Free access
Reviews
28 April 2020

Benefits and Harms of Prescription Drugs and Supplements for Treatment of Clinical Alzheimer-Type Dementia: A Systematic Review and Meta-analysisFREE

Publication: Annals of Internal Medicine
Volume 172, Number 10

Abstract

Background:

Effects of drug treatment of clinical Alzheimer-type dementia (CATD) are uncertain.

Purpose:

To summarize evidence on the effects of prescription drugs and supplements for CATD treatment.

Data Sources:

Electronic bibliographic databases (inception to November 2019), ClinicalTrials.gov (to November 2019), and systematic review bibliographies.

Study Selection:

English-language trials of prescription drug and supplement treatment in older adults with CATD that report cognition, function, global measures, behavioral and psychological symptoms of dementia (BPSD), or harms. Minimum treatment was 24 weeks (≥2 weeks for selected BPSD).

Data Extraction:

Studies with low or medium risk of bias (ROB) were analyzed. Two reviewers rated ROB. One reviewer extracted data; another verified extraction accuracy.

Data Synthesis:

Fifty-five studies reporting non-BPSD outcomes (most ≤26 weeks) and 12 reporting BPSD (most ≤12 weeks) were analyzed. Across CATD severity, mostly low-strength evidence suggested that, compared with placebo, cholinesterase inhibitors produced small average improvements in cognition (median standardized mean difference [SMD], 0.30 [range, 0.24 to 0.52]), no difference to small improvement in function (median SMD, 0.19 [range, −0.10 to 0.22]), no difference in the likelihood of at least moderate improvement in global clinical impression (median absolute risk difference, 4% [range, 2% to 4%]), and increased withdrawals due to adverse events. In adults with moderate to severe CATD receiving cholinesterase inhibitors, low- to insufficient-strength evidence suggested that, compared with placebo, add-on memantine inconsistently improved cognition and improved global clinical impression but not function. Evidence was mostly insufficient about prescription drugs for BPSD and about supplements for all outcomes.

Limitation:

Most drugs had few trials without high ROB, especially for supplements, active drug comparisons, BPSD, and longer trials.

Conclusion:

Cholinesterase inhibitors and memantine slightly reduced short-term cognitive decline, and cholinesterase inhibitors slightly reduced reported functional decline, but differences versus placebo were of uncertain clinical importance. Evidence was mostly insufficient on drug treatment of BPSD and on supplements for all outcomes.

Primary Funding Source:

Agency for Healthcare Research and Quality. (PROSPERO: CRD42018117897)
Dementia, a clinical syndrome in which acquired cognitive deficits interfere with independence in daily activities (1), affects about 10% of older U.S. adults (2, 3). In most persons, Alzheimer disease (AD) is the primary cause of dementia. Clinical Alzheimer-type dementia (CATD) impairs cognition and function, lowers quality of life, burdens caregivers, and increases institutionalization (4). Behavioral and psychological symptoms of dementia (BPSD) (such as agitation and psychosis) are common in advanced CATD (5).
A 2008 evidence synthesis (6) and a guideline from the American Academy of Family Physicians (AAFP) and American College of Physicians (7) concluded that cholinesterase inhibitors and memantine improve cognition versus placebo. However, the authors further concluded that the average magnitude of effect was not clinically important and that evidence was limited about whether these drugs increase the likelihood of clinically important improvements in cognition or function. The review did not address prescription drugs for treatment of CATD-associated BPSD or the effects of supplements in CATD. However, off-label use of prescription drugs for BPSD is common, and many supplements are promoted for CATD treatment, including for BPSD, despite uncertainty about their benefits and harms (8–13). In addition, CATD-associated BPSD is a common qualifying condition for state medical marijuana programs (14), although effects of cannabinoids in CATD are also unclear.
To address uncertainties regarding CATD drug treatment and provide an updated evidence base for a new AAFP guideline on CATD management, this systematic review examined 2 questions. First, what are the effects of prescription drugs and supplements for treatment of patients with CATD, including those with BPSD? Second, do treatment effects of CATD drugs vary by patient or drug characteristics?

Methods

We developed and followed a standard protocol that is registered in PROSPERO (CRD42018117897) and available at https://effectivehealthcare.ahrq.gov/topics/alzheimers-type-dementia/protocol. The full technical report contains search strategies, flow diagrams, evidence tables, study quality assessment tables, and detailed results (https://effectivehealthcare.ahrq.gov/products/alzheimers-type-dementia/research).

Data Sources and Searches

We searched MEDLINE, the Cochrane Library, EMBASE, and PsycINFO from inception to November 2019 using search terms for AD or dementia. We searched ClinicalTrials.gov to November 2019 and bibliographies of relevant systematic reviews published since 2013 for additional studies.

Study Selection

We included English-language trials of adults with CATD that compared drug treatment versus placebo, inactive control, or active treatment (drug or nondrug) and reported efficacy or harms. Studies were excluded if participants had dementia attributed solely to a non-AD cause. Cholinesterase inhibitors, memantine, and all supplements were eligible drugs for assessment of non-BPSD efficacy outcomes (cognition, function, global measures of clinical impression, CATD stage, level of independence, and quality of life). Cholinesterase inhibitors; memantine; antipsychotic, antidepressant, anxiolytic, and antiseizure drugs; hormones; supplements; and cannabinoids were eligible drugs for assessment of BPSD efficacy outcomes (patient aggression, agitation, psychosis, disinhibited sexual behavior, general behavior, depression, anxiety, and quality of life, and caregiver depression, distress, burden, and quality of life). All of these drugs were eligible for assessment of the following harms: serious adverse events, withdrawals due to adverse events, somnolence, confusion, falls, extrapyramidal symptoms, stroke, and mortality. Caregiver efficacy and BPSD were evaluated only in participants with both CATD and BPSD at baseline. Minimum study duration was 2 weeks for agitation, aggression, psychosis, and disinhibited sexual behavior and 24 weeks for all other BPSD and non-BPSD efficacy outcomes.
Studies considered possibly eligible by at least 1 of 2 independent reviewers (among P.J.D., M.A.M., P.C.S., M. Brasure, R.M., V.A.N., and M. Butler) after title and abstract review were forwarded for full-text screening. Two reviewers (among P.J.D., M.A.M., P.C.S., M.L.F., M. Brasure, M. Butler, V.A.N., and R.M.) independently screened full-text articles and resolved discrepancies in eligibility decisions by consensus.

Data Extraction and Quality Assessment

For each eligible study, 2 reviewers (among E.J.L., P.J.D., P.C.S., M.L.F., W.N., M. Brasure, M. Butler, K.M.S., K.M.C.T., V.A.N., and R.M.) rated risk of bias (ROB) as low, medium, or high on the basis of guidance from the Agency for Healthcare Research and Quality (AHRQ) (selection, attrition, performance, detection, and reporting bias) (15). High-ROB studies were excluded from further analyses. For studies with low or medium ROB, 1 reviewer (E.J.L., P.J.D., P.C.S., M.L.F., K.M.S., R.M., V.A.N., or M. Brasure) extracted details on study design, inclusion criteria, participant characteristics, drug treatment and control interventions, treatment duration, and efficacy and harms outcomes. A second reviewer (E.J.L., P.C.S., W.N., M. Butler, K.M.C.T., R.M., V.A.N., or M. Brasure) verified extraction accuracy. For each treatment comparison and outcome of interest that was evaluated by at least 2 studies with low or medium ROB or 1 study with low or medium ROB and at least 100 participants, 2 reviewers (among E.J.L., P.J.D., P.C.S., M.L.F., W.N., M. Butler, K.M.S., K.M.C.T., R.M., V.A.N., and M. Brasure) graded strength of evidence (SOE) as insufficient, low, moderate, or high on the basis of study limitations, directness, consistency, and precision (16). When there was only 1 study with low or medium ROB and fewer than 100 participants, SOE was considered insufficient. We confirmed ROB and SOE assessments by consensus.

Data Synthesis and Analysis

For individual studies with low or medium ROB, we used Comprehensive Meta-Analysis, version 3 (Biostat), to calculate standardized mean differences (SMDs) with corresponding 95% CIs for continuous outcomes and risk ratios or Peto odds ratios (the latter for mortality, given few events) with corresponding 95% CIs for binary outcomes.
When data were appropriate for pooling (that is, minimal clinical heterogeneity of participants, interventions, and outcomes), we synthesized data in R (R Foundation) (17, 18). We applied the Hartung–Knapp–Sidik–Jonkman method for random-effects models to calculate SMD for continuous outcomes and relative measures of effect for categorical outcomes with corresponding 95% CIs (19). However, when there were fewer than 5 trials and no between-study variance (τ2 at or near 0), data were meta-analyzed with a fixed-effects model (20). When a treatment group had no events, we used the treatment group continuity correction. For continuous outcomes, we followed existing conventions (21) to characterize SMDs as small (≥0.2 to <0.5), medium (≥0.5 to <0.8), or large (≥0.8). Absolute risk differences (ARDs) were calculated for binary outcomes.
We measured the magnitude of statistical heterogeneity with the I 2 statistic (22). When results suggested substantial heterogeneity (that is, I 2 ≥ 75%) (22), we stratified results by participant or study characteristics or explored sensitivity analyses. When data allowed, we evaluated whether treatment outcomes varied by participant age, sex, race/ethnicity, living setting, prevalence of depression, cognition, function, or study-defined severity of CATD or BPSD, or by drug dose, delivery route, or treatment duration.

Role of the Funding Source

This review was funded by AHRQ. The AAFP, AHRQ staff, and a key informant panel helped refine the project scope. The draft report was reviewed by a technical expert panel, peer and public reviewers, and AHRQ staff. The authors are solely responsible for the content of this article.

Results

We identified 6550 publications from electronic bibliographic databases and hand-searching, of which 66 unique studies met eligibility criteria, had low or medium ROB, and were analyzed (Appendix Figure 1). Only 8 of these trials reported efficacy outcomes beyond 26 weeks (23–30), and just 2 reported BPSD efficacy outcomes beyond 12 weeks (27, 31).
Appendix Figure 1. Evidence search and selection. BPSD = behavioral and psychological symptoms of dementia; ROB = risk of bias. * Because 1 eligible trial with low to medium ROB of persons with BPSD reported both non-BPSD and BPSD outcomes, the total numbers of eligible references and unique studies are smaller than the sums of eligible references and unique studies for the non-BPSD and BPSD outcome groups, and the total number of eligible unique studies with low or medium ROB is lower than the sum of eligible studies with low to medium ROB for the non-BPSD and BPSD outcome groups. † The total number of studies reporting non-BPSD outcomes is smaller than the sum for the separately listed drug classes because individual studies may have included comparisons of drug treatment with placebo, a different formulation or dose of the same drug, a different drug in the same class, or a different drug class.
Appendix Figure 1. Evidence search and selection.
BPSD = behavioral and psychological symptoms of dementia; ROB = risk of bias.
* Because 1 eligible trial with low to medium ROB of persons with BPSD reported both non-BPSD and BPSD outcomes, the total numbers of eligible references and unique studies are smaller than the sums of eligible references and unique studies for the non-BPSD and BPSD outcome groups, and the total number of eligible unique studies with low or medium ROB is lower than the sum of eligible studies with low to medium ROB for the non-BPSD and BPSD outcome groups.
† The total number of studies reporting non-BPSD outcomes is smaller than the sum for the separately listed drug classes because individual studies may have included comparisons of drug treatment with placebo, a different formulation or dose of the same drug, a different drug in the same class, or a different drug class.
Studies most frequently reported between-group differences in mean score change from baseline. Less often, they reported the proportion of participants who had a categorical outcome, including a change of at least 4 points on the Alzheimer's Disease Assessment Scale–Cognitive Subscale and minimal, moderate, or marked improvement on measures of global clinical impression (for example, Alzheimer's Disease Cooperative Study–Clinical Global Impression of Change). Data for some categorical outcomes, including for specific harms, were sparse.

Non-BPSD Outcomes

Cholinesterase Inhibitors

Cholinesterase inhibitors were compared with placebo in 25 trials (n = 9476) (32–56), including 18 of donepezil, 5 of rivastigmine, and 2 of galantamine. Eleven trials (n = 5893) compared different dosages or formulations of the same cholinesterase inhibitor (35, 39, 44, 51–53, 56–60). All trial results were based on 24 to 26 weeks of follow-up. Mean participant age was 75 years; 65% of participants were female; 64% had mild or moderate CATD; baseline Mini-Mental State Examination score was 15; and, in the 17 trials reporting race/ethnicity, 66% of participants were white.
Trials provided mostly low-strength evidence favoring standard dosages of cholinesterase inhibitors (that is, donepezil, 10 mg/d; rivastigmine, 12 mg/d oral or 9.5 mg/d patch; or galantamine, 32 mg/d) over placebo for cognition, function, and global clinical impression (Table 1; Appendix Figures 2 and 3).
Table 1. Efficacy of Standard Doses of CATD Drug Treatments for Cognition, Function, and Global Clinical Impression of Change From Placebo-Controlled Trials of at Least 24 Weeks' Duration and With Low or Medium Risk of Bias*
Table 1. Efficacy of Standard Doses of CATD Drug Treatments for Cognition, Function, and Global Clinical Impression of Change From Placebo-Controlled Trials of at Least 24 Weeks' Duration and With Low or Medium Risk of Bias*
Appendix Figure 2. Forest plots for drug treatment: donepezil vs. placebo. ADAS-Cog = Alzheimer's Disease Assessment Scale–Cognitive Subscale; CATD = clinical Alzheimer-type dementia; MMSE = Mini-Mental State Examination; OR = odds ratio; RR = risk ratio; SMD = standardized mean difference.
Appendix Figure 2. Forest plots for drug treatment: donepezil vs. placebo.
ADAS-Cog = Alzheimer's Disease Assessment Scale–Cognitive Subscale; CATD = clinical Alzheimer-type dementia; MMSE = Mini-Mental State Examination; OR = odds ratio; RR = risk ratio; SMD = standardized mean difference.
Appendix Figure 2—Continued
Appendix Figure 2. Continued
Appendix Figure 2—Continued
Appendix Figure 2. Continued
Appendix Figure 2—Continued
Appendix Figure 2. Continued
Appendix Figure 3. Forest plots for drug treatment: rivastigmine vs. placebo. ADAS-Cog = Alzheimer's Disease Assessment Scale–Cognitive Subscale; CATD = clinical Alzheimer-type dementia; MMSE = Mini-Mental State Examination; OR = odds ratio; RR = risk ratio; SMD = standardized mean difference. * Two other trials of rivastigmine vs. placebo in patients with mild to moderate CATD did not report on the Progressive Deterioration Scale. Instead, Winblad and colleagues (52) reported on the Alzheimer's Disease Cooperative Study–Activities of Daily Living, and Nakamura and colleagues (53) reported on the Disability Assessment for Dementia.
Appendix Figure 3. Forest plots for drug treatment: rivastigmine vs. placebo.
ADAS-Cog = Alzheimer's Disease Assessment Scale–Cognitive Subscale; CATD = clinical Alzheimer-type dementia; MMSE = Mini-Mental State Examination; OR = odds ratio; RR = risk ratio; SMD = standardized mean difference.
* Two other trials of rivastigmine vs. placebo in patients with mild to moderate CATD did not report on the Progressive Deterioration Scale. Instead, Winblad and colleagues (52) reported on the Alzheimer's Disease Cooperative Study–Activities of Daily Living, and Nakamura and colleagues (53) reported on the Disability Assessment for Dementia.
Appendix Figure 3—Continued
Appendix Figure 3. Continued
Average benefits versus placebo were small for mean change in cognitive test score (median pooled drug-specific SMD, 0.30 [range, 0.24 to 0.52]) (32–35, 37, 38, 40–42, 44–54, 56) and ranged from no difference to a small benefit for mean change in function (median pooled drug-specific SMD, 0.19 [range, −0.10 to 0.22]) (34, 37–39, 49–53, 56).
Results for the likelihood of improvement from baseline favored cholinesterase inhibitors over placebo for cognition (≥4-point change in Alzheimer's Disease Assessment Scale–Cognitive Subscale) (median ARD, 16% [range, 8% to 19%]) (45, 50–52, 56). Similarly, results favored cholinesterase inhibitors for the proportion of participants with minimal or greater improvement in global clinical impression (median ARD, 10% [range, 8% to 13%]) (37–41, 44, 45, 48, 50–52, 54). However, moderate or marked improvement in global clinical impression was uncommon and did not differ between treatments (median ARD, 4% [range, 2% to 4%]) (37, 39, 40, 48, 50, 52, 53). Quality of life was reported in only 2 trials (44, 45), and evidence was insufficient.
Compared with placebo, standard doses of donepezil and rivastigmine increased withdrawals due to adverse events (32–48, 51–56) and inconsistently increased serious adverse events (34–37, 39, 41–46, 48, 51–54, 56) (Table 2; Appendix Figures 2 and 3). Low doses of cholinesterase inhibitors did not differ from placebo for these harms (39, 44, 45, 51, 53).
Table 2. Harms of Standard Doses of CATD Drug Treatments for Cognition, Function, or Global Clinical Impression of Change From Placebo-Controlled Trials of at Least 24 Weeks' Duration and With Low or Medium Risk of Bias*
Table 2. Harms of Standard Doses of CATD Drug Treatments for Cognition, Function, or Global Clinical Impression of Change From Placebo-Controlled Trials of at Least 24 Weeks' Duration and With Low or Medium Risk of Bias*
Evidence conflicted about whether the efficacy of cholinesterase inhibitors versus placebo varied by baseline cognitive level (34, 48–50), and no trials reported whether efficacy varied by other participant characteristics. On the basis of direct and indirect comparisons, we found mostly low- to moderate-strength evidence that efficacy did not differ by donepezil dose, that standard rivastigmine doses had greater efficacy than placebo but low doses did not, and that evidence on efficacy by galantamine dosing was mostly insufficient. For harms, compared with 10 mg of donepezil per day, higher dosing increased withdrawals due to adverse events (moderate SOE) (57, 58) and lower dosing reduced serious adverse events (low SOE) and withdrawals due to adverse events (moderate SOE). Evidence was insufficient about differences in harms between galantamine doses (59) or rivastigmine doses or formulations (51–53, 60).

Memantine

Six trials (n = 2227) compared memantine versus placebo, of which 5 evaluated dosages up to 20 mg/d (23, 61–64) and 1 evaluated 28 mg/d (65). Follow-up results were available through 30 weeks for 1 trial (23) and 24 weeks for the others. Mean participant age was 77 years; 64% of participants were female; 63% had moderate to severe CATD; mean baseline score on the Mini-Mental State Examination was 13; and, in 4 trials reporting race/ethnicity, 93% of participants were white.
In 2 trials in adults with mild to moderate CATD, memantine and placebo did not differ for mean change in function (low SOE) and evidence was insufficient for mean change in cognition (Table 1). Results for measures of global clinical impression were inconsistent. In 1 trial in which participants did not receive a cholinesterase inhibitor, those assigned memantine had a small improvement in mean change in global clinical impression and were more likely to experience a composite outcome of no change or any improvement (low SOE) (61). By comparison, in 1 trial in patients who received a cholinesterase inhibitor, mean change in global clinical impression did not differ between memantine and placebo (low SOE) (63).
In 4 trials in adults with moderate to severe CATD, all of whom were receiving a cholinesterase inhibitor, add-on memantine did not differ from placebo for mean change in function (low SOE), there was a small benefit with memantine in mean change in global clinical impression (low SOE), and evidence for memantine benefit in continuous cognition measures varied by type of cognitive measure (23, 62, 64, 65) (Table 1).
No memantine trials reported data on the likelihood that persons improved in cognition, function, or global measures, let alone whether they had moderate or marked improvement.
Memantine and placebo did not differ for serious adverse events (61, 63, 65) or withdrawals due to adverse events (61, 63–65) (Table 2; Appendix Figure 4). Somnolence was higher with memantine than placebo (4.4% vs. 1.1%; risk ratio, 3.75 [95% CI, 1.36 to 10.30]) (61, 65), but treatment groups did not statistically differ for confusion (61, 63–65), falls (23, 61, 63–65), stroke (23, 65), or mortality (23, 61, 63, 65). No studies reported data on extrapyramidal symptoms.
Appendix Figure 4. Forest plots for drug treatment: memantine plus cholinesterase inhibitor vs. placebo plus cholinesterase inhibitor. CATD = clinical Alzheimer-type dementia; OR = odds ratio; RR = risk ratio.
Appendix Figure 4. Forest plots for drug treatment: memantine plus cholinesterase inhibitor vs. placebo plus cholinesterase inhibitor.
CATD = clinical Alzheimer-type dementia; OR = odds ratio; RR = risk ratio.
Other than the possible effect modification by CATD severity detailed earlier, no studies reported whether differences in efficacy between memantine and placebo vary by other participant characteristics.

Supplements

Twelve trials of supplements versus placebo (n = 2214) and 4 of supplements versus prescription drugs (n = 258) reported effects on cognition, function, or harms in persons with CATD.
Two 24-week trials (n = 786) reported no difference between Souvenaid (Nutricia Advanced Medical Nutrition) and placebo for function, serious adverse events, or withdrawals due to adverse events (all low SOE), but evidence was insufficient for cognition (66–68) (Tables 1 and 2; Appendix Figure 5). Two trials (n = 230) that studied omega-3 fatty acids versus placebo for 6 and 12 months reported no difference for cognition (low SOE) and insufficient evidence about function, other efficacy outcomes, and harms (24, 69, 70). One trial (n = 210) reported low-strength evidence that oral vitamin D3, 800 IU/d, versus placebo for 12 months improved mean change in cognition but found insufficient evidence about other outcomes (30).
Appendix Figure 5. Forest plots for drug treatment: Souvenaid vs. placebo. CATD = clinical Alzheimer-type dementia; OR = odds ratio; RR = risk ratio.
Appendix Figure 5. Forest plots for drug treatment: Souvenaid vs. placebo.
CATD = clinical Alzheimer-type dementia; OR = odds ratio; RR = risk ratio.
By comparison, evidence was insufficient for all evaluated outcomes for the following supplements compared with placebo: omega-3 fatty acids combined with α-lipoic acid (n = 26) (24), an antioxidant combination (n = 52) (71), choline alfoscerate (n = 261) (72), prolonged-release melatonin (n = 80) (73), sodium selenite (n = 40) (74), soy isoflavones (n = 65) (75), copper (n = 68) (28), and folic acid combined with vitamin B (n = 409) (29).

Prescription Drugs Versus Prescription Drug or Supplement

Seven trials compared prescription drugs with each other or supplements. All provided insufficient-strength evidence for between-treatment differences in all reported efficacy and harms outcomes. These comparisons were donepezil versus galantamine (n = 188) (25), memantine (n = 67) (76), ginkgo biloba (n = 50) (47), vitamin E (n = 40) (77), or Huannao Yicong formula (Beijing CMages Pharmaceutical) (n = 60) (78); memantine versus continued antipsychotic drug therapy (n = 199) (31) or saffron (n = 68) (26); and vitamin E versus rivastigmine (n = 40) (77).

BPSD Outcomes in Persons With CATD and BPSD

We found no eligible trials that enrolled adults with CATD and BPSD, had low or medium ROB, and reported BPSD outcomes with antianxiety drugs, hormones, or cannabinoids.

Antipsychotic Drugs

Four trials (n = 522) compared antipsychotic drugs (haloperidol [79], quetiapine [80], aripiprazole [81], and pimavanserin [82, 83]) versus placebo. Two comprised community-dwelling adults (n = 279) (79, 81), and 2 were limited to nursing home residents (n = 243) (80, 82, 83). One trial enrolled participants with agitation, 2 enrolled those with psychosis, and 1 required agitation or psychosis. Study durations were 6 to 12 weeks.
Evidence was insufficient about differences between antipsychotics and placebo for agitation and aggression (79, 80, 82, 83). Although antipsychotics were statistically significantly better than placebo for a few of many reported psychosis outcomes (79, 81–83), evidence for psychosis was also deemed insufficient, primarily because of imprecision and inconsistency of results within trials (Table 3). No trials reported data on disinhibited sexual behavior, and none reported data at 24 weeks or later for depression, anxiety, general behavior, or caregiver outcomes.
Table 3. CATD Drug Treatment Efficacy for BPSD From Trials of at Least 2 Weeks' Duration and With Low or Medium Risk of Bias
Table 3. CATD Drug Treatment Efficacy for BPSD From Trials of at Least 2 Weeks' Duration and With Low or Medium Risk of Bias
Evidence was insufficient about differences between antipsychotics and placebo for serious adverse events or withdrawals due to adverse events (Table 4; Appendix Figure 6). Falls seemed similar between groups in 1 trial (23% vs. 23%) (82, 83). In a second trial, both injurious falls (8% vs. 1%) and somnolence (8% vs. 1%) seemed more common with antipsychotics than placebo (81). Two trials (n = 279) reported that changes in extrapyramidal symptoms and abnormal movements did not statistically differ between placebo and standard-dose haloperidol (79) or aripiprazole (81). In 3 trials reporting it (n = 451), mortality over 6 to 12 weeks was 4.4% in the antipsychotic group and 1.8% with placebo (ARD, 2.6% [CI, −0.6% to 5.8%]) (80–83). No trials reported data on confusion or stroke.
Table 4. Harms of CATD Drug Treatments From Placebo-Controlled Trials Reporting BPSD of at Least 2 Weeks' Duration and With Low or Medium Risk of Bias
Table 4. Harms of CATD Drug Treatments From Placebo-Controlled Trials Reporting BPSD of at Least 2 Weeks' Duration and With Low or Medium Risk of Bias
Appendix Figure 6. Forest plots for drug treatment: antipsychotic drugs vs. placebo. OR = odds ratio; RR = risk ratio.
Appendix Figure 6. Forest plots for drug treatment: antipsychotic drugs vs. placebo.
OR = odds ratio; RR = risk ratio.
In 1 exploratory analysis, the likelihood that participants' psychosis symptoms improved with pimavanserin compared with placebo seemed greater in those with worse psychosis (n = 57) than in trial participants overall (82, 83).

Antidepressant Drugs

Four trials (n = 836) compared antidepressant drugs (citalopram [84–86], sertraline [27, 87], and mirtazapine [27]) versus placebo. Two were restricted to participants with agitation or aggression (84–86), 1 to those with depression (27), and 1 to those with increased neuropsychiatric symptom scores (87). Eighty-nine percent of participants were community-dwelling. Both citalopram trials used 30-mg/d dosing, exceeding the maximum currently recommended dose for patients older than 60 years (88). Study durations were 8 to 39 weeks.
Evidence was insufficient about differences between antidepressants and placebo for treatment of BPSD. Among persons with agitation or aggression (n = 430), results for psychosis were insufficient, and no data were reported for disinhibited sexual behavior (Table 3). Although high-dose citalopram was statistically significantly better than placebo for some reported agitation outcomes, results were not consistent, and evidence was considered insufficient. Citalopram was associated with statistically significantly more improvement in caregiver distress than placebo (84–86), but sertraline and placebo did not statistically differ for change in caregiver burden (87). Among persons with depression (n = 326), low-strength evidence showed no difference between antidepressants and placebo for participant depression, general behavior, or quality of life (27). No studies reported data on participant anxiety or caregiver depression.
Evidence was insufficient to draw conclusions about differences between antidepressants and placebo for serious adverse events or withdrawals due to adverse events (Table 4; Appendix Figure 7). In 1 trial reporting confusion, falls, and somnolence (n = 176), citalopram and placebo did not seem to differ for these outcomes (84, 85). In 2 trials reporting mortality (n = 404), citalopram and placebo also did not seem to differ (27, 84, 85). No studies reported data on extrapyramidal symptoms or stroke.
Appendix Figure 7. Forest plots for drug treatment: antidepressant drugs vs. placebo. OR = odds ratio; RR = risk ratio.
Appendix Figure 7. Forest plots for drug treatment: antidepressant drugs vs. placebo.
OR = odds ratio; RR = risk ratio.
In separate trials, neither change in posttreatment depression score (27) nor caregiver burden (87) varied between antidepressant and placebo groups by baseline BPSD severity.

Cholinesterase Inhibitors

In a 12-week trial (n = 272) in persons with agitation who were living in a residential care facility and were unresponsive to psychosocial interventions, donepezil and placebo did not differ for change in agitation (low SOE) (Table 3) (89). Falls (1.6% with donepezil and 1.5% with placebo) and stroke (0.8% with donepezil and 0% with placebo) were rare. Mortality was 2.3% and 3.1% in the donepezil and placebo groups, respectively. Results for other efficacy and harms outcomes either were of high ROB and were not analyzed or were not reported (Tables 3 and 4).

Antiseizure Drugs

In a 6-week trial (n = 153) of divalproex versus placebo in nursing home residents with agitation, evidence was insufficient about between-treatment differences in change in agitation from baseline (Table 3) (90). No data were reported on other participant BPSD outcomes, caregiver outcomes, serious adverse events, or withdrawals due to adverse events (Table 4). Falls occurred in 21% of the divalproex group and 17% of the placebo group, and 1 divalproex participant died. No studies reported data on confusion, somnolence, stroke, or extrapyramidal symptoms.

Supplements

In a 4-week trial (n = 145) of yokukansan (Zenyaku Japan and Tsumura) versus placebo in mostly community-dwelling adults with agitation, aggression, or irritability, evidence was insufficient about between-treatment differences in all reported efficacy and harms outcomes (Tables 3 and 4) (91).

Discussion

This systematic review summarized the evidence on the efficacy and harms of prescription drugs and supplements for CATD, including effects on non-BPSD outcomes (such as cognition, function, and global clinical impression), BPSD outcomes (such as agitation, aggression, psychosis, depression, and anxiety), and caregiver outcomes.
We found that cholinesterase inhibitors compared with placebo were associated with small reductions in the average amount of worsening in cognition and function over about 6 months. Although likelihood of at least minimal improvement in global measures was greater with cholinesterase inhibitors than placebo, at least moderate improvement was uncommon and no more likely with treatment than placebo. Data were mixed regarding differences in efficacy by cholinesterase inhibitor dose, but standard doses more consistently increased harms versus placebo, whereas low doses did not. Memantine generally did not differ from placebo in patients with mild to moderate CATD but had inconsistent benefits and few harms when added to a cholinesterase inhibitor in those with moderate to severe CATD. Evidence for the effects of supplements on cognition and function was mostly insufficient. For treatment of BPSD, despite isolated statistically significant findings favoring antipsychotics over placebo for psychosis and high-dose citalopram over placebo for agitation, findings were insufficient because of few studies, small sample sizes, and inconsistent results within and between trials. Antipsychotic trials were small and short and reported relatively few total deaths, but findings were not inconsistent with warnings from the U.S. Food and Drug Administration about increased risk (9, 10). Evidence on efficacy and harms of BPSD treatment with other antidepressants, antiseizure drugs, antianxiety drugs, hormones, cannabinoids, and supplements was insufficient or absent. Among trials that directly compared drug treatments, no 2 with low or medium ROB evaluated the same treatment comparison, and all provided insufficient evidence to conclude anything about relative benefits and harms.
Our findings have several clinical implications. They suggest that cholinesterase inhibitors may have symptomatic benefits in cognition, function, and global measures compared with placebo. Only 1 small trial (n = 67) directly compared a cholinesterase inhibitor versus memantine, but evidence of benefit versus placebo seems weaker for memantine than for cholinesterase inhibitors. However, the clinical importance of these treatment differences versus placebo is unclear. Based on conventional interpretations of effect size (21), the average differences in efficacy of cholinesterase inhibitors and memantine versus placebo as estimated by SMD are small. Although cholinesterase inhibitors were more likely than placebo to produce a change of at least 4 points in the Alzheimer's Disease Assessment Scale–Cognitive Subscale, this commonly cited metric for clinically important change in cognition is based on U.S. Food and Drug Administration expert opinion (92) and has not been adequately validated on the basis of patient and caregiver values and preferences (93). Although cholinesterase inhibitors were more likely than placebo to at least minimally improve global clinical impression, this threshold probably correlates with a minimal detectable difference (93). The outcome of at least moderate improvement in global clinical impression is more likely to meet or exceed the minimal clinically important difference, but this outcome occurred no more frequently in participants assigned cholinesterase inhibitors than placebo. We also found no trial data on the effects of drug treatments on function at home or delay in the need for higher-level care (for example, transfer from independent to assisted living)—outcomes that are important to participants. Current data do not provide clear guidance on whether any prescription drugs improve BPSD outcomes in patients with CATD, let alone whether benefits outweigh harms, overall or in any patient subgroups. Finally, from trials included in this review and rated as having low or medium ROB, we could not establish whether any supplements or cannabinoids have clinically important benefits for any outcome that would support their use in persons with CATD, including those with BPSD.
Evidence for this review was limited in several ways. Although more than half of the analyzed studies were published after the search date for the 2008 evidence synthesis and guideline from the AAFP and American College of Physicians (6, 7), substantially augmenting the evidence base (including for supplements and treatment of BPSD), the general findings of our review are similar, and significant evidence gaps remain. We identified few eligible trials for many individual drug treatments. Among eligible trials, many had high ROB due to attrition, especially longer trials and trials of supplements, active drug comparisons, and treatments for BPSD. We found no trials that compared drug versus nondrug treatments. By design, this review did not examine the effects of drug treatment on cognition or function for less than 24 weeks or drug treatment of BPSD for less than 2 weeks. By design, this review also did not examine risk for many individual harms defined by trials as nonserious and not associated with study withdrawals. Because few trials evaluating cognition or function exceeded 26 weeks and few for BPSD exceeded 12 weeks, longer-term drug effects were unclear—although patients generally live with CATD for years (94, 95). Most trials had small sample sizes, and treatment effects were inconsistent within and between trials. This limited statistical power to detect potentially meaningful differences between treatments in both efficacy and harms. In addition, some trials analyzed results using methods of accounting for missing data that may overestimate treatment benefit in a progressive disease like CATD (such as last observation carried forward). Few trials reported data on participant quality of life or caregiver outcomes, and none with low or medium ROB reported data on disinhibited sexual behavior. Harms reporting was poor. Last, few trials evaluated whether treatment efficacy or harms vary by participant characteristics, and none evaluated whether they vary by accuracy of CATD diagnosis.
Future trials of CATD drug treatment should be large enough to detect the likelihood of treatment response as defined for validated, clinically important outcomes related to cognition, function, global function, and BPSD. Future trials should investigate treatment efficacy and harms beyond 6 months. They should enroll more diverse participants, including nonwhite persons, and prespecify analyses to examine whether treatment effects are modified by participant characteristics, including age, sex, race/ethnicity, CATD and BPSD severity, and living setting. In addition to agitation, aggression, and psychosis, BPSD treatment trials should routinely report data on participant depression, anxiety, disinhibited sexual behavior, and quality of life and caregiver outcomes. Future trials should examine prescription drug treatments for which data, although currently insufficient, raise questions about possible benefit. For example, high-dose citalopram data suggest that lower-dose citalopram for agitation warrants investigation. Future trials also should rigorously evaluate nonprescription drug treatments (such as supplements and cannabinoids) for which claims of efficacy have been made in comparison with both placebo and prescription drugs approved by the U.S. Food and Drug Administration. Last, future BPSD trials should directly compare benefits and harms of drug treatments with nondrug treatment strategies.

References

1.
McKhann GMKnopman DSChertkow Het al. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7:263-9. [PMID: 21514250]  doi: 10.1016/j.jalz.2011.03.005
2.
Plassman BLLanga KMFisher GGet al. Prevalence of dementia in the United States: the Aging, Demographics, and Memory Study. Neuroepidemiology. 2007;29:125-32. [PMID: 17975326]
3.
Langa KMLarson EBCrimmins EMet al. A comparison of the prevalence of dementia in the United States in 2000 and 2012. JAMA Intern Med. 2017;177:51-8. [PMID: 27893041]  doi: 10.1001/jamainternmed.2016.6807
4.
Hurd MDMartorell PDelavande Aet al. Monetary costs of dementia in the United States. N Engl J Med. 2013;368:1326-34. [PMID: 23550670]  doi: 10.1056/NEJMsa1204629
5.
Zhao QFTan LWang HFet al. The prevalence of neuropsychiatric symptoms in Alzheimer's disease: systematic review and meta-analysis. J Affect Disord. 2016;190:264-71. [PMID: 26540080]  doi: 10.1016/j.jad.2015.09.069
6.
Raina PSantaguida PIsmaila Aet al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148:379-97. [PMID: 18316756]
7.
Qaseem ASnow VCross JT Jret alAmerican College of Physicians/American Academy of Family Physicians Panel on Dementia. Current pharmacologic treatment of dementia: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med. 2008;148:370-8. [PMID: 18316755]
8.
Kales HCGitlin LNLyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350:h369. [PMID: 25731881]  doi: 10.1136/bmj.h369
9.
U.S. Food and Drug Administration. Public health advisory: deaths with antipsychotics in elderly patients with behavioral disturbances. 11 April 2005. Updated 16 August 2013. Accessed at https://wayback.archive-it.org/7993/20170113112252/ http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm053171.htm on 10 July 2019.
10.
U.S. Food and Drug Administration. Information for healthcare professionals: conventional antipsychotics. Updated 15 August 2013. Accessed at https://wayback.archive-it.org/7993/20170722190727/ https://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm124830.htm on 10 July 2019.
11.
U.S. Food and Drug Administration. Advisory letters: products illegally marketed for serious diseases. Accessed at www.fda.gov/inspections-compliance-enforcement-and-criminal-investigations/compliance-actions-and-activities/advisory-letters on 17 February 2020.
12.
The Alzheimer's Organization. Vitamins for Alzheimer's and dementia. Accessed at www.alzheimersorganization.org/vitamins on 17 February 2020.
13.
Alzheimer's Research & Prevention Foundation. Pillar 1: diet & supplements. Accessed at https://alzheimersprevention.org/4-pillars-of-prevention/pillar-1-diet-supplements on 17 February 2020.
14.
Qualifying conditions for a medical marijuana card by state. Leafly. 2019. Accessed at www.leafly.com/news/health/qualifying-conditions-for-medical-marijuana-by-state on 24 June 2019.
15.
Viswanathan MAnsari MTBerkman NDet al. Methods Guide for Effectiveness and Comparative Effectiveness Reviews. Agency for Healthcare Research and Quality; 2008.
16.
Owens DKLohr KNAtkins Det al. AHRQ series paper 5: grading the strength of a body of evidence when comparing medical interventions—Agency for Healthcare Research and Quality and the Effective Health-Care Program. J Clin Epidemiol. 2010;63:513-23. [PMID: 19595577]  doi: 10.1016/j.jclinepi.2009.03.009
17.
Schwarzer G. Meta: an R package for meta-analysis. R News. 2007;7:40-5.
18.
Viechtbauer W. Conducting meta-analyses in R with the metafor package. J Stat Softw. 2010;36:1-48.  doi: 10.18637/jss.v036.i03
19.
IntHout JIoannidis JPBorm GF. The Hartung-Knapp-Sidik-Jonkman method for random effects meta-analysis is straightforward and considerably outperforms the standard DerSimonian-Laird method. BMC Med Res Methodol. 2014;14:25. [PMID: 24548571]  doi: 10.1186/1471-2288-14-25
20.
Veroniki AAJackson DBender Ret al. Methods to calculate uncertainty in the estimated overall effect size from a random-effects meta-analysis. Res Synth Methods. 2019;10:23-43. [PMID: 30129707]  doi: 10.1002/jrsm.1319
21.
Cohen J. Statistical Power Analysis for the Behavioral Sciences. 2nd ed. Lawrence Erlbaum Associates; 1988.
22.
Higgins JPThompson SGDeeks JJet al. Measuring inconsistency in meta-analyses. BMJ. 2003;327:557-60. [PMID: 12958120]
23.
Howard RMcShane RLindesay Jet al. Donepezil and memantine for moderate-to-severe Alzheimer's disease. N Engl J Med. 2012;366:893-903. [PMID: 22397651]  doi: 10.1056/NEJMoa1106668
24.
Shinto LQuinn JMontine Tet al. A randomized placebo-controlled pilot trial of omega-3 fatty acids and alpha lipoic acid in Alzheimer's disease. J Alzheimers Dis. 2014;38:111-20. [PMID: 24077434]  doi: 10.3233/JAD-130722
25.
Wilcock GHowe IColes Het alGAL-GBR-2 Study Group. A long-term comparison of galantamine and donepezil in the treatment of Alzheimer's disease. Drugs Aging. 2003;20:777-89. [PMID: 12875613]
26.
Farokhnia MShafieeSabet MIranpour Net al. Comparing the efficacy and safety of Crocus sativus L. with memantine in patients with moderate to severe Alzheimer's disease: a double-blind randomized clinical trial. Hum Psychopharmacol. 2014;29:351-9. [PMID: 25163440]  doi: 10.1002/hup.2412
27.
Banerjee SHellier JDewey Met al. Sertraline or mirtazapine for depression in dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-controlled trial. Lancet. 2011;378:403-11. [PMID: 21764118]  doi: 10.1016/S0140-6736(11)60830-1
28.
Kessler HBayer TABach Det al. Intake of copper has no effect on cognition in patients with mild Alzheimer's disease: a pilot phase 2 clinical trial. J Neural Transm (Vienna). 2008;115:1181-7. [PMID: 18587525]  doi: 10.1007/s00702-008-0080-1
29.
Aisen PSSchneider LSSano Met alAlzheimer Disease Cooperative Study. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA. 2008;300:1774-83. [PMID: 18854539]  doi: 10.1001/jama.300.15.1774
30.
Jia JHu JHuo Xet al. Effects of vitamin D supplementation on cognitive function and blood Aβ-related biomarkers in older adults with Alzheimer's disease: a randomised, double-blind, placebo-controlled trial. J Neurol Neurosurg Psychiatry. 2019;90:1347-52. [PMID: 31296588]  doi: 10.1136/jnnp-2018-320199
31.
Ballard CThomas AGerry Set alMAIN-AD investigators. A double-blind randomized placebo-controlled withdrawal trial comparing memantine and antipsychotics for the long-term treatment of function and neuropsychiatric symptoms in people with Alzheimer's disease (MAIN-AD). J Am Med Dir Assoc. 2015;16:316-22. [PMID: 25523285]  doi: 10.1016/j.jamda.2014.11.002
32.
Moraes Wdos SPoyares DRGuilleminault Cet al. The effect of donepezil on sleep and REM sleep EEG in patients with Alzheimer disease: a double-blind placebo-controlled study. Sleep. 2006;29:199-205. [PMID: 16494088]
33.
Tune LTiseo PJIeni Jet al. Donepezil HCl (E2020) maintains functional brain activity in patients with Alzheimer disease: results of a 24-week, double-blind, placebo-controlled study. Am J Geriatr Psychiatry. 2003;11:169-77. [PMID: 12611746]
34.
Maher-Edwards GWatson CAscher Jet al. Two randomized controlled trials of SB742457 in mild-to-moderate Alzheimer's disease. Alzheimers Dement (N Y). 2015;1:23-36. [PMID: 29854923]  doi: 10.1016/j.trci.2015.04.001
35.
Tariot PNCummings JLKatz IRet al. A randomized, double-blind, placebo-controlled study of the efficacy and safety of donepezil in patients with Alzheimer's disease in the nursing home setting. J Am Geriatr Soc. 2001;49:1590-9. [PMID: 11843990]
36.
Winblad BEngedal KSoininen Het alDonepezil Nordic Study Group. A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology. 2001;57:489-95. [PMID: 11502918]
37.
Winblad BKilander LEriksson Set alSevere Alzheimer's Disease Study Group. Donepezil in patients with severe Alzheimer's disease: double-blind, parallel-group, placebo-controlled study. Lancet. 2006;367:1057-65. [PMID: 16581404]  doi: 10.1016/S0140-6736(06)68350-5
38.
Black SEDoody RLi Het al. Donepezil preserves cognition and global function in patients with severe Alzheimer disease. Neurology. 2007;69:459-69. [PMID: 17664405]
39.
Homma AImai YTago Het al. Donepezil treatment of patients with severe Alzheimer's disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25:399-407. [PMID: 18391486]  doi: 10.1159/000122961
40.
Homma ATakeda MImai Yet al. Clinical efficacy and safety of donepezil on cognitive and global function in patients with Alzheimer's disease. A 24-week, multicenter, double-blind, placebo-controlled study in Japan. E2020 Study Group. Dement Geriatr Cogn Disord. 2000;11:299-313. [PMID: 11044775]
41.
Jia JWei CJia Let al. Efficacy and safety of donepezil in Chinese patients with severe Alzheimer's disease: a randomized controlled trial. J Alzheimers Dis. 2017;56:1495-1504. [PMID: 28157100]  doi: 10.3233/JAD-161117
42.
Seltzer BZolnouni PNunez Met alDonepezil “402” Study Group. Efficacy of donepezil in early-stage Alzheimer disease: a randomized placebo-controlled trial. Arch Neurol. 2004;61:1852-6. [PMID: 15596605]
43.
Mohs RCDoody RSMorris JCet al“312” Study Group. A 1-year, placebo-controlled preservation of function survival study of donepezil in AD patients. Neurology. 2001;57:481-8. [PMID: 11502917]
44.
Burns ARossor MHecker Jet al. The effects of donepezil in Alzheimer's disease - results from a multinational trial. Dement Geriatr Cogn Disord. 1999;10:237-44. [PMID: 10325453]
45.
Rogers SLFarlow MRDoody RSet al. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer's disease. Donepezil Study Group. Neurology. 1998;50:136-45. [PMID: 9443470]
46.
Feldman HGauthier SHecker Jet alDonepezil MSAD Study Investigators Group. A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer's disease. Neurology. 2001;57:613-20. [PMID: 11524468]
47.
Mazza MCapuano ABria Pet al. Ginkgo biloba and donepezil: a comparison in the treatment of Alzheimer's dementia in a randomized placebo-controlled double-blind study. Eur J Neurol. 2006;13:981-5. [PMID: 16930364]
48.
Maher-Edwards GDixon RHunter Jet al. SB-742457 and donepezil in Alzheimer disease: a randomized, placebo-controlled study. Int J Geriatr Psychiatry. 2011;26:536-44. [PMID: 20872778]  doi: 10.1002/gps.2562
49.
Burns ABernabei RBullock Ret al. Safety and efficacy of galantamine (Reminyl) in severe Alzheimer's disease (the SERAD study): a randomised, placebo-controlled, double-blind trial. Lancet Neurol. 2009;8:39-47. [PMID: 19042161]  doi: 10.1016/S1474-4422(08)70261-8
50.
Wticock GKLilienfeld SGaens EGalantamine International-1 Study Group. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer's disease: multicentre randomised controlled trial. BMJ. 2000;321:1445-9. [PMID: 11110737]
51.
Rösler MAnand RCicin-Sain Aet al. Efficacy and safety of rivastigmine in patients with Alzheimer's disease: international randomised controlled trial. BMJ. 1999;318:633-8. [PMID: 10066203]
52.
Winblad BCummings JAndreasen Net al. A six-month double-blind, randomized, placebo-controlled study of a transdermal patch in Alzheimer's disease—rivastigmine patch versus capsule. Int J Geriatr Psychiatry. 2007;22:456-67. [PMID: 17380489]
53.
Nakamura YImai YShigeta Met al. A 24-week, randomized, double-blind, placebo-controlled study to evaluate the efficacy, safety and tolerability of the rivastigmine patch in Japanese patients with Alzheimer's disease. Dement Geriatr Cogn Dis Extra. 2011;1:163-79. [PMID: 22163242]  doi: 10.1159/000328929
54.
Lopez-Pousa S. Pilot, multicenter, randomized, doubleblind, controlled, parallel efficacy and safety study of rivastigmine vs placebo in the treatment of cognitive and non-cognitive symptoms in patients with moderate-to-severe Alzheimer's disease. IFPMA Register. 2005. (As cited in Birks JS, Chong LY, Grimley Evans J. Rivastigmine for Alzheimer's disease. Cochrane Database Syst Rev. 2015;9:CD001191. [PMID: 26393402]
55.
Krishnan KRCharles HCDoraiswamy PMet al. Randomized, placebo-controlled trial of the effects of donepezil on neuronal markers and hippocampal volumes in Alzheimer's disease. Am J Psychiatry. 2003;160:2003-11. [PMID: 14594748]
56.
Feldman HHLane RStudy 304 Group. Rivastigmine: a placebo controlled trial of twice daily and three times daily regimens in patients with Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2007;78:1056-63. [PMID: 17353259]
57.
Farlow MRCummings JLOlin JTet al. Effects of oral rivastigmine on cognitive domains in mild-to-moderate Alzheimer's disease. Am J Alzheimers Dis Other Demen. 2010;25:347-52. [PMID: 20392862]  doi: 10.1177/1533317510365344
58.
Homma AAtarashi HKubota Net al. Efficacy and safety of sustained release donepezil high dose versus immediate release donepezil standard dose in Japanese patients with severe Alzheimer's disease: a randomized, double-blind trial. J Alzheimers Dis. 2016;52:345-57. [PMID: 26967222]  doi: 10.3233/JAD-151149
59.
Kano OIto HTakazawa Tet al. Clinically meaningful treatment responses after switching to galantamine and with addition of memantine in patients with Alzheimer's disease receiving donepezil. Neuropsychiatr Dis Treat. 2013;9:259-65. [PMID: 23431041]  doi: 10.2147/NDT.S40682
60.
Cummings JFroelich LBlack SEet al. Randomized, double-blind, parallel-group, 48-week study for efficacy and safety of a higher-dose rivastigmine patch (15 vs. 10 cm2) in Alzheimer's disease. Dement Geriatr Cogn Disord. 2012;33:341-53. [PMID: 22796905]  doi: 10.1159/000340056
61.
Peskind ERPotkin SGPomara Net al. Memantine treatment in mild to moderate Alzheimer disease: a 24-week randomized, controlled trial. Am J Geriatr Psychiatry. 2006;14:704-15. [PMID: 16861375]
62.
Lorenzi MBeltramello AMercuri NBet al. Effect of memantine on resting state default mode network activity in Alzheimer's disease. Drugs Aging. 2011;28:205-17. [PMID: 21250762]  doi: 10.2165/11586440-000000000-00000
63.
Porsteinsson APGrossberg GTMintzer Jet alMemantine MEM-MD-12 Study Group. Memantine treatment in patients with mild to moderate Alzheimer's disease already receiving a cholinesterase inhibitor: a randomized, double-blind, placebo-controlled trial. Curr Alzheimer Res. 2008;5:83-9. [PMID: 18288936]
64.
Tariot PNFarlow MRGrossberg GTet alMemantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291:317-24. [PMID: 14734594]
65.
Grossberg GTManes FAllegri RFet al. The safety, tolerability, and efficacy of once-daily memantine (28 mg): a multinational, randomized, double-blind, placebo-controlled trial in patients with moderate-to-severe Alzheimer's disease taking cholinesterase inhibitors. CNS Drugs. 2013;27:469-78. [PMID: 23733403]  doi: 10.1007/s40263-013-0077-7
66.
Scheltens PTwisk JWBlesa Ret al. Efficacy of Souvenaid in mild Alzheimer's disease: results from a randomized, controlled trial. J Alzheimers Dis. 2012;31:225-36. [PMID: 22766770]  doi: 10.3233/JAD-2012-121189
67.
Olde Rikkert MGVerhey FRBlesa Ret al. Tolerability and safety of Souvenaid in patients with mild Alzheimer's disease: results of multi-center, 24-week, open-label extension study. J Alzheimers Dis. 2015;44:471-80. [PMID: 25322923]  doi: 10.3233/JAD-141305
68.
Shah RCKamphuis PJLeurgans Set al. The S-Connect study: results from a randomized, controlled trial of Souvenaid in mild-to-moderate Alzheimer's disease. Alzheimers Res Ther. 2013;5:59. [PMID: 24280255]  doi: 10.1186/alzrt224
69.
Freund-Levi YEriksdotter-Jönhagen MCederholm Tet al. Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch Neurol. 2006;63:1402-8. [PMID: 17030655]
70.
Eriksdotter MVedin IFalahati Fet al. Plasma fatty acid profiles in relation to cognition and gender in Alzheimer's disease patients during oral omega-3 fatty acid supplementation: the OmegAD study. J Alzheimers Dis. 2015;48:805-12. [PMID: 26402079]  doi: 10.3233/JAD-150102
71.
Cornelli U. Treatment of Alzheimer's disease with a cholinesterase inhibitor combined with antioxidants. Neurodegener Dis. 2010;7:193-202. [PMID: 20224285]  doi: 10.1159/000295663
72.
De Jesus Moreno Moreno M. Cognitive improvement in mild to moderate Alzheimer's dementia after treatment with the acetylcholine precursor choline alfoscerate: a multicenter, double-blind, randomized, placebo-controlled trial. Clin Ther. 2003;25:178-93. [PMID: 12637119]
73.
Wade AGFarmer MHarari Get al. Add-on prolonged-release melatonin for cognitive function and sleep in mild to moderate Alzheimer's disease: a 6-month, randomized, placebo-controlled, multicenter trial. Clin Interv Aging. 2014;9:947-61. [PMID: 24971004]  doi: 10.2147/CIA.S65625
74.
Malpas CBVivash LGenc Set al. A phase IIa randomized control trial of VEL015 (sodium selenate) in mild-moderate Alzheimer's disease. J Alzheimers Dis. 2016;54:223-32. [PMID: 27447428]  doi: 10.3233/JAD-160544
75.
Gleason CEFischer BLDowling NMet al. Cognitive effects of soy isoflavones in patients with Alzheimer's disease. J Alzheimers Dis. 2015;47:1009-19. [PMID: 26401779]  doi: 10.3233/JAD-142958
76.
Modrego PJFayed NErrea JMet al. Memantine versus donepezil in mild to moderate Alzheimer's disease: a randomized trial with magnetic resonance spectroscopy. Eur J Neurol. 2010;17:405-12. [PMID: 19874395]  doi: 10.1111/j.1468-1331.2009.02816.x
77.
Thomas AIacono DBonanni Let al. Donepezil, rivastigmine, and vitamin E in Alzheimer disease: a combined P300 event-related potentials/neuropsychologic evaluation over 6 months. Clin Neuropharmacol. 2001;24:31-42. [PMID: 11290880]
78.
Yang YLiu JPFang JYet al. Effect and safety of Huannao Yicong formula in patients with mild-to-moderate Alzheimer's disease: a randomized, double-blinded, donepezil-controlled trial. Chin J Integr Med. 2019;25:574-81. [PMID: 30109588]  doi: 10.1007/s11655-018-3054-7
79.
Devanand DPMarder KMichaels KSet al. A randomized, placebo-controlled dose-comparison trial of haloperidol for psychosis and disruptive behaviors in Alzheimer's disease. Am J Psychiatry. 1998;155:1512-20. [PMID: 9812111]
80.
Ballard CMargallo-Lana MJuszczak Eet al. Quetiapine and rivastigmine and cognitive decline in Alzheimer's disease: randomised double blind placebo controlled trial. BMJ. 2005;330:874. [PMID: 15722369]
81.
De Deyn PJeste DVSwanink Ret al. Aripiprazole for the treatment of psychosis in patients with Alzheimer's disease: a randomized, placebo-controlled study. J Clin Psychopharmacol. 2005;25:463-7. [PMID: 16160622]
82.
Ballard CBanister CKhan Zet alADP Investigators. Evaluation of the safety, tolerability, and efficacy of pimavanserin versus placebo in patients with Alzheimer's disease psychosis: a phase 2, randomised, placebo-controlled, double-blind study. Lancet Neurol. 2018;17:213-22. [PMID: 29452684]  doi: 10.1016/S1474-4422(18)30039-5
83.
Ballard CYouakim JMCoate Bet al. Pimavanserin in Alzheimer's disease psychosis: efficacy in patients with more pronounced psychotic symptoms. J Prev Alzheimers Dis. 2019;6:27-33. [PMID: 30569083]  doi: 10.14283/jpad.2018.30
84.
Leonpacher AKPeters MEDrye LTet alCitAD Research Group. Effects of citalopram on neuropsychiatric symptoms in Alzheimer's dementia: evidence from the CitAD study. Am J Psychiatry. 2016;173:473-80. [PMID: 27032628]  doi: 10.1176/appi.ajp.2016.15020248
85.
Porsteinsson APDrye LTPollock BGet alCitAD Research Group, Effect of citalopram on agitation in Alzheimer disease: the CitAD randomized clinical trial. JAMA. 2014;311:682-91. [PMID: 24549548]  doi: 10.1001/jama.2014.93
86.
Zhou TWang JXin Cet al. Effect of memantine combined with citalopram on cognition of BPSD and moderate Alzheimer's disease: a clinical trial. Exp Ther Med. 2019;17:1625-30. [PMID: 30783429]  doi: 10.3892/etm.2018.7124
87.
Finkel SIMintzer JEDysken Met al. A randomized, placebo-controlled study of the efficacy and safety of sertraline in the treatment of the behavioral manifestations of Alzheimer's disease in outpatients treated with donepezil. Int J Geriatr Psychiatry. 2004;19:9-18. [PMID: 14716694]
88.
U.S. Food and Drug Administration. FDA drug safety communication: revised recommendations for Celexa (citalopram hydrobromide) related to a potential risk of abnormal heart rhythms with high doses. 28 March 2012. Accessed at www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-revised-recommendations-celexa-citalopram-hydrobromide-related on 29 November 2019.
89.
Howard RJJuszczak EBallard CGet alCALM-AD Trial Group. Donepezil for the treatment of agitation in Alzheimer's disease. N Engl J Med. 2007;357:1382-92. [PMID: 17914039]
90.
Tariot PNRaman RJakimovich Let alAlzheimer's Disease Cooperative Study. Divalproex sodium in nursing home residents with possible or probable Alzheimer disease complicated by agitation: a randomized, controlled trial. Am J Geriatr Psychiatry. 2005;13:942-9. [PMID: 16286437]
91.
Furukawa KTomita NUematsu Det al. Randomized double-blind placebo-controlled multicenter trial of yokukansan for neuropsychiatric symptoms in Alzheimer's disease. Geriatr Gerontol Int. 2017;17:211-8. [PMID: 26711658]  doi: 10.1111/ggi.12696
92.
U.S. Food and Drug Administration. Peripheral and Central Nervous System Drugs Advisory Committee Meeting. Department of Health and Human Services; 1989:227. (As cited in reference 93.)
93.
Molnar FJMan-Son-Hing MFergusson D. Systematic review of measures of clinical significance employed in randomized controlled trials of drugs for dementia. J Am Geriatr Soc. 2009;57:536-46. [PMID: 19187414]  doi: 10.1111/j.1532-5415.2008.02122.x
94.
Zanetti OSolerte SBCantoni F. Life expectancy in Alzheimer's disease (AD). Arch Gerontol Geriatr. 2009;49 Suppl 1:237-43. [PMID: 19836639]  doi: 10.1016/j.archger.2009.09.035
95.
Tom SEHubbard RACrane PKet al. Characterization of dementia and Alzheimer's disease in an older population: updated incidence and life expectancy with and without dementia. Am J Public Health. 2015;105:408-13. [PMID: 25033130]  doi: 10.2105/AJPH.2014.301935

Comments

0 Comments
Sign In to Submit A Comment

Information & Authors

Information

Published In

cover image Annals of Internal Medicine
Annals of Internal Medicine
Volume 172Number 1019 May 2020
Pages: 656 - 668

History

Published online: 28 April 2020
Published in issue: 19 May 2020

Keywords

Authors

Affiliations

Howard A. Fink, MD, MPH
Minneapolis VA Health Care System and University of Minnesota, Minneapolis, Minnesota (H.A.F., J.R.M., L.S.H., B.C.T., T.J.W.)
Eric J. Linskens, BS
Minneapolis VA Health Care System, Minneapolis, Minnesota (E.J.L., R.M., M.A.M.)
Roderick MacDonald, MS
Minneapolis VA Health Care System, Minneapolis, Minnesota (E.J.L., R.M., M.A.M.)
Pombie C. Silverman, BA
University of Iowa Carver College of Medicine, Iowa City, Iowa (P.C.S.)
J. Riley McCarten, MD
Minneapolis VA Health Care System and University of Minnesota, Minneapolis, Minnesota (H.A.F., J.R.M., L.S.H., B.C.T., T.J.W.)
Kristine M.C. Talley, PhD, RN
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Mary L. Forte, PhD, DC
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Priyanka J. Desai, MSPH
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Victoria A. Nelson, MSc
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Margaret A. Miller, MA
Minneapolis VA Health Care System, Minneapolis, Minnesota (E.J.L., R.M., M.A.M.)
Laura S. Hemmy, PhD
Minneapolis VA Health Care System and University of Minnesota, Minneapolis, Minnesota (H.A.F., J.R.M., L.S.H., B.C.T., T.J.W.)
Michelle Brasure, PhD, MSPH, MLIS
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Brent C. Taylor, MPH, PhD
Minneapolis VA Health Care System and University of Minnesota, Minneapolis, Minnesota (H.A.F., J.R.M., L.S.H., B.C.T., T.J.W.)
Weiwen Ng, MPH
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Jeannine M. Ouellette, MFA
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Kerry M. Sheets, MD
Hennepin Healthcare and Minneapolis VA Health Care System, Minneapolis, Minnesota (K.M.S.)
Timothy J. Wilt, MD, MPH
Minneapolis VA Health Care System and University of Minnesota, Minneapolis, Minnesota (H.A.F., J.R.M., L.S.H., B.C.T., T.J.W.)
Mary Butler, PhD, MBA
University of Minnesota, Minneapolis, Minnesota (K.M.T., M.L.F., P.J.D., V.A.N., M.B., W.N., J.M.O., M.B.)
Disclaimer: The findings and conclusions in this document are those of the authors, who are responsible for its contents; the findings and conclusions do not necessarily represent the views of AHRQ. Therefore, no statement in this report should be construed as an official position of AHRQ or the U.S. Department of Health and Human Services. AHRQ retains a license to display, reproduce, and distribute the data and the report from which the manuscript was derived under the terms of the agency's contract with the author.
Acknowledgment: The authors thank Cheryl Cole-Hill for administrative, technical, and logistic support.
Financial Support: This article is based on research conducted by the Minnesota Evidence-based Practice Center under contract HHSA 290-2015-00008-I to the AHRQ.
Disclosures: Drs. Fink, Desai, Taylor, and Sheets report that the evidence synthesis on which this manuscript was based was funded by a contract with AHRQ. Mr. Linskens reports that the project received funding from AHRQ. Drs. Talley and Brasure, Ms. Nelson, and Ms. Miller report grants from AHRQ during the conduct of the study. Dr. Hemmy reports support from AHRQ during the conduct of the study. Dr. Ng reports stock ownership in AmerisourceBergen, Cerner, and Express Scripts outside the submitted work. Dr. Wilt reports grants and personal fees from the AHRQ Evidence-based Practice Center program during the conduct of the study. Authors not named here have disclosed no conflicts of interest. Disclosures can also be viewed at www.acponline.org/authors/icmje/ConflictOfInterestForms.do?msNum=M19-3887.
Editors' Disclosures: Christine Laine, MD, MPH, Editor in Chief, reports that her spouse has stock options/holdings with Targeted Diagnostics and Therapeutics. Darren B. Taichman, MD, PhD, Executive Editor, reports that he has no financial relationships or interests to disclose. Cynthia D. Mulrow, MD, MSc, Senior Deputy Editor, reports that she has no relationships or interests to disclose. Jaya K. Rao, MD, MHS, Deputy Editor, reports that she has stock holdings/options in Eli Lilly and Pfizer. Christina C. Wee, MD, MPH, Deputy Editor, reports employment with Beth Israel Deaconess Medical Center. Sankey V. Williams, MD, Deputy Editor, reports that he has no financial relationships or interests to disclose. Yu-Xiao Yang, MD, MSCE, Deputy Editor, reports that he has no financial relationships or interest to disclose.
Reproducible Research Statement: Study protocol: Registered in PROSPERO (CRD42018117897) and available at https://effectivehealthcare.ahrq.gov/topics/alzheimers-type-dementia/protocol. Statistical code: Not available. Data set: The full technical report contains search strategies, flow diagrams, evidence tables, study quality assessment tables, and detailed results (https://effectivehealthcare.ahrq.gov/products/alzheimers-type-dementia/research).
Corresponding Author: Howard A. Fink, MD, MPH, Geriatric Research Education & Clinical Center, Minneapolis VA Health Care System (11-G), One Veterans Drive, Minneapolis, MN 55417; e-mail, [email protected].
Current Author Addresses: Drs. Fink, McCarten, and Hemmy; Mr. Linskens; and Ms. Miller: Geriatric Research Education & Clinical Center, Minneapolis VA Health Care System, 11-G, One Veterans Drive, Minneapolis, MN 55417.
Mr. MacDonald and Drs. Sheets and Wilt: Center for Care Delivery & Outcomes Research, Minneapolis VA Health Care System, 111-0, One Veterans Drive, Minneapolis, MN 55417.
Ms. Silverman: University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52246.
Dr. Talley: School of Nursing, University of Minnesota, 5-140 Weaver-Densford Hall, 308 Harvard Street SE, Minneapolis, MN 55455.
Dr. Forte: University of Minnesota, Division of Health Policy and Management, School of Public Health, MMC 729 (8729A), D330-4 Mayo Memorial Building, 420 Delaware Street SE, Minneapolis, MN 55455.
Ms. Desai, Ms. Nelson, Dr. Brasure, Mr. Ng, and Ms. Ouellette: University of Minnesota, Division of Health Policy and Management, School of Public Health, MMC 197, D330-6 Mayo Memorial Building, 420 Delaware Street SE, Minneapolis, MN 55455.
Dr. Taylor: Center for Care Delivery & Outcomes Research, Minneapolis VA Health Care System, 152, One Veterans Drive, Minneapolis, MN 55417.
Dr. Butler: University of Minnesota, Division of Health Policy and Management, School of Public Health, MMC 729, D381 Mayo Memorial Building, 420 Delaware Street SE, Minneapolis, MN 55455.
Author Contributions: Conception and design: H.A. Fink, J.R. McCarten, K.M.C. Talley, L.S. Hemmy, M. Brasure, T.J. Wilt, M. Butler.
Analysis and interpretation of the data: H.A. Fink, E.J. Linskens, R. MacDonald, P.C. Silverman, J.R. McCarten, K.M.C. Talley, M.L. Forte, P.J. Desai, V.A. Nelson, M.A. Miller, M. Brasure, B.C. Taylor, K.M. Sheets, T.J. Wilt, M. Butler.
Drafting of the article: H.A. Fink, E.J. Linskens, R. MacDonald, P.C. Silverman, J.R. McCarten, M.L. Forte, P.J. Desai, V.A. Nelson, M.A. Miller, M. Brasure, W. Ng.
Critical revision of the article for important intellectual content: H.A. Fink, E.J. Linskens, R. MacDonald, P.C. Silverman, J.R. McCarten, K.M.C. Talley, M.L. Forte, B.C. Taylor, J.M. Ouellette, K.M. Sheets, T.J. Wilt, M. Butler.
Final approval of the article: H.A. Fink, E.J. Linskens, R. MacDonald, P.C. Silverman, J.R. McCarten, K.M.C. Talley, M.L. Forte, P.J. Desai, V.A. Nelson, M.A. Miller, L.S. Hemmy, M. Brasure, B.C. Taylor, W. Ng, J.M. Ouellette, K.M. Sheets, T.J. Wilt, M. Butler.
Statistical expertise: H.A. Fink, R. MacDonald, B.C. Taylor, T.J. Wilt.
Obtaining of funding: H.A. Fink, T.J. Wilt, M. Butler.
Administrative, technical, or logistic support: H.A. Fink, P.J. Desai, V.A. Nelson, M.A. Miller, M. Brasure, T.J. Wilt, M. Butler.
Collection and assembly of data: H.A. Fink, E.J. Linskens, R. MacDonald, P.C. Silverman, K.M.C. Talley, M.L. Forte, P.J. Desai, V.A. Nelson, M.A. Miller, M. Brasure, W. Ng, K.M. Sheets, M. Butler.
This article was published at Annals.org on 28 April 2020.

Metrics & Citations

Metrics

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format





Download article citation data for:
Howard A. Fink, Eric J. Linskens, Roderick MacDonald, et al. Benefits and Harms of Prescription Drugs and Supplements for Treatment of Clinical Alzheimer-Type Dementia: A Systematic Review and Meta-analysis. Ann Intern Med.2020;172:656-668. [Epub 28 April 2020]. doi:10.7326/M19-3887

View More

Get Access

Login Options:
Purchase

You will be redirected to acponline.org to sign-in to Annals to complete your purchase.

Create your Free Account

You will be redirected to acponline.org to create an account that will provide access to Annals.

View options

PDF/ePub

View PDF/ePub

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media