Erb R, Leibovici A, et al. 1994. Carbamazepine treatment of agitation in nursing home patients with dementia: A preliminary study. JAGS, 42:1160–6. Tariot P, Farlow M, Grossberg G, et al. 2004. Memantine treatment in patients with moderate to severe Alzheimer’s Disease already receiving Donepezil: A randomized controlled trial. JAMA, 291:317–24. Tariot P, Profenno L, Ismail M. 2004. Effi cacy of atypical antipsychot- ics in elderly patients with dementia. J Clin Psychiatry, 65(Suppl 11):11–15. Teri L, Gibbons L, McCurry S, et al. 2003. Exercise plus behavioral manage- ment in patients with Alzheimer Disease. JAMA, 290:2015–22. Wang P, Schneeweiss S, Avorn J, et al. 2005. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. NEJM, 353:2335–41. Weiner M, Koss E, Wild K, et al. 1996. Measure of psychiatric symp- toms in Alzheimer patients: A review. Alzheimer Dis Assoc Disord, 10:20–30. Zaudig M. 2000. A risk-benefi t assessment of risperidone for the treatment of behavioral and psychological symptoms in dementia. Drug Safety, 23:183–95. << /ASCII85EncodePages false /AllowTransparency false /AutoPositionEPSFiles true /AutoRotatePages /None /Binding /Left /CalGrayProfile (Dot Gain 20%) /CalRGBProfile (sRGB IEC61966-2.1) /CalCMYKProfile (U.S. Web Coated \050SWOP\051 v2) /sRGBProfile (sRGB IEC61966-2.1) /CannotEmbedFontPolicy /Error /CompatibilityLevel 1.4 /CompressObjects /Tags /CompressPages true /ConvertImagesToIndexed true /PassThroughJPEGImages true /CreateJDFFile false /CreateJobTicket false /DefaultRenderingIntent /Default /DetectBlends true /ColorConversionStrategy /LeaveColorUnchanged /DoThumbnails false /EmbedAllFonts true /EmbedJobOptions true /DSCReportingLevel 0 /EmitDSCWarnings false /EndPage -1 /ImageMemory 1048576 /LockDistillerParams false /MaxSubsetPct 100 /Optimize true /OPM 1 /ParseDSCComments true /ParseDSCCommentsForDocInfo true /PreserveCopyPage true /PreserveEPSInfo true /PreserveHalftoneInfo false /PreserveOPIComments false /PreserveOverprintSettings true /StartPage 1 /SubsetFonts true /TransferFunctionInfo /Apply /UCRandBGInfo /Preserve /UsePrologue false /ColorSettingsFile () /AlwaysEmbed [ true ] /NeverEmbed [ true ] /AntiAliasColorImages false /DownsampleColorImages true /ColorImageDownsampleType /Bicubic /ColorImageResolution 300 /ColorImageDepth -1 /ColorImageDownsampleThreshold 1.50000 /EncodeColorImages true /ColorImageFilter /DCTEncode /AutoFilterColorImages true /ColorImageAutoFilterStrategy /JPEG /ColorACSImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /ColorImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /JPEG2000ColorACSImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /JPEG2000ColorImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /AntiAliasGrayImages false /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /GrayImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /JPEG2000GrayACSImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /JPEG2000GrayImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /AntiAliasMonoImages false /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 1200 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict << /K -1 >> /AllowPSXObjects false /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile () /PDFXOutputCondition () /PDFXRegistryName (http://www.color.org) /PDFXTrapped /Unknown /Description << /ENU (Use these settings to create PDF documents with higher image resolution for high quality pre-press printing. The PDF documents can be opened with Acrobat and Reader 5.0 and later. These settings require font embedding.) /JPN /FRA /DEU /PTB /DAN /NLD /ESP /SUO /ITA /NOR /SVE >> >> setdistillerparams << /HWResolution [2400 2400] /PageSize [612.000 792.000] >> setpagedevice