83
Views
21
CrossRef citations to date
0
Altmetric
Review

Treatment of Alzheimer’s disease across the spectrum of severity

&
Pages 131-142 | Published online: 18 Oct 2022

Abstract

Alzheimer’s disease (AD) is the most common cause of dementia affecting nearly 18 million people around the world and 4.5 million in the US. It is a progressive neurodegenerative condition that is estimated to dramatically increase in prevalence as the elderly population continues to grow. As the cognitive and neuropsychiatric signs and symptoms of AD progresses in severity over time, affected individuals become increasingly dependent on others for assistance in performing all activities of daily living. The burden of caring for someone affected by the disorder is great and has substantial impact on a family’s emotional, social and financial well-being. In the US, the currently approved medications for the treatment of mild to moderate stages of AD are the cholinesterase inhibitors (ChEIs). Cholinesterase inhibitors have shown modest efficacy in terms of symptomatic improvement and stabilization for periods generally ranging from 6 to 12 months. There are additional data that have emerged, which suggest longer-term benefits. For the moderate to severe stages of AD, memantine, an N-methyl-D-aspartate (NMDA) receptor antagonist is in widespread use and has shown modest benefit as monotherapy and in combination with ChEIs. The cost effectiveness of the currently available therapeutic agents for AD has undergone great scrutiny and remains controversial, especially outside the US. Neuropsychiatric symptoms such as agitation and psychosis are common in AD. Unfortunately, in the US there are no Food and Drug Administration (FDA)-approved agents for the treatment of these symptoms, although atypical antipsychotics have shown some efficacy and have been widely used. However, the use of these agents has recently warranted special caution due to reports of associated adverse effects such as weight gain, hyperlipidemia, glucose intolerance, cerebrovascular events, and an increased risk for death. Alternative agents used to treat neuropsychiatric symptoms include serotonergic antidepressants, benzodiazepines, and anticonvulsant medications.

Introduction

Alzheimer’s disease (AD) is an acquired neurodegenerative disease that causes persistent and progressively severe memory loss accompanied by other cognitive deficits that include aphasia, apraxia, agnosia, dyscalculia, executive function impairment, and changes in behavior. These clinical features have a major impact on the affected individual’s ability to perform the activities of daily living. As AD progresses in severity across its different clinical stages, the affected patient becomes increasingly more dependent on others for care. As the most common cause of dementia, AD exerts a substantial toll on an aging population worldwide and is of great public health interest. The neuropathological hallmarks of AD include amyloid plaques, neurofibrillary tangles, inflammation, neuronal loss, and depletion of neurochemicals such as acetylcholine (CitationWhitehouse et al 1982). Alterations in other neurotransmitter systems such as the glutamatergic system have also been identified as contributory to the pathogenesis of the disease.

There is no cure for AD. Current treatment of the disorder involves the use of medications to obtain symptomatic cognitive, behavioral, and functional improvement or stabilization. In addition, individualization of the treatment plan to address medical co-morbidities, the social and financial impact of the disease on the family, and caregiver well-being is vital to achieving successful outcomes throughout all clinical stages of AD.

Treatment of cognition and function

Mild cognitive impairment

Alzheimer disease is a gradually progressive neurodegenerative disorder whose neuropathological alterations may precede the onset of clinical symptoms by decades. Mild cognitive impairment (MCI) is considered by some to represent the earliest clinical manifestations of impending dementia. In MCI, the affected individual subjectively complains of memory problems but has generally normal cognitive and daily functioning despite a demonstrable decrease in memory performance on testing (CitationPeterson et al 2001). Mild cognitive impairment has been described as a transitional state between healthy cognitive aging and overt dementia caused by AD, vascular dementia, or other causes of progressive cognitive decline in the elderly. Approximately 10% to 15% of individuals with MCI progress to diagnosed AD each year. There is no treatment approved for MCI. CitationPeterson and colleagues (2005) have reported on the use of donepezil and vitamin E in patients with MCI in a randomized double-blind, placebo-controlled study. They found that compared with placebo, at 18 months donepezil delayed the onset of AD by 6 months, however, donepezil had no beneficial effects in preventing the onset of AD by the end of the 36 month trial. Vitamin E was not shown to be of significant benefit.

Mild to moderate AD

Cholinesterase inhibitors

Available consensus treatment guidelines and practice parameters, including those of the American Academy of Neurology (AAN) recommend the use of cholinesterase inhibitors (ChEIs) as standard therapy for mild to moderate AD based on evidence accumulated from several 3- to 6-month, randomized, double-blind, placebo-controlled clinical trials (CitationDoody et al 2001). These studies suggest a modest therapeutic effect of this class of medications on the symptoms of AD. The primary outcome measures used across these studies most often include the Alzheimer’s Disease Assessment Scale – cognitive subscale (ADAS-cog) (CitationRosen et al 1984) and the clinician’s interview-based impression of change plus caregiver input (CIBIC-Plus) (CitationSchneider et al 1997). The ADAS-cog is a highly sensitive scale that measures multiple cognitive outcomes. Scores range from 0 to 70 (high to low cognitive functioning). Untreated patients with mild to moderate AD typically progress by a 3 to 4 point increase over 6 months. Patients with moderate to severe AD have an increase of 4 to 6 points over 6 months if untreated. The CIBIC-Plus is a tool that assesses patient functioning via a subjective examination of the patient and an interview with a caregiver (range of scores 1 to 7; score of 1 indicating substantial improvement and 7 indicating marked deterioration). The pivotal 12 to 24 week duration clinical trials examining the efficacy of ChEIs in AD have shown improvement in study subjects on the ADAS-cog of 2.5 to 3.5 points and on the CIBIC-Plus of 0.3 to 0.5 points as compared with deterioration in placebo-treated subjects (CitationCummings 2004).

Donepezil

Donepezil is a rapidly reversible inhibitor of acetylcholinesterase. Subjects treated with a daily dose of donepezil 5–10 mg, in several 12 to 24 week placebo-controlled trials of patients with mild to moderate probable AD (n=1759), demonstrated statistically significant differences on the ADAS-cog and the CIBIC-Plus scales favoring the active drug (CitationRogers, Doody, et al 1998; CitationRogers, Farlow, et al 1998; CitationBurns et al 1999). During single-blind, 6 week placebo washout periods, ADAS-cog and CIBIC-Plus scores in the donepezil-treated groups reverted to levels similar to those in the placebo group (CitationRogers et al 1998; CitationBurns et al 1999). This has been interpreted to suggest that donepezil treatment for 6 months has a symptomatic effect, but not a “disease modifying” effect.

Two double-blind, 12 month placebo-controlled trials of donepezil with 10 mg daily have been completed in patients with mild to moderate AD (CitationWinblad et al 2001; CitationMohs et al 2001). Winblad and colleagues reported a difference in favor of donepezil over placebo using the Gottfries-Brane-Steen scale (GBS) (CitationBrane et al 2001) as the primary outcome measure. The GBS scale is a measure of global functioning. This scale assesses patient function in four general areas using a semi-structured interview by a clinician with a patient and caregiver. It measures changes in the symptoms of dementia over a specified amount of time. Overall, GBS scale scores for subjects (observed cases) in the active treatment group showed a decline half as large as that reported for patients in the placebo group. Mohs and colleagues reported that the median amount of time to clinically evident functional decline as determined by several measures of activities of daily living was delayed by 5 months in the donepezil treated group compared with the placebo group ().

Table 1 Summary of major clinical trials of donepezil

In a Cochrane Library review of the efficacy of donepezil for mild to moderate AD, 4 studies were included covering treatment of 12 or 24 weeks duration in highly selected patients. The authors concluded that in selected patients with mild to moderate AD treated for 12 or 24 weeks, donepezil produced modest improvements in cognitive function and no improvements were noted on patient self-assessed quality of life. In addition, the authors noted significantly higher number of withdrawals before the end of treatment from the 10 mg/day donepezil group compared with placebo, which could have potentially resulted in overestimation of the beneficial effects at 10 mg/day, as last available measures were used in the analyses. It is not known whether these gains translate into gains for the caregiver or in substantial changes in the life of the patient (CitationBirks et al 2000). A recent study on the long-term use of donepezil has attempted to address the controversies regarding the practical effectiveness of the agent from a cost–benefit analysis. Courtney and co-workers (CitationCourtney et al 2004) studied 565 community residing subjects with mild to moderate AD. The subjects were referred from memory clinics and clinically diagnosed with AD with or without cerebrovascular disease. They were initially randomly assigned to treatment with donepezil 5 mg/day or placebo during a 12-week run-in period to assess initial effects. 486 subjects who completed this phase of the study were then re-randomized to receive either donepezil or placebo, with double blind treatment continuing for as long as “judged appropriate”. At the end of 48 weeks of treatment, a 6 week washout period was commenced. Patients could then subsequently choose to continue with the same double-blinded treatment that they had been on for a further 48 weeks. At the end of every 48 week treatment period a further 4-week washout occurred, after which patients could again continue on for another 48 weeks. Primary endpoints were entry to institutionalized care and progression of disability in activities of daily living. The Mini-Mental State Examination (MMSE) (CitationFolstein et al 1975), a widely used screening test for cognitive impairment in older adults was also used in this study as an outcome measure. The donepezil-treated group improved from baseline by 0.9 MMSE points during the first 12 weeks, however, no change was seen in the placebo group. After the 12th week, both groups declined at similar rates. Over the first two years, donepezil-treated subjects showed some advantage over placebo-treated subjects in MMSE scores (0.8 points) and functional ability. At three years, no significant difference was noted between the donepezil-treated group and the placebo-treated group in rates of institutionalization or progression of disability. The study also did not detect any significant difference between donepezil and placebo on the treatment of behavioral symptoms. There were no significant differences between donepezil and placebo in caregiver well-being, time of caregiving, and costs of care. However, the study confirmed the same degree of modest efficacy of donepezil in treating cognition as did the pivotal trials of shorter duration. The authors concluded that donepezil is not a cost effective treatment for AD.

CitationRelkin and colleagues (2003) conducted an open-label trial of donepezil in a large community-based population. They included 1035 patients with mild to moderate AD, out of which 894 completed the trial. Nearly all patients had at least one co-morbid medical condition or were taking a minimum of one concomitant medication. Efficacy was measured using the standardized MMSE (sMMSE) (CitationMolloy et al 1991). The goal of the sMMSE is to impose clear and explicit guidelines for administration and scoring to improve the reliability of the instrument. The sMMSE has significantly better inter-rater and intra-rater reliability compared with the MMSE. Over the initial 12 week study period the sMMSE increased by 1.54 points over baseline. Donepezil was shown to be well-tolerated with the occurrence of adverse events significantly lower after a dose increase at 4 weeks as compared with a dose increase after 1 week as in previous trials. In addition, the investigators found that concomitant medication use such as aspirin or nonsteroidal antiinflammatory drugs did not increase the risk of gastrointestinal side effects with use of donepezil. Concomitant medications such as digoxin, calcium channel blockers, or beta blockers did not increase the risk of bradycardia, a potential side-effect of cholinergic medications.

CitationSeltzer and colleagues (2004) studied the efficacy of donepezil in mild AD in subjects with MMSE scores of 21 to 26. In a double blind fashion, subjects were randomized to receive either donepezil or placebo in a 2:1 ratio for 24 weeks. Donepezil-treated subjects performed better than placebo-treated subjects on the ADAS-cog and MMSE at all time points studied.

None of the ChEIs have been approved for treatment of the severe stages of AD. However, in a 6 month placebo-controlled trial of donepezil in moderate to severe AD, donepezil was shown to be of benefit (CitationFeldman et al 2001). At 6 months, 63% of moderate to severely affected patients in the donepezil-treated group versus 42% of the placebo-treated group showed improvement or no change in CIBIC-Plus scores.

Based on the results from the pivotal double-blind, randomized, placebo-controlled clinical trials in patients with mild to moderate AD, the US Food and Drug Administration (FDA) has approved use of donepezil at doses of 5 mg to 10 mg once daily. The starting dose of donepezil is 5 mg once daily, with an increase to 10 mg recommended at 4 to 6 weeks.

Rivastigmine

Rivastigmine inhibits acetylcholinesterase and butyryl-cholinesterase (CitationBallard 2002). In two 26-week trials involving patients with mild to moderate AD (n=1424), patients receiving a daily dose of 6 mg to 12 mg of rivastigmine demonstrated favorable and significant differences in ADAS-cog and CIBIC-Plus scores as compared with the placebo-treated group (CitationCorey-Bloom et al 1998; CitationRosler et al 1999) ().

Table 2 Summary of major clinical trials of rivastigmine

Sustained benefits of rivastigmine have been reported for over a 1-year period by CitationFarlow and colleagues (2000). After receiving double-blind placebo or rivastigmine at daily doses of 1 mg to 4 mg or 6 mg to 12 mg for 26 weeks, patients were eligible to enter an open label extension phase. The withdrawal rate for subjects treated with rivastigmine 6 mg to 12 mg with more rapid dose titration in the initial double-blind phase of the study was 35% (CitationCorey-Bloom et al 1998). For the open label extension phase, dose titration was more flexible and the rate of withdrawal was decreased to 19% (CitationFarlow et al 2000). At 52 weeks, a large difference (5.7 points) was seen in ADAS-cog scores favoring the rivastigmine-treated group over the placebo group (CitationFarlow et al 2000). Although impairment advanced in both groups, subjects originally receiving placebo for the initial 6 months of the study did not show the same amount of benefit as subjects treated with effective doses of rivastigmine for the entire 52 weeks. This observation suggests the benefit of starting rivastigmine treatment early in the course of the illness.

There is some evidence from a retrospective analysis of rivastigmine therapy that it might have beneficial effects in moderately severe AD (CitationBurns et al 2004). Data were pooled from three 6-month randomized, placebo-controlled, double-blind trials. Patients with severe cognitive impairment were identified if they had MMSE scores of 10 to 12. 112 patients met the inclusion criteria and had received placebo or daily rivastigmine, 6 mg to 12 mg. After 6 months, subjects in the rivastigmine group showed a 0.2 point decrease in mean ADAS-cog scores (decreased impairment) compared with baseline, whereas placebo group subjects had an increase of 6.3 points (increased impairment) (p<0.001). Clinical improvement was also observed with the MMSE, Progressive Deterioration Scale (PDS), and the Behavioral Pathology in Alzheimer’s Disease Rating Scale (BEHAVEAD).

The FDA has approved rivastigmine (like all other available ChEIs) for mild to moderate AD. The dose of rivastigmine increases from 1.5 mg twice daily to 3 mg twice daily, then to 4.5 mg twice daily, and to a maximum of 6 mg twice daily. Dose increments are recommended at 1 to 4 week intervals; fewer side effects emerge when the intervals are longer.

Galantamine

Galantamine has been proposed to both inhibit acetylcholinesterase and to act as an allosteric potentiating ligand on nicotinic acetylcholine receptors (CitationMaelicke et al 2001). Several large 5- to 6-month placebo-controlled trials for patients with mild to moderate AD (n = 2267) have found beneficial drug effects on the ADAS-cog and CIBIC-Plus scales for patients treated with galantamine, 16 mg to 24 mg per day versus placebo ().

Table 3 Summary of major clinical trials of galantamine

There is some evidence of longer-term benefits of galantamine in 12-month open label extension studies (CitationRaskind et al 2000). After 12 months of therapy, patients who received 24 mg per day of galantamine had ADAS-cog scores that remained stable relative to baseline measures. Comparatively, the placebo group had increased impairment in ADAS-cog scores (increase of 4 to 5 points). The group that received galantamine throughout the 12-month study period had better ADAS-cog outcomes at endpoint than did patients who received placebo during the first 6 months of the study and were then switched to galantamine. All findings were observed case (OC) analyses which were confirmed with intent to treat analyses (ITT) using the last observation carried forward method (LOCF). CitationLyketsos and colleagues (2004) assessed the long-term efficacy as well as tolerability of galantamine. The authors conducted a 12-month open-label extension of an earlier 5-month, double-blind, placebo-controlled trial with a 6-week withdrawal phase. The authors concluded that patients treated with galantamine continuously throughout the double-blind and open-label phases (n=288) showed sustained cognitive benefits at 18.5 months. In addition, the safety and tolerability of galantamine was comparable with other large studies of the drug.

CitationBlesa and colleagues (2003) investigated the benefits of galantamine in patients with more severe disease by performing a post-hoc analysis using data collected from two long-term galantamine studies. Patients included in this study were those with MMSE scores <14, or ADAS-cog subscale scores of >30. The authors concluded that galantamine offered both cognitive and functional benefit in this population. Galantamine is approved by the FDA for mild to moderate AD. It is initiated at a dose of 4 mg twice daily for 1 month. The dose is increased to 8 mg twice daily and if desired, up to 12 mg twice daily. Longer intervals between dosage increases are associated with a lower incidence of gastrointestinal side effects. Recently, galantamine was introduced in an extended release form for once per day dosing. The starting dose is 8 mg per day for one month followed by titration up to 16 mg or 24 mg per day.

Despite the postulated differences in mechanisms of action of the ChEIs, all three drugs (donepezil, rivastigmine, and galantamine) have similar degrees of efficacy. This was demonstrated by CitationRitchie and colleagues (2004) who conducted a meta-analysis of randomized trials of the efficacy and safety of these three agents. The authors conducted regression analyses to compare the effect of dose on clinical outcomes and completion rates, analyzing ten donepezil, six galantamine, and five rivastigmine studies. All three drugs showed beneficial effects on cognitive tests, as compared with placebo. For donepezil and rivastigmine, larger doses were associated with larger symptomatic effects as assessed by the ADAS-cog. Clinical global improvement was shown to be superior for each drug over placebo with no dose effects noted.

The optimal duration of treatment with ChEIs is uncertain. The duration of most blinded trials has been 6 months. Trials lasting one year have shown a sustained difference in efficacy between actively treated groups of patients and patients receiving placebo (CitationRaskind et al 2000; CitationWinblad et al 2001). In addition, studies in which the rate of deterioration in the placebo group was extrapolated and compared with the level of function of patients continuing treatment with a ChEI suggest that patients continue to derive modest benefit from treatment for two to three years (CitationRogers et al 2000).

Although the ChEIs as a class are generally well tolerated, the commonest adverse effects noted in the pivotal trials include nausea, vomiting, diarrhea, loss of appetite, loss of weight, muscle cramps, and insomnia. The dosing recommendation provided with these agents is to have them administered with a meal to lower the risk of side effects. As a class, ChEIs are contraindicated in the presence of cardiac conduction abnormalities such as left bundle branch block and sick sinus syndrome as well as gastric ulcer disease in which bleeding has occurred. From a comparative tolerability perspective, data from the pivotal clinical trials of all three agents suggest that rivastigmine is associated with the greatest risk of gastrointestinal side-effects (nausea, emesis, diarrhea) followed by galantamine and then donepezil. A more gradual dose titration of rivastigmine than initially recommended with the use of this agent appears to attenuate the frequency of these symptoms. Donepezil has been more often associated with muscles cramps and sleep disturbance than the other two agents in the class.

Moderate to severe AD

Memantine

Memantine was approved by the FDA in October 2003 for the treatment of moderate to severe AD having been previously widely available in Germany for several years. Memantine is an N-methyl-D-aspartate (NMDA) receptor noncompetitive antagonist that is proposed to lessen the neurotoxic effects of excessive excitatory glutamatergic neurotransmission that occurs in AD. To assess the impact of treatment, pivotal trials of memantine for moderate to severe AD have used the Severe Impairment Battery (SIB) to evaluate abilities to perform basic cognitive tasks (CitationPanisset et al 1994), the Alzheimer’s disease cooperative study – activities of daily living scale (ADCS–ADL) (CitationGalasko et al 1997); and the CIBIC-Plus scale ().

Table 4 Summary of major clinical trials of memantine

As noted in , results from the trial by Reisberg and colleagues indicated benefit of memantine treatment on cognitive and functional measures. As measured by the SIB, cognition was stable for the first 12 weeks and the extent of impairment in the memantine-treated group at end point was significantly less than that in the placebo treated group (−4.0 vs −10.1). Memantine treatment also delayed decline in functional ability versus placebo (ADCS–ADL scores: −3.1 with memantine vs −5.2 with placebo).

CitationTariot and colleagues (2004) reports results from a double-blind combination study in which moderate to severely impaired AD subjects were randomized to receive donepezil plus placebo (monotherapy) or donepezil plus memantine. The investigators reported that combination therapy with donepezil and memantine had a greater influence on cognitive improvement, reduced decline in activities of daily living, and a reduced frequency of new behavioral symptoms versus the donepezil monotherapy group.

In another study conducted by CitationWinblad and Poritis (1999), patients with severe dementia living in a nursing home setting or a single psychiatric hospital were recruited to assess the clinical efficacy and safety of memantine during a 12 week trial. Patients with AD or vascular dementia with a MMSE score of less than 10 were included. It was a double-blind, parallel-group comparison study utilizing memantine 5 mg/day during the first week and 10 mg/day during the next 11 weeks or placebo. Notably, this dosing is in contrast to 10 mg twice per day utilized in the pivotal trials reported by Reisberg and colleagues and Tariot and colleagues. Outcome scales used in this study included the Clinical Global Impression of change (GCI-C) (CitationNIMH 1986) and the Behavioral Rating scale for Geriatric Patients (BGP) subscore ‘care dependence’, (Citationvan de Kam et al 1971). The GCI-C is a 7-point global rating scale related to the CIBIC-Plus. The BGP is an observer-rated scale for the assessment of functional and behavioral disturbances in geriatric patients. The investigators reported that a positive response was seen in 73% of the memantine-treated subjects versus 45% of the placebo-treated patients on the CGI-C. As measured by the BGP, memantine treatment improved functioning and behavioral symptoms to a statistically significant greater extent than placebo

Adverse effects with memantine are uncommon, however, they include headache, sedation, fatigue, constipation, increased confusion, irritability, and agitation. The recommended starting dose is 5 mg once daily, with a gradual dose titration that increases the daily dose in 5 mg increments each week to reach a maximum dose of 10 mg twice daily. Although memantine is approved for use in moderate to severe AD, there is preliminary published evidence to suggest that memantine may be effective for mild to moderate AD (CitationPeskind et al 2004). Peskind and colleagues studied memantine in a 24 week placebo-controlled, randomized trial (n= 403, baseline MMSE score of 10 to 22) and noted significant improvement in cognition and observed function. Two additional unpublished studies of memantine in mild to moderate AD, a monotherapy study conducted in Europe and a combination study with a ChEI conducted in the US, failed to show statistically significant benefits over placebo in the primary outcome measures. Despite the one positive study cited above, the FDA recently denied a supplemental new drug application to broaden the agent’s indication to mild to moderate AD.

The cost-effectiveness of AD treatments has been questioned. Specifically, the use of ChEIs has been recently challenged in the UK by the National Institute for Clinical Excellence (NICE). NICE proposed in their preliminary guideline to withdraw ChEIs and memantine from the UK National Health Service (NHS) (CitationKmietowicz 2005). The NICE assessment group has indicated that there is insufficient evidence that acetylcholinesterase inhibitors have measurable effects on quality of life and time to admission to nursing home care. This proposal has caused significant international concern. The NICE guideline does not adequately take into consideration the effect of treatment on caregivers who bear a significant care burden. In addition, the NICE recommendation does not consider the wishes of the individual patient and caregiver, denying them a chance to benefit from consistently proven effective therapies.

Treatment of neuropsychiatric symptoms

Neuropsychiatric symptoms associated with dementia are common and have been reported in more than 80% of subjects in most studies (CitationCummings 2004). These features of the disorder contribute to poor outcomes for patients and caregivers. There is some controversy about the frequency of these symptoms in patients with varying severity of dementia, although psychotic symptoms seem to be more common in advanced dementia (CitationLyketsos et al 2000). CitationLyketsos and colleagues (2000) reported findings from a study of 5092 community residents who represented 90% of the elderly population of Cache County in Utah, US. Several disturbances (delusions, anxiety, apathy, irritability, elation, and disinhibition) were reported with similar severity at all stages of dementia. In contrast, aggression/agitation and aberrant motor behavior were more common at later stages of AD. The study also identified a slightly increased occurrence of depression and hallucinations in moderately severe dementia as compared with mild stage dementia. As behavior disturbances emerge in individuals with dementia, they should be managed initially by nonpharmacologic interventions to avoid the potential adverse effects of psychotropic agents (CitationCohen-Mansfield 2001). A wide spectrum of nonpharmacologic interventions have been studied for the treatment of behavior disturbances in Alzheimer’s disease (CitationCohen-Mansfield 2001). These include music, videotapes of family members, audiotapes of the voices of caregivers, walking, exercise, and sensory stimulation.

In behaviorally disturbed demented patients, few studies of the psychotropic effects of the ChEIs and memantine have been published. However, in some of the pivotal trials of the ChEIs and memantine, the data suggest that treatment with active drug is associated with less behavioral deterioration over time than placebo treatment. CitationHolmes and co-workers (2004) conducted a 12 week, randomized withdrawal study in 134 patients with donepezil. Patients with mild to moderate stage AD with neuropsychiatric symptoms as indicated by a baseline Neuropsychiatric Inventory (NPI) scale of more than 11 points were treated with donepezil 5 mg daily for 6 weeks, followed by 10 mg daily for a further 6 weeks. Patients were then randomized to either placebo or 10 mg donepezil daily. The patients were assessed using the NPI and NPI-caregiver distress (NPI-D) (CitationCummings et al 1994) scales at 6 weeks and then again at 12 weeks. The NPI is a caregiver-based interview to assess 10 behavioral disturbances: delusions, hallucinations, euphoria, anxiety, agitation, disinhibition, irritability, apathy, and aberrant motor behavior. Frequency is rated from 1 (occasional, less than once per week) to 4 (very frequent, daily or continuous). Severity is rated from 1 (mild) to 3 (severe). Total score ranges from 1 to 120 points. The NPI-D scale assesses the caregiver distress caused by the individual behaviors. Total scores range from 0 to 50 points. In the randomization phase as well as the open label phase of the study, there was improvement in the NPI and NPI-D scores in the donepezil-treated group. For patients who were on donepezil in the first 6 weeks and later randomized to placebo, there was a significant worsening of neuropsychiatric symptoms and caregiver distress at 6 and 12 weeks.

CitationOlin and colleagues (2002) have reported in a Cochrane review on 2 randomized controlled trials of galantamine for AD that included the NPI as an outcome measure. In one trial, there was no benefit of galantamine 24 mg/day or 32 mg/day treatment on behavioral symptoms versus placebo treatment. In the second trial, the 16 mg/day dose was found to be significantly better than placebo. Two randomized-controlled trials with memantine for the treatment of moderate to severe stage AD have included neuropsychiatric symptom measures as outcomes. CitationReisberg and colleagues (2003) reported that the memantine-treated group was not significantly different than the placebo-treated group in terms of NPI scores. CitationTariot and colleagues (2004) reported a statistically significant difference in favor of the memantine/donepezil-treated group versus the donepezil/placebo-treated group in terms of the NPI scores. However, the magnitude of this difference was small.

Only a few placebo-controlled clinical trials have addressed the treatment of depression in patients with AD. The most abundant data exist for sertraline and citalopram in which the former agent has shown some efficacy for depressive symptoms and the latter, depression, agitation, and lability ().

Table 5 Summary of antidepressant trials in elderly dementia patients

Risperidone, olanzapine, and quetiapine are frequently used atypical antipsychotic agents for the treatment of psychosis and agitation in dementia. There are several open label trials reporting the efficacy of these agents and a limited number of placebo-controlled studies ().

Table 6 Overview of placebo-controlled trials of atypical antipsychotics in dementia

Studies of the efficacy and tolerability of two of the newer atypical antipsychotics, aripiprazole and ziprazidone in dementia with behavior disturbance, have not yet appeared in the peer-reviewed literature. The available evidence suggests that overall, atypical antipsychotic agents are better tolerated than conventional neuroleptics and more efficacious than placebo in decreasing agitation in patients with dementia. Importantly, it has been recently reported that medications in this class may increase the risk of glucose intolerance and cerebrovascular adverse events. Clozapine is occasionally recommended for treatment refractory elderly patients with movement disorders such as Parkinsonism or tardive dyskinesia and psychosis. One of the major drawbacks with clozapine use is a significantly increased risk of agranulocytosis and the need for standardized blood monitoring (CitationKasckow et al 2004).

In April 2005, the FDA requested that all manufacturers of atypical antipsychotic medications add a “black box warning” to their prescribing information regarding the use of these medications in elderly patients with dementia-related psychosis (CitationFDA 2005). According to the FDA, “analyses of seventeen placebo-controlled trials that enrolled 5106 elderly patients with dementia-related behavior disturbances revealed a risk of death in the drug-treated patients of 1.6 to 1.7 times that seen in placebo-treated patients”. The data reviewed were gathered from dementia trials of olanzapine, aripiprazole, risperidone, and quetiapine. Over the course of these trials, averaging about 10 weeks, the rate of death in drug treated patients was about 4.5% compared with a rate of 2.6 % in the placebo group. The causes of death varied, appearing to be either cardiovascular or infectious in origin. The black box warning further goes on to state that the FDA has not approved aripiprazole, quetiapine, ziprazidone, risperidone, and olanzapine for the treatment of patients with dementia-related psychosis. Given the black box warning as well as increased cardiovascular risks (stroke), metabolic side effects such as weight gain, hyperlipidemia, insulin resistance, and diabetes, the clinician should very carefully consider the choice of pharmacological agent for the treatment of dementia-related psychosis and other neuropsychiatric symptoms. Before prescribing an atypical antipsychotic in dementia care, one should be able to identify target symptoms of psychosis such as delusional thinking and hallucinations. Only in the presence of sustained patient distress, danger to self or others, or impairment of functioning (resistance to care) is the use of atypical antipsychotic medications recommended.

In addition to the emergence of atypical antipsychotic agents as commonly used agents for the treatment of dementia-associated neuropsychiatric symptoms, the use of anticonvulsant medications such as carbamazepine and sodium valproate has commanded significant attention. One placebo-controlled study using an average dose of 300 mg per day of carbamazepine showed that the drug was well tolerated by patients and significantly decreased aggression and agitation (CitationTariot et al 1998). Potential side effects of carbamazepine include drowsiness, gastrointestinal distress, ataxia, rash, elevated hepatic enzymes, and drug interactions with other agents. This agent has also been associated with aplastic anemia and agranulocytosis.

Sodium valproate has been reportedly effective in the treatment of behavioral disturbances in dementia and has fewer drug interactions than carbamazepine. CitationPorsteinsson and colleagues (2001) reported on the use of sodium valproate in a randomized placebo-controlled six week study of 56 nursing home patients with dementia and agitation. The average dose of divalproex sodium used was 840 mg per day. The authors noted that their results suggested improvement in the agitation subscale of the Brief Psychiatric Rating Scale (BPRS). Potential side effects of this agent include nausea, vomiting, sedation, diarrhea, ataxia, and tremor. Hepatotoxicity and pancreatitis have been noted to be rare complications of the use of sodium valproate. When this medication is administered, complete blood counts, hepatic enzymes, and serum drug concentrations need to be monitored regularly to prevent toxicity.

Gabapentin, lamotrigine, and topiramate are newer anticonvulsants whose efficacy and tolerability have not been systematically evaluated for efficacy and tolerability in the treatment of dementia.

Benzodiazepines such as lorazepam and oxazepam are often used in the management of agitation, but their efficacy has not been comprehensively studied. These agents are generally safe when used in low doses, but with increasing doses, common side effects include over-sedation, ataxia, confusion, and paradoxical agitation. In addition, with long-term use, tolerance and dependence is likely. Zolpidem, a hypnotic, was shown to reduce nighttime wandering in two dementia patients (CitationShelton et al 1997).

The antianxiety agent buspirone has shown efficacy in case reports in reducing anxiety, aggression, and agitation in patients with dementia (CitationSakauye et al 1993). The drug is generally well-tolerated, however, if used in combination with selective serotonin reuptake inhibitors (SSRIs), the patient is at increased risk for the development of serotonin syndrome (confusion, tremors, hyperthermia, hypertension, and seizure).

In a recent review of the pharmacological treatment of neuropsychiatric symptoms of dementia, CitationSink and colleagues (2005) conducted a systematic review of English language articles published from 1996 to 2004. They utilized Medline, the Cochrane database of systematic reviews and a manual search of bibliographies for this review. Only double-blind, placebo-controlled, randomized control trials (RCT) or meta-analyses of dementia trials reporting effects on neuropsychiatric symptoms were included. Twenty-nine articles met inclusion criteria. Five trials of antidepressants were included. No efficacy was demonstrated for symptoms other than depression, except one study of citalopram. For mood stabilizers, three trials of sodium valproate showed no efficacy. Two trials of carbamazepine showed mixed results. Four studies of typical antipsychotics showed small benefit and no differences were shown among specific agents. Six trials of atypical antipsychotics were included. Results indicated modest, statistically significant, efficacy of olanzapine and risperidone. Two meta-analyses and six randomized clinical trials of ChEIs showed small, statistically significant, efficacy. Two randomized clinical trials of memantine had mixed results for the treatment of neuropsychiatric symptoms.

Prior to the recent FDA warning regarding the use of atypical antipsychotic agents in elderly patients with dementia, expert consensus guidelines were published to address the treatment of agitation in dementia (CitationAlexopoulos et al 2005). The guidelines recommend initiating therapy with risperidone at low doses of 0.25 mg to 0.5 mg per day with an average maximum dose of 1 mg to 1.5 mg per day if there is evidence of psychosis. These guidelines also recommend the use of environmental interventions in addition to medication treatment. If agitation is due to depression and/or anxiety, an SSRI trial is the first recommended option. If insomnia is a contributing factor, trazodone use is indicated. Other consensus guidelines (CitationAGS–AAGP 2003) recommend that nonpharmacological interventions alone are indicated if there is no threat to the patient or others, medical conditions are being addressed, and if no psychotic symptoms are evident.

At present, no psychotropic agent presently available within the US is FDA-approved for use in dementia. Atypical antipsychotics are the first line agents of choice for severe behavioral symptoms with psychosis. When medications are used to manage behavior disturbances, one must document the specific target symptoms being addressed. Combination therapy is indicated only after two different trials with two different classes of agents have proven unsuccessful. Taper or discontinuation of medications should be attempted and documented by six months following response and every six months, thereafter. There is no consensus on the choice of medication for nonpsychotic behavioral symptoms. There are no comparison studies between pharmacological and nonpharmacological interventions, which makes it difficult to prioritize a specific approach for most nonpsychotic behavioral symptoms ().

Table 7 Commonly prescribed drugs, dosage guidelines in the elderly

Conclusions

ChEIs are the mainstay of treatment for mild to moderate AD and memantine is indicated as therapy for moderate to severe AD. The degree of efficacy of these agents in improving or stabilizing cognition and function is modest, but has been consistently demonstrated across numerous clinical trials. Some controversy exists regarding whether the putative benefits of these agents outweigh their expense. As AD advances, neuropsychiatric symptoms contribute substantially to caregiver distress and are especially difficult to manage. Preliminary data suggest that memantine and ChEIs may reduce the severity or delay the emergence of neuropsychiatric symptoms. Despite their recent association with an increased risk of death, cerebrovascular adverse events, and metabolic alterations such as hyperglycemia and hyperlipidemia, atypical antipsychotics are widely used in treating psychosis and agitation. Although supporting data are limited, serotonergic antidepressants and anticonvulsant agents are frequently used for the treatment of neuropsychiatric symptoms such as depression, agitation, and aggression.

Tables

References

  • [AGS–AAGP] American Geriatrics Society and American Association for Geriatric Psychiatry2003Consensus statement on improving the quality of mental health care in U.S. nursing homes: Management of depression and behavioral symptoms associated with dementiaJ Am Geriatr Soc5112879812919243
  • AlexopoulosGJesteDCarpenterD2005Expert Consensus Guideline Series: Treatment of dementia and its behavioral disturbancesPostgraduate Medicine Special ReportMcGraw-Hill
  • BallardCG2002Advances in the treatment of Alzheimer’s disease: benefits of dual cholinesterase inhibition (review)Eur Neurol47647011803198
  • BirksJSMelzerD2000Donepezil for mild and moderate Alzheimer’s diseaseCochrane Database Syst Rev4CD00119011034704
  • BlesaRDavidsonMKurzA2003Galantamine provides sustained benefits in patients with “Advanced moderate” Alzheimer’s disease for at least 12 monthsDement Geriatr Cogn Disord15798712566596
  • BraneGGottfriesCGWinbladB2001The Gottfries-Brane-Steen scale: validity, reliability, and application in anti-dementia drug trialsDement Geriatr Cogn Disord1211411125236
  • BurnsARossorMHeckerJ1999The effects of donepezil in Alzheimer’s disease-results from a multinational trialDement Geriatr Cogn Disord102374410325453
  • BurnsASpiegelRQuargP2004Efficacy of rivastigmine in subjects with moderately severe Alzheimer’s diseaseInt J Geriatr Psychiatry19243915027039
  • Cohen-MansfieldJ2001Nonpharmacologic interventions for inappropriate behaviors in dementia: a review, summary, and critiqueAm J Geriatr Psychiatry93618111739063
  • Corey-BloomJAnandRVeachJ1998A randomized trial evaluating the efficacy and safety of ENA 713 (rivastigmine tartrate), a new cholinesterase inhibitor, in patients with mild to moderately severe Alzheimer’s diseaseInt Geriatr Psychopharmacol25565
  • CourtneyCFarrellDGrayRAD 2000 Collaborative group2004Long term donepezil in 565 patients with Alzheimer’s disease (AD 2000): randomized double-blind trialLancet36321051515220031
  • CummingsJLMegaMGrayK1994The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementiaNeurology442308147991117
  • CummingsJL2004Alzheimer’s disease: drug therapyN Engl J Med351566715229308
  • DeDeynPRabheruKRasmussenA1999A randomized trial of risperidone, placebo and haloperidol for behavioral symptoms of dementiaNeurology539465510496251
  • DoodyRSStevensJCBeckC2001Practice parameter: management of dementia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of NeurologyNeurology5611546611342679
  • FarlowMAnandRMessinaJJr2000A 52-week study of the efficacy of rivastigmine in patients with mild to moderately severe Alzheimer’s diseaseEur Neurol2442364111096224
  • [FDA] Food and Drug Administration2005FDA Public Health Advisory. Deaths with antipsychotics in elderly patients with behavioral disorders [online]Accessed 1 October 2005. URL: http://www.fda.gov/cder/drug/advisory/antipsychotics.htm
  • FeldmanHGauthierSHeckerJDonepezil MSAD Study Investigators group2001A 24-week randomized, double-blind study of donepezil in moderate to severe Alzheimer’s diseaseNeurology5761320Correction in Neurology, 57:2153.11524468
  • FinkelSIMintzerJEDyskenM2004A randomized, placebo-controlled study of the efficacy and safety of sertraline in the treatment of behavioral manifestations of Alzheimer’s disease in outpatients treated with donepezilInt J Geriatr Psychiatry1991814716694
  • FolsteinMFFolsteinSEMcHughPR1975“Mini-mental state”: a practical method for grading the cognitive state of subjects for the clinicianJ Psychiatr Res12189981202204
  • GalaskoDBennettDSanoM1997An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. The Alzheimer’s disease cooperative studyAlzheimer Dis Assoc Disord11Suppl 2S3399236950
  • GelinasIGauthierLMcIntyreM1999Development of a functional measure for persons with Alzheimer’s disease: the disability assessment for dementiaAm J Occup Ther534718110500855
  • HolmesCWilkinsonDDeanC2004The efficacy of donepezil in the treatment of neuropsychiatric symptoms in Alzheimer’s diseaseNeurology632141915277611
  • KasckowJWMulchaheyJJMohamedS2004The use of novel antipsychotics in the older patient with neurodegenerative disorders in the long-term care settingJ Am Med Dir Assoc5242815228634
  • KatzIRJesteDVMintzerJERisperidone study group1999Comparison of risperidone and placebo for psychosis and behavioral disturbances associated with dementia: a randomized, double-blind trialJ Clin Psychiatry6010711510084637
  • KmietowiczZ2005NICE proposes to withdraw Alzheimer’s drugs from NHSBMJ33049515746112
  • LyketsosCGDelCampoLSteinbergM2003Treating depression in Alzheimer’s disease: efficacy and safety of sertraline therapy and the benefits of depression reduction: the DIADSArch Gen Psychiatry607374612860778
  • LyketsosCGReichmanWEKershawP2004Long-term outcomes of galantamine treatment in patients with Alzheimer’s diseaseAm J Geriatr Psychiatry124738215353385
  • LyketsosCGSteinbergMTschanzJT2000Mental and behavioral disturbances in dementia: findings from the Cache County Study on memory in agingAm J Psychiatry1577081410784462
  • MaelickeASamochockiMJostockR2001Allosteric sensitization of nicotinic receptors by galantamine, a new treatment strategy for Alzheimer’s diseaseBiol Psychiatry492798811230879
  • MohsRCDoodyRSMorrisJC2001A 1-year placebo-controlled preservation of function survival study of donepezil in Alzheimer’s disease patientsNeurology574818Correction in Neurology, 57:194211502917
  • MolloyDWAlemayehuERobertsR1991Reliability of a standardized mini-mental state examination compared with the traditional mini-mental state examinationAm J Psychiatry14810251984692
  • [NIMH] National Institute of Mental Health1986Clinical global impressionsGuyWECDEU assessment manual for psychopharmacologyRockville, MA21722
  • OlinJSchneiderL2002Galantamine for Alzheimer’s diseaseCochrane Database Syst Rev3CD00174712137632
  • PanissetMRoudierMSaxtonJ1994Severe Impairment battery. A neuropsychological test for severely demented patientsArch Neurol514158274108
  • PeskindERPotkinSGPomaraN2004Memantine monotherapy is effective and safe for the treatment of mild to moderate Alzheimer’s disease: a randomized, controlled trial [abstract]Eur J Neurol11Suppl 2186
  • PetersonRCStevensJCGanguliM2001Practice parameter: early detection of dementia: mild cognitive impairment (an evidence based review). Report of the Quality Standards Subcommittee of the American Academy of NeurologyNeurology5611334211342677
  • PetersenRCThomasRGGrundmanM2005Vitamin E and donepezil for the treatment of mild cognitive impairmentN Engl J Med35223798815829527
  • PollockBGMulsantBHRosenJ2002Comparison of citalopram, perphenazine and placebo for the acute treatment of psychosis and behavioral disturbances in hospitalized, demented patientsAm J Psychiatry159460511870012
  • PorteinssonAPTariotPErbR2001Placebo-controlled study of divalproex sodium for agitation in dementiaAm J Geriatr Psychiatry9586611156753
  • RaskindMAPeskindERWesselT2000Galantamine in Alzheimer’s disease. A 6-month randomized placebo-controlled trial with a 6-month extension. The galantamine USA-1 Study groupNeurology5422617610881250
  • ReissbergBDoodyRStofflerA2003Memantine in moderate to severe Alzheimer’s diseaseN Engl J Med34813334112672860
  • RelkinNRReichmanWEOrazemJ2003A large, community-based, open-label trial of donepezil in the treatment of Alzheimer’s diseaseDement Geriatr Cogn Disord161152412714795
  • RitchieCWAmesDClaytonT2004Metaanalysis of randomized trials of the efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer’s diseaseAm J Geriatr Psychiatry123586915249273
  • RogersSLDoodyRSMohsR1998Donepezil improves cognition and global function in Alzheimer’s disease: a 15-week, double-blind, placebo-controlled study. Donepezil Study groupArch Intern Med1581021319588436
  • RogersSLDoodyRSPrattRD2000Long-term efficacy and safety of donepezil in the treatment of Alzheimer’s disease: final analysis of a US multicentre open-label studyEur Neuropsychopharmacol1019520310793322
  • RogersSLFarlowMRDoodyRS1998A 24-week, double-blind placebo-controlled trial of donepezil in patients with Alzheimer’s disease. Donepezil Study GroupNeurology50136459443470
  • RosenWGMohsRCDavisKL1984A new rating scale for Alzheimer’s diseaseAm J Psychiatry1411356646496779
  • RoslerMAnandRCicin-SainA1999Efficacy and safety of rivastigmine in patients with Alzheimer’s disease: international randomized controlled trialBMJ318633810066203
  • SakauyeKMCampCJFordPA1993Effects of buspirone on agitation associated with dementiaAm J Geriatr Psychiatry1824
  • SchneiderLSOlinJTDoodyRS1997Validity and reliability of the Alzheimer’s disease cooperative study – clinical global impression of change. The Alzheimer’s disease Co-operative StudyAlzheimer Dis Assoc Disord11Suppl2S22329236949
  • SeltzerBZolnouniPNunezM2004Efficacy of donepezil in early-stage Alzheimer’s disease. A randomized Placebo-controlled trialArch Neurol611852615596605
  • SheltonPSHockingLB1997Zolpidem for dementia-related insomnia and nighttime wanderingAnn Pharmacother31319229066939
  • SinkKMHoldenKFYaffeK2005Pharmacological treatment of neuropsychiatric symptoms of dementia, a review of the evidenceJAMA29359660815687315
  • StreetJClarkSGannonK2000Olanzapine treatment of psychotic and behavioral symptoms in patients with Alzheimer’s disease in nursing care facilitiesArch Gen Psychiatry579687611015815
  • TariotPNFarlowMRGrossbergGTMemantine Study Group2004Memantine treatment in patients with moderate to severe Alzheimer’s disease already receiving donepezil, a randomized controlled trialJAMA2913172414734594
  • TariotPNRosemaryRNPodgorskiCA1998Efficacy and tolerability of carbamazepine for agitation and aggression in dementiaAm J Psychiatry15554619433339
  • TariotPNSchneiderLKatzI2002Quetiapine in nursing home residents with Alzheimer’s dementia and psychosisAm J Geriatr Psychiatry10Suppl 193
  • TariotPNSolomonPRMorrisJC2000A 5-month, randomized, placebo-controlled trial of galantamine in Alzheimer’s disease. The Galantamine USA-10 Study GroupNeurology542269227610881251
  • van der KamPMolFWimmersMF1971Beoordelingsschaal voor oudere patienten (BOP)Van Loghum SlaterusThe Netherlands
  • WhitehousePJPriceDLStrubleRG1982Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrainScience215123797058341
  • WilcockGKLilienfieldSGaensE2000Efficacy and safety of galantamine in patients with mild to moderate Alzheimer’s disease: multicenter randomized controlled trial. Galantamine International-1 Study GroupBMJ3211710875807
  • WinbladBEngedalKSoininenHDonepezil Nordic Study2001A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate Alzheimer’s diseaseNeurology574899511502918
  • WinbladBPoritisN1999Memantine in severe dementia: results of the M-BEST study (Benefit and efficacy in severely demented patients during treatment with memantine)Int J Geriatr Psychiatry141354610885864