Final Evidence Summary

Cognitive Impairment in Older Adults: Screening

February 25, 2020

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

By Carrie D. Patnode, PhD; Leslie A. Perdue, MPH; Rebecca C. Rossom, MD; Megan C. Rushkin, MPH; Nadia Redmond, MSPH; Rachel G. Thomas, MPH; Jennifer S. Lin, MD

The information in this article is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This article is intended as a reference and not as a substitute for clinical judgment.

This article may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied.

This article was published in JAMA on February 25, 2020 (JAMA. 2020;323(8):764-785. doi:10.1001/jama.2019.22258)

Return to Table of Contents

Importance: Early identification of cognitive impairment may improve patient and caregiver health outcomes.

Objective: To systematically review the test accuracy of cognitive screening instruments and benefits and harms of interventions to treat cognitive impairment in older adults (≥65 years) to inform the US Preventive Services Task Force.

Data Sources: MEDLINE, PubMed, PsycINFO, and Cochrane Central Register of Controlled Trials through January 2019, with literature surveillance through November 22, 2019.

Study Selection: Fair- to good-quality English-language studies of cognitive impairment screening instruments, and pharmacologic and nonpharmacologic treatments aimed at persons with mild cognitive impairment (MCI), mild to moderate dementia, or their caregivers.

Data Extraction and Synthesis: Independent critical appraisal and data abstraction; random-effects meta-analyses and qualitative synthesis.

Main Outcomes and Measures: Sensitivity, specificity; patient, caregiver, and clinician decision-making; patient function, quality of life, and neuropsychiatric symptoms; caregiver burden and well-being.

Results: The review included 287 studies with more than 280,000 older adults. One randomized clinical trial (RCT) (n = 4005) examined the direct effect of screening for cognitive impairment on patient outcomes, including potential harms, finding no significant differences in health-related quality of life at 12 months (effect size, 0.009 [95% CI, –0.063 to 0.080]). Fifty-nine studies (n = 38,531) addressed the accuracy of 49 screening instruments to detect cognitive impairment. The Mini-Mental State Examination was the most-studied instrument, with a pooled sensitivity of 0.89 (95% CI, 0.85 to 0.92) and specificity of 0.89 (95% CI, 0.85 to 0.93) to detect dementia using a cutoff of 23 or less or 24 or less (15 studies, n = 12,796). Two hundred twenty-four RCTs and 3 observational studies including more than 240,000 patients or caregivers addressed the treatment of MCI or mild to moderate dementia. None of the treatment trials were linked with a screening program; in all cases, participants were persons with known cognitive impairment. Medications approved to treat Alzheimer disease (donepezil, galantamine, rivastigmine, and memantine) improved scores on the ADAS-Cog 11 by 1 to 2.5 points over 3 months to 3 years. Psychoeducation interventions for caregivers resulted in a small benefit for caregiver burden (standardized mean difference, –0.24 [95% CI, –0.36 to –0.13) over 3 to 12 months. Intervention benefits were small and of uncertain clinical importance.

Conclusions and Relevance: Screening instruments can adequately detect cognitive impairment. There is no empirical evidence, however, that screening for cognitive impairment improves patient or caregiver outcomes or causes harm. It remains unclear whether interventions for patients or caregivers provide clinically important benefits for older adults with earlier detected cognitive impairment or their caregivers.

Return to Table of Contents

Dementia is a burdensome disease, not only to the health and longevity of individuals with the disease but also to their families and informal caregivers. According to the most recent Global Burden of Disease classification system, Alzheimer disease rose from the 12th most burdensome disease or injury in the United States in 1990 to the 6th in 2016 in terms of disability-adjusted life-years.1 It has been projected that by 2050 Alzheimer dementia will affect 13.8 million US residents.2 Early identification of cognitive impairment through screening would ideally allow patients and their families to receive care at an earlier stage in the disease process, potentially facilitating discussions regarding health, financial, and legal decision-making while the patient still retains decision-making capacity.

In 2014, the US Preventive Services Task Force (USPSTF) concluded that evidence was insufficient to assess the balance of benefits and harms of screening for cognitive impairment in older adults (I statement).3 The objective of this review was to inform an updated recommendation by the USPSTF.

Return to Table of Contents

Scope of Review

This review is an update of the 2013 review4,5 that supported the 2014 USPSTF recommendation. The update retained the analytic framework and key questions (KQs) that guided the 2013 review (Figure 1) and included studies published since the previous review, as well as studies from the previous review that met updated inclusion criteria. No substantive changes were made to the scope of the review for this update, other than to exclude the medication tacrine from the list of included interventions as it is no longer available in the United States. The full report is available at https://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/cognitive-impairment-in-older-adults-screening1. All main results presented in the full report are also presented in this article; more detailed methods and additional forest plots are included in the full report.

Data Sources and Searches

Ovid MEDLINE, PubMED (for publisher-supplied records only), PsycINFO, and the Cochrane Central Register of Controlled Trials were searched for relevant English-language literature. Searches encompassed literature published through January 2019. The searches were supplemented by examining the reference lists of other previously published reviews and primary studies and by suggestions from experts. ClinicalTrials.gov was searched for ongoing randomized clinical trials (RCTs) related to KQ1. Active surveillance was conducted through November 22, 2019, via article alerts and targeted journal searches to identify major studies that might affect the conclusions or understanding of the evidence. No new studies were identified.

Study Selection

Because of the large volume of search results, we first used a single-screen process (ie, 1 reviewer screened for exclusion) for records with terms clearly outside the scope of the review in the title or abstract (eg, “mice,” “HIV,” “brain injury”). Two independent reviewers then screened the titles and abstracts and relevant full-text articles to ensure consistency with a priori inclusion and exclusion criteria. For all KQs, studies that were relevant to community-dwelling, noninstitutionalized adults 65 years or older cared for in primary care in the United States were included. Only treatment studies (KQ4) conducted among community-dwelling older adults with mild cognitive impairment (MCI) or mild to moderate dementia were included (ie, those populations more representative of screen-detected older adults with cognitive impairment); studies of treatment of severe dementia were excluded.

For KQ1 and KQ3, we included RCTs and nonrandomized controlled studies that compared individuals who received screening with those who received no screening or usual care.

For KQ2, studies that evaluated any brief screening instrument that could be administered by a clinician in 10 minutes or less or self-administered in 20 minutes or less were included. Studies of screening performed by diagnostic imaging or biomarker testing were excluded. Studies needed to report sensitivity and specificity (or data needed to calculate them) of a screening test compared with a diagnostic reference standard (ie, clinical assessment or neuropsychological testing with explicit diagnostic criteria with or without expert consensus/conference). Case-control studies and studies that selectively recruited patients with known or clinically suspected dementia or MCI (or control patients with normal cognition) were excluded because of the high risk of bias in patient selection for these studies.

For treatment effectiveness (KQ4), studies were limited to RCTs or nonrandomized controlled intervention studies of major pharmacologic and nonpharmacologic interventions intended for use during the early and mild stages of cognitive impairment and aimed at improving patient cognition, physical function, quality of life (QOL), caregiver burden or well-being, or a combination of these. Interventions with a primary aim of improving patient behavioral and psychological symptoms of dementia (eg, agitation, aggression, depressive symptoms), improving markers of physical performance, or reducing falls were excluded. Studies reporting outcomes on decision-making for patients, families, or clinicians (eg, health care planning, including advance directives; safety planning; legal and financial planning); patient health outcomes (ie, mortality, health care utilization, institutionalization, global function, cognitive function, physical function, QOL, and neuropsychiatric symptoms including depression and anxiety); caregiver outcomes (ie, caregiving burden, symptoms of depression and anxiety, QOL); or societal outcomes (eg, automobile crashes) were included.

For harms (KQ5),we included all KQ4 RCTs as well as cohort or case-control studies with Ns 1000 or greater. Open-label extension data (unblinded and uncontrolled data collected on medication use subsequent to the blinded placebo-controlled phase of an RCT) were excluded because there were no comparison groups. Studies were required to report total adverse events, withdrawals attributable to adverse events, or serious adverse events that resulted in unexpected medical care, morbidity, or mortality.

Data Extraction and Quality Assessment

Two reviewers independently assessed the methodological quality of eligible studies. Disagreements were resolved by consensus and, if needed, consultation with a third reviewer. Each study was assigned a quality rating of “good,” “fair,” or “poor” according to the USPSTF study design–specific criteria.6 Studies rated as of poor quality because of serious methodological shortcomings were excluded.6 One reviewer abstracted descriptive and outcome data from fair- and good-quality studies into standardized evidence tables; a second checked for accuracy and completeness.

Data Synthesis and Analysis

For test accuracy studies (KQ2), the primary outcomes of interest were sensitivity and specificity. Results were synthesized by instrument type (according to length of administration as very brief [administered in ≤5 minutes], brief [administered in 6-10 minutes], or longer [self-administered in >10 minutes] instruments) and separated by screening for dementia, MCI and dementia, or MCI only. Only 1 instrument had adequate data to conduct a quantitative synthesis: the Mini-Mental State Examination (MMSE) at a cutoff of 23 or less or 24 or less to detect dementia. A bivariate model was used to model sensitivity and specificity simultaneously, thus accounting for the correlation between these variables. For other instruments, ranges of sensitivity and specificity are reported.

For treatment studies, the interventions were grouped into 4 broad categories: (1) US Food and Drug Administration (FDA)–approved medications to treat Alzheimer disease (ie, acetylcholinesterase inhibitors [AChEIs] and memantine); (2) other medications or dietary supplements (eg, nonsteroidal anti-inflammatory drugs, gonadal steroids, and vitamins); (3) nonpharmacologic interventions aimed primarily at the patient, including cognitive training, stimulation, and/or rehabilitation, exercise interventions, and multicomponent and other interventions; and (4) nonpharmacologic interventions aimed primarily at the caregiver or caregiver-patient dyad, including psychoeducation, care and case management, and other caregiver-focused interventions.

Meta-analyses were conducted on the most commonly reported outcomes for each body of evidence. As a result, pooled analyses were conducted for FDA-approved medications on global cognitive function outcomes, global function outcomes, and harms; for nonpharmacologic patient-level interventions on global cognitive function outcomes; and for caregiver and caregiver-patient dyad interventions on caregiver burden and caregiver depression measures. For consistency across the body of evidence, quantitative analyses focused on 6- to 12-month outcomes and included only shorter- or longer-term results when 6- or 12-month outcomes were not available.

Random-effects models using the DerSimonian and Laird method were used.7 For analyses with fewer than 10 studies, a sensitivity analysis was conducted using a more conservative restricted maximum likelihood analysis with the Knapp-Hartung correction.8 In cases in which continuous outcomes were measured using a variety of different instruments with differing scales (eg, caregiver burden), a standardized effect size (Hedges g) based on the differences in change between groups from baseline to follow-up was analyzed. A pooled risk ratio (for binary data) was used to analyze harms outcomes and improvement or maintenance in global function for AChEI and memantine interventions.

The presence of statistical heterogeneity among the studies was assessed using standard χ2 tests, and the magnitude of heterogeneity was estimated using the I2 statistic. For outcomes with 10 or more studies in the meta-analysis, funnel plots were generated and an Egger test was conducted to evaluate small study effects and potential publication bias.9,10

Stata version 15.1 (StataCorp LP) was used for all analyses. All significance testing was 2-sided, and results were considered statistically significant if the P value was 0.05 or less.

The aggregate strength of evidence was assessed for each KQ using the approach described in the Methods Guide for Effectiveness and Comparative Effectiveness Reviews,11 based on the number, quality, and size of studies and the consistency and precision of results between studies.

Return to Table of Contents

Investigators reviewed 11,645 unique citations and 967 full-text articles for all KQs (Figure 2). Overall, 287 studies including more than 285,000 older adults were included. Ninety-two studies were newly identified in this update and 195 were carried forward from the previous review. Fifty-nine studies that addressed the test accuracy of screening instruments (KQ2) were included, as well as another 224 RCTs and 3 observational studies that addressed the benefits and harms of screening or treatment (KQ1, KQ3, KQ4, and KQ5).

Benefits of Screening

Key Question 1. Does screening for cognitive impairment in community-dwelling older adults in primary care–relevant settings improve decision-making, patient-family/caregiver, or societal outcomes?

One RCT (IU CHOICE [conducted from October 2012 to September 2016]) examined the direct effect of screening for cognitive impairment on patient outcomes.12,13 This RCT was specifically designed and funded to address the lack of empirical data included in the previous USPSTF review. Primary care patients 65 years or older with no indication of cognitive impairment were randomized to screening for Alzheimer disease and related dementia (n = 2008) or no screening (n = 1997). Patients in the screening group were screened using the Memory Impairment Screen or the Mini-Cog and were referred for a voluntary diagnostic assessment if they screened positive on either or both tests. After a positive diagnostic assessment, a local memory care program worked with the caregivers and patients to provide or facilitate care and resources. Measures of health-related QOL using Health Utilities Index (HUI) scores (range, 0.36-1.00; 0 = dead and 1.00 = no impairment) were not significantly different between groups and across time. Among patients in the screening group, HUI scores were 0.67 (95% CI, 0.65 to 0.68) at baseline, 0.71 (95% CI, 0.69 to 0.72) at 1 month, 0.69 (95% CI, 0.67 to 0.71) at 6 months, and 0.68 (95% CI, 0.66 to 0.69) at 12 months. For those in the no screening group, HUI scores were not significantly different from scores in the screening group at all 4 time points (0.67 [95% CI, 0.66 to 0.69] at baseline, 0.69 [95% CI, 0.68 to 0.71] at 1 month, 0.70 [95% CI, 0.68 to 0.72] at 6 months, and 0.68 [95% CI, 0.66 to 0.70] at 12 months). Mixed-effects models showed no statistically significant differences between groups at any time point (eg, effect size at 12 months, 0.009 [95% CI, –0.063 to 0.080]). Furthermore, no significant differences in health care utilization, advance care planning, and dementia recognition by physicians were detected at 12 months.

Accuracy of Screening

Key Question 2. What is the accuracy of screening instruments to detect cognitive impairment in community-dwelling older adults?

Fifty-nine studies (n = 38,531) that addressed the test accuracy of screening for MCI or dementia were identified.14-72 The number of participants screened ranged from 46 to 8805. Among the included studies, the prevalence of cognitive impairment varied widely; dementia ranged from 1% to 47%, MCI ranged from 10% to 52%, and cognitive impairment (inclusive of MCI and dementia) ranged from 17% to 90%.

The reference standard used to diagnose dementia or MCI usually consisted of a neuropsychological battery of tests and often was supplemented by a clinical examination, laboratory testing, imaging, assessment of depression and physical function, and/or an informant interview. The reference standard was administered by research staff, neurologists, psychiatrists, psychologists, psychometricians, other physicians, and/or nurses, and the diagnosis was usually made by consensus. Diagnostic and Statistical Manual of Mental Disorders (Fourth Edition, Third Edition Revised, and Third Edition) criteria were most often used to diagnose dementia, sometimes in conjunction with National Institute of Neurological and Communicative Disorders and Stroke–Alzheimer's Disease and Related Disorders Association (NINCDS-ADRDA) criteria73 (for Alzheimer dementia) and National Institute for Neurological Disorders and Stroke–Association Internationale pour la Recherche et l'Enseignement en Neurosciences (NINDS-AIREN) criteria74 (for vascular dementia). No studies used Diagnostic and Statistical Manual of Mental Disorders (Fifth Edition) criteria. MCI was more variably diagnosed, with criteria including that from the International Working Group on MCI,75 performance 1 SD or more or 1.5 SD below normal, performance less than the 10th percentile on at least 1 cognitive test, a Clinical Dementia Rating scale score of 0.5, reported impairment that did not meet criteria for dementia, criteria developed by Petersen,76 criteria developed by a specific aging and disability resource center, or NINCDS-ADRDA criteria (for amnestic MCI).

Despite a large body of evidence examining cognitive screening instruments, most instruments were tested in only a few well-designed studies. The tests most likely relevant to screening in primary care are very brief instruments, with an administration time of 5 minutes or less. Eight very brief instruments were examined in more than 1 study (Clock Drawing Test, Lawton Instrumental Activities of Daily Living, Memory Impairment Screen, Mental State Questionnaire, Mini-Cog, verbal fluency tests, 8-item Interview to Differentiate Aging and Dementia [AD8], Functional Activities Questionnaire), with sensitivity to detect dementia usually at 0.75 or higher (range, 0.43-1.0) and specificity usually at 0.80 or higher (range, 0.54-1.0) (eFigure 1 in the Supplement). The MMSE, a brief test that takes 7 to 10 minutes to complete, was the most-studied instrument (32 studies). Pooled estimates across 15 studies (n = 12,796) resulted in a sensitivity of 0.89 (95% CI, 0.85 to 0.92) and a specificity of 0.89 (95% CI, 0.85 to 0.93) of the MMSE to detect dementia at a cutpoint of 23 or less or 24 or less. The test accuracy of the MMSE to detect MCI was based on a much smaller body of literature (13 studies) with a variety of cutoffs and resulted in less consistent estimates for test accuracy, with a range in sensitivity from 0.20 to 0.93 and range in specificity from 0.48 to 0.93. The test accuracy of 5 additional brief tests (7-Minute Screen, Abbreviated Mental Test, Montreal Cognitive Assessment, Saint Louis University Mental Status Examination, Telephone Interview for Cognitive Status) was reported in more than 1 study, with sensitivity to detect dementia ranging from 0.74 to 1.0 and specificity ranging from 0.65 to 0.96. The test performances of very brief and brief screening tests evaluated in only 1 study varied substantially. For self-administered, longer tests (>10 minutes), only 1 instrument (the Informant Questionnaire on Cognitive Decline in the Elderly) was assessed in more than 1 study, with sensitivity to detect dementia ranging from 0.80 to 0.88 and specificity ranging from 0.51 to 0.91. Across all instruments, test performance was generally higher in the detection of dementia vs MCI, although confidence intervals overlapped.

Harms of Screening

Key Question 3. What are the harms of screening for cognitive impairment in community-dwelling older adults?

The IU CHOICE RCT (n = 4005) compared symptoms of depression and anxiety among patients randomized to screening for dementia vs those randomized to no screening.12,13 At 1 month after screening, depressive symptoms (as measured by the Patient Health Questionnaire 9 [PHQ-9]) and anxiety symptoms (as measured by the Generalized Anxiety Disorder 7 [GAD-7]) were not significantly different between groups after adjusting for baseline values. A similar pattern was evident at 6 and 12 months as well, suggesting no significant differences in feelings of depression or anxiety after screening for dementia.

Benefits of Interventions

Key Question 4. Do interventions for mild to moderate dementia or MCI in community-dwelling older adults improve decisionmaking, patient, family/caregiver, or societal outcomes?

Two hundred twenty-four RCTs77-300 representing more than 50,000 patients, caregivers, or both and 3 cohort studies301-303 with more than 190,000 patients were identified that addressed the treatment or management of MCI or mild to moderate dementia (Table 1).

Acetylcholinesterase Inhibitors and Memantine

Based on 48 RCTs (n = 22,431) that evaluated AChEIs (ie, donepezil [18 RCTs; n = 6209], galantamine [10 RCTs; n = 7464], rivastigmine [8 RCTs; n = 4569]), and memantine (12 RCTs; n = 4189), these medications may improve measures of global cognitive function and global function in the short term (≤6 months’ follow-up), but the magnitude of change was small (Table 1).77,81,83,85,93,96,100,110,116,124,127,130,131,133,134,153,159,163,167,179,203,204,208,209,211,218,224-226,230,236,239-243,245,249, 252,266,275,286-290,294,295 In meta-analyses, the differences in changes between patients receiving AChEIs or memantine compared with those receiving placebo ranged from approximately 1 to 2.5 points on the Alzheimer Disease Assessment Scale–Cognitive 11 (ADAS-Cog-11; scale range, 0-70) (n = 10,994) and 0.5 to 1 point on the MMSE (scale range, 0-30) (n = 8589) over 3 months to 3 years of follow-up. AChEIs and memantine appeared to increase the likelihood of improving or maintaining patients’ global function by 15% (for memantine) to 50% (for rivastigmine) over 3 to 12 months (pooled 95% CI range, 0.49 to 2.69) (n = 8405); change at longer follow-up was not reported. Outcome measures of physical function were reported in only 60% of the studies and showed mixed results. Other important measures such as neuropsychiatric symptoms and rates of institutionalization were rarely reported; no medication studies included measures of QOL. Only 8 studies of medications examined outcomes beyond 6 months and generally found persistent effects that were consistent with shorter term outcomes.

Most of the available evidence on the effectiveness of FDA-approved medications came from studies involving people with dementia, particularly among those with moderate vs mild forms of dementia, most commonly Alzheimer disease. Four RCTs (n = 1919; mean age, 74 years) tested these medications in people with MCI; these studies, testing donepezil and memantine, showed no benefit on global cognitive function. Only 1 RCT (n = 769) reported on progression of MCI to Alzheimer disease, finding no significant differences in the rate of conversion between people receiving donepezil vs placebo at 3 years.

Other Medications and Supplements

Twenty-nine RCTs (n = 6489; mean age, 75 years) evaluated other medications or supplements, including antihypertensives, 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors (atorvastatin and simvastatin), nonsteroidal anti-inflammatory drugs (ibuprofen, naproxen, indomethacin, and celecoxib), gonadal steroids (estrogen [plus or minus progesterone] and testosterone), and dietary supplements and vitamins (multivitamins, B vitamins, vitamin E, and omega-3 fatty acids).78,79,114,120,121,127,132,138,155,156, 182,195,210,213,223,226,227,234,247,248,254-256,258,260,264,277,285,300 There was no consistent benefit on global cognition or physical function in people with mild to moderate dementia or MCI for any of these medications or supplements.

Nonpharmacologic Patient-Level Interventions

Sixty-one RCTs (n = 7847; mean age, 76 years) evaluated nonpharmacologic patient-level interventions, including cognitive-focused, exercise, and multicomponent and other interventions for people with MCI or dementia.80,82,84,88,90,91,94,98,101,102,104,106, 117,119,123,135,152,158,161,162,164-166,169-171,174,175,177,180,185,187-189,191,197,212, 215,217,219,220,222,229,232,233,235,237,244,251,253,257,263,265,270,272-274,278, 280,282,297 In general, these studies were quite small and of limited duration.

Among all interventions, there was no clear benefit on global or domain-specific measures of cognitive function compared with control conditions at 3 months to 2 years of follow-up among people with MCI or dementia (Table 1). Effect estimates generally favored intervention over control, but the magnitude of effect was inconsistent and had very wide confidence intervals (ranging from no effect to a large effect).

Although a pooled analysis of cognitive training, stimulation, and rehabilitation intervention studies found a small, statistically significant mean difference of 1.33 points on MMSE scores (95% CI, 0.29 to 2.37; 15 RCTs, n = 1341) favoring cognitive-focused interventions compared with control conditions at 3 to 12 months of follow-up, there was substantial clinical and statistical heterogeneity. Furthermore, combining 8 RCTs that reported changes in ADAS-Cog scores found a slightly greater improvement of 0.66 points (scale range, 0-70; higher scores indicate greater cognitive impairment) among intervention vs control group participants, but this difference was not statistically significant (mean difference, –0.66 [95% CI, –1.60 to 0.29]; n = 842). There was no evidence that the effect of the interventions was modified by study, population, or intervention characteristics and no evidence of longer-term (up to 2 years) effects on cognitive function. Physical function outcomes, including change in activities of daily living and instrumental activities of daily living, as well as QOL and mental and neuropsychiatric symptoms, were inconsistently reported. Cognitive training, stimulation, and rehabilitation interventions consistently resulted in very little change over time or in small and relatively equal decline in these measures from baseline to 3 months to 2 years across intervention and control groups, and few studies reported any statistically significant benefit.

For RCTs of exercise interventions, pooled, conservative estimates of differences in measures of global cognitive function showed no to small effects based on the MMSE (mean difference, 1.17 [95% CI, 0.45 to 1.90]; 10 studies, n = 1168) and ADAS-Cog (mean difference, –1.05 [95% CI, –1.60 to 0.29]; 6 studies, n = 1071) at 3 to 12 months (Table 1). There was, however, a pattern of effect for exercise interventions, with small improvements in measures of physical function and symptoms for intervention groups but declines for control groups. The clinical meaningfulness of these differences and the possibility of selective reporting limit the understanding of this finding. There was no consistent benefit of multicomponent and other patient-level interventions across outcomes.

Caregiver or Caregiver-Patient Dyad Interventions

Eighty-eight RCTs (n = 14,880; mean patient age, 78 years) evaluated the effect of multiple types of caregiver or caregiver-patient dyad interventions (Table 1).86,87,89,92,95,97,99,103,105,107-109,111-113, 115,118,122,125,126,128,129,136,137,139-151,154,157,160,168,172,173,176,178,181,183,184, 186,190,192-194,196,198-202,205-207,214,216,221,228,231,238,246,250,259,261, 262,267-269,271,276,279,281,283,284,291-293,296,298,299 Most randomized more than 100 caregivers or caregiver-patient dyads. About one-half of the studies followed up participants for 1 year or longer, and almost all focused on patients with moderate dementia. More than one-half targeted caregivers only, while the remaining trials targeted both the patient and caregiver or the entire family.

Overall, psychoeducation and care and case management interventions consistently benefited caregiver burden and depression outcomes. Effect sizes were mostly small, however, and of unclear clinical significance. Psychoeducation interventions resulted in a small but statistically significant benefit on caregiver burden at 3 to 12 months (standardized mean difference, –0.24 [95% CI, –0.36 to –0.13]; 27 RCTs, n = 2776; I2 = 50.2%) and in a medium effect on caregiver burden for care and/or case management interventions (standardized mean difference, –0.54 [95% CI, –0.96 to –0.12]; 8 RCTs, n = 1215; I2 = 82.9%) (Table 1). The clinical importance of these changes in self-reported caregiver burden scores is unclear, with standardized effects translating to a between-group difference of approximately 2 to 4 points on the 22-Item Zarit Burden Interview (Zarit-22; scale range, 0-88). Similar small effect sizes were seen for caregiver depression outcomes. The effect sizes of both caregiver depression and burden outcomes had wide confidence intervals, suggesting a range in the magnitude of benefit or, in some cases, a lack of benefit. There was no evidence in the meta-regression analyses that one type of intervention (psychoeducation vs care or case management vs other caregiver or caregiver-patient dyad interventions) was more effective than the others on measures of caregiver burden or caregiver depression. Likewise, there were no study, population, or intervention characteristics that consistently and robustly associated with larger effects on caregiver burden or depression outcomes.

Other outcomes such as caregiver or patient QOL, rates of or time to institutionalization, patient mental health and neuropsychiatric symptoms, and patient functional ability were sparsely reported across the studies, with no consistent evidence of benefit. Decision-making and preparation for meeting dementia-related needs were reported by only 1 RCT each (n = 414), with neither demonstrating statistically significant benefit.

Harms of Interventions

Key Question 5: What are the harms of interventions for mild to moderate dementia or MCI in community-dwelling older adults?

Acetylcholinesterase Inhibitors and Memantine

Overall, adverse effects from medications were quite common. Adverse events were reported in all 48 RCTs77,81,83,85,93,96,100,110,116, 124,127,130,131,133,134,153,159,163,167,179,203,204,208,209,211,218,224-226,230, 236, 239-243,245,249,252,266,275,286-290,294,295 (n = 22,431) in addition to 3 large observational studies301-303 (n = 190,076) (Table 1). Discontinuation was more common with AChEIs than with placebo (13% [donepezil and rivastigmine], 14% [galantamine], 8% [placebo]). Total adverse events were also statistically significantly higher for all 3 types of AChEI vs placebo. In studies that tested various doses of medications, there was some evidence of slightly higher total adverse events and withdrawals with higher doses (ie, 10 mg vs 5 mg [donepezil], 32 mg vs 24 mg [galantamine], and 6-12 mg vs 1-4 mg [rivastigmine]), although no formal tests of differences between these groups were reported. Memantine appeared to be better tolerated (8% withdrew), with no significant difference in discontinuation rates or total adverse events compared with placebo. Overall, there did not appear to be differences in total serious adverse events for these medications across studies with limited duration of follow-up. However, individual studies, including observational evidence, reported increased rates of bradycardia and, relatedly, of syncope, falls, and need for pacemaker placement among those exposed vs unexposed to AChEIs.

Other Medications and Supplements

Twenty-one of the RCTs (n = 5688) reported on harms, with harms not significantly increased in intervention groups compared with control groups.78,79,120,121,127,132,155,195,210,213,223,226,234,247,254-256,258,264,277,300

Nonpharmacologic Patient-Level Interventions

Little harm was evident in the few studies (12 RCTs, n = 2370) that reported harms.119,123,135,162,175,187,188,197,212,229,263,270

Caregiver or Caregiver-Patient Dyad Interventions

Only 4 RCTs (n = 486) reported monitoring harms, and no harms were evident.86,145,151,184

Return to Table of Contents

This review updates the 2013 USPSTF review on screening for cognitive impairment in older adults.4,5 A summary of findings, including an assessment of the strength of evidence for each key question, is presented in Table 2. To date, only 1 RCT has addressed the direct effect of screening for cognitive impairment and found no evidence of benefits or harms. As such, this review answers 2 primary questions: How well does screening detect dementia or MCI in primary care? and How effective are interventions to improve patient or caregiver outcomes in people with mild to moderate dementia or MCI? More than 260 studies were identified that addressed these questions; more than one-fourth of those studies were identified as part of this update. Despite the accumulation of new data, the conclusions for these key questions are essentially unchanged from the prior review.4,5

There is a large body of well-conducted test accuracy studies, but only a few instruments applicable to primary care have been examined in more than 1 study. Although the MMSE has the largest body of evidence to support its use and has adequate test accuracy, its utility is limited by the longer administration time (10-15 minutes) and cost (approximately $1.86 per form plus a test manual [$88], as of January 2020).304 Other instruments examined in at least 2 studies with adequate test performance to detect dementia among primary care–relevant populations include very brief instruments such as the Clock Drawing Test, the Memory Impairment Screen, the Mini-Cog, verbal fluency tests, the AD8, and the Functional Activities Questionnaire; brief instruments such as the Abbreviated Mental Test, Montreal Cognitive Assessment, 7-Minute Screen, and Saint Louis University Mental Status Examination; and the longer, self-administered Informant Questionnaire on Cognitive Decline in the Elderly.

One rationale for routine screening for cognitive impairment in older adults is facilitation of earlier diagnosis that may positively influence decision-making, leading to improved patient outcomes and reduced caregiver burden. This may include implementing medical, educational, and psychosocial interventions to suit individual patient and caregiver needs and encouraging patient participation in medical, legal, and financial decisions.305 While these are logical arguments, there is currently little empirical evidence, including qualitative evidence, to support them.306

Screening for cognitive impairment may have direct or indirect harms as a result of diagnostic inaccuracy (false-positive and false-negative results) or negative emotions and stigma that may arise with diagnosis.307,308 Recent systematic reviews regarding patients’ attitudes and preferences about screening for dementia found mixed evidence. Some studies suggested that patients have no concerns, whereas others suggested that few people would agree to routine screening for memory problems for reasons such as stigma.306,309 Evidence suggests that caregivers and the general public believe they will benefit from being screened for dementia, in part because they believe there are effective treatments and financial benefits.306,310,311

This review was not a comprehensive synthesis of all treatment and management options for people with cognitive impairment; instead, the focus was on selected interventions aimed at people with mild to moderate dementia or MCI. Based on the large body of evidence, there is support that AChEIs (donepezil, galantamine, and rivastigmine) and memantine and interventions that support caregivers, including care coordination, can result in small improvements in patient and caregiver health outcomes in the short term. The average effects of these benefits are quite small and likely not clinically significant. Any benefits are further limited by the commonly experienced adverse effects of medications and the limited availability of complex caregiver and care coordination interventions. Cognitive stimulation and training, exercise interventions, and other medications and supplements showed some favorable effects on patients’ cognitive and physical function, but study evidence lacked consistency and the estimates of benefit were imprecise.

Limitations

There is a lack of evidence around how screening for and treating MCI and early-stage dementia affects decision-making outcomes. Furthermore, there has been little reproducibility in testing specific screening instruments in primary care populations. The treatment literature is limited by a lack of consistency in the specific outcomes reported and short follow-up duration. It is difficult to interpret the clinical importance of the small average effects seen among treatment studies, and many measures likely have limited responsiveness for patients with less pronounced cognitive impairment. Consistent and standardized reporting of results according to meaningful thresholds of clinical significance would be helpful in interpreting the small average effects on continuous outcome measures. Other important measures such as QOL, physical function, and institutionalization were inconsistently reported.

Return to Table of Contents

Screening instruments can adequately detect cognitive impairment. There is no empirical evidence, however, that screening for cognitive impairment improves patient or caregiver outcomes or causes harm. It remains unclear whether interventions for patients or caregivers provide clinically important benefits for older adults with earlier detected cognitive impairment or their caregivers.

Return to Table of Contents

Source: This article was published in JAMA on February 25, 2020 (JAMA. 2020;323(8):764-785. doi:10.1001/jama.2019.22258)

Conflict of Interest Disclosures: Dr Rossom reported receiving grants from the National Institutes of Health and the US Food and Drug Administration. No other disclosures were reported.

Funding/Support: This research was funded under contract HHSA-290-2015-00007-I-EPC5, Task Order 3, from the Agency for Healthcare Research and Quality (AHRQ), US Department of Health and Human Services, under a contract to support the US Preventive Services Task Force (USPSTF).

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ had no role in study selection, quality assessment, or synthesis. AHRQ staff provided project oversight, reviewed the report to ensure that the analysis met methodological standards, and distributed the draft for peer review. Otherwise, AHRQ had no role in the conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript findings. The opinions expressed in this document are those of the authors and do not reflect the official position of AHRQ or the US Department of Health and Human Services.

Additional Information: A draft version of this evidence report underwent external peer review from 4 content experts (Amy Sanders, MD, SUNY Upstate Medical Center Department of Neurology; Deborah Barnes, PhD, University of California, San Francisco, Division of Psychiatry; Joseph Gaugler, PhD, University of Minnesota; Parminder Raina, PhD, McMaster University Institute for Research on Aging) and 2 federal partners (National Institute on Aging and National Institute of Mental Health). Comments from reviewers were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Return to Table of Contents

1. Mokdad AH, Ballestros K, Echko M, et al; US Burden of Disease Collaborators. The state of US health, 1990-2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444-1472. doi:10.1001/jama.2018.0158
2. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 2013;9(1):63-75.e2. doi:10.1016/j.jalz.2012.11.007
3. Moyer VA; U.S. Preventive Services Task Force. Screening for cognitive impairment in older adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2014;160(11):791-797. doi:10.7326/M14-0496
4. Lin JS, O’Connor E, Rossom RC, Perdue LA, Eckstrom E. Screening for cognitive impairment in older adults: a systematic review for the U.S. Preventive Services Task Force. Ann Intern Med. 2013;159(9):601-612. doi:10.7326/0003-4819-159-9-201311050-00730
5. Lin JS, O’Connor E, Rossom RC, et al. U.S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews. In: Agency for Healthcare Research and Quality, ed. Screening for Cognitive Impairment in Older Adults: An Evidence Update for the U.S. Preventive Services Task Force. Rockville, MD: Agency for Healthcare Research and Quality; 2013.
6. US Preventive Services Task Force. U.S. Preventive Services Task Force Procedure Manual. Rockville, MD: US Preventive Services Task Force; 2015.
7. DerSimonian R, Kacker R. Random-effects model for meta-analysis of clinical trials: an update. Contemp Clin Trials. 2007;28(2):105-114. doi:10.1016/j.cct.2006.04.004
8. Veroniki AA, Jackson D, ViechtbauerW, et al. Methods to estimate the between-study variance and its uncertainty inmeta-analysis. Res Synth Methods. 2016;7(1):55-79. doi:10.1002/jrsm.1164
9. Sterne JAC, Harbord RM. Funnel plots in meta-analysis. In: Sterne JAC, ed. Meta-Analysis in Stata: An Updated Collection from the Stata Journal. College Station, TX: Stata Press; 2009:109-123.
10. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315(7109):629-634. doi:10.1136/bmj.315.7109.629
11. Berkman ND, Lohr KN, Ansari M, et al. Grading the strength of a body of evidence when assessing health care interventions for the Effective Health Care Program of the Agency for Healthcare Research and Quality: an update. In: Agency for Healthcare Research and Quality, ed. Methods Guide for Effectiveness and Comparative Effectiveness Reviews. Rockville, MD: Agency for Healthcare Research and Quality; 2014:314-349. AHRQ publication 10(14)-EHC063-EF.
12. Fowler N, Perkins A, Gao S, Sachs G, Boustani M. Risks and benefits of screening for dementia in primary care: the Indiana University Cognitive Health Outcomes Investigation of the Comparative Effectiveness of Dementia Screening (IU CHOICE) trial [published online December 2, 2019]. J Am Geriatr Soc. doi:10.1111/jgs.16247
13. Fowler NR, Harrawood A, Frame A, et al. The Indiana University Cognitive Health Outcomes Investigation of the Comparative Effectiveness of Dementia Screening (CHOICE) study: study protocol for a randomized controlled trial. Trials. 2014;15:209. doi:10.1186/1745-6215-15-209
14. Grut M, Fratiglioni L, Viitanen M, Winblad B. Accuracy of the Mini-Mental Status Examination as a screening test for dementia in a Swedish elderly population. Acta Neurol Scand. 1993;87(4):312-317. doi:10.1111/j.1600-0404.1993.tb05514.x
15. Galvin JE, Roe CM, Powlishta KK, et al. The AD8: a brief informant interview to detect dementia. Neurology. 2005;65(4):559-564. doi:10.1212/01.wnl.0000172958.95282.2a
16. Reischies FM, Geiselmann B. Age-related cognitive decline and vision impairment affecting the detection of dementia syndrome in old age. Br J Psychiatry. 1997;171:449-451. doi:10.1192/bjp.171.5.449
17. Lee JY, Cho SJ, Na DL, et al; DongWoo Lee; Hong Jin Jeon; You Ra Lee; Maeng Je Cho. Brief screening for mild cognitive impairment in elderly outpatient clinic: validation of the Korean version of the Montreal Cognitive Assessment. J Geriatr Psychiatry Neurol. 2008;21(2):104-110. doi:10.1177/0891988708316855
18. Chan QL, Xu X, Shaik MA, et al. Clinical utility of the informant AD8 as a dementia case finding instrument in primary healthcare. J Alzheimers Dis. 2016;49(1):121-127. doi:10.3233/JAD-150390
19. Ehreke L, Luck T, Luppa M, König HH, Villringer A, Riedel-Heller SG. Clock drawing test - screening utility for mild cognitive impairment according to different scoring systems: results of the Leipzig Longitudinal Study of the Aged (LEILA 75+). Int Psychogeriatr. 2011;23(10):1592-1601. doi:10.1017/S104161021100144X
20. Lee KS, Kim EA, Hong CH, Lee DW, Oh BH, Cheong HK. Clock drawing test in mild cognitive impairment: quantitative analysis of four scoring methods and qualitative analysis. Dement Geriatr Cogn Disord. 2008;26(6):483-489. doi:10.1159/000167879
21. Kirby M, Denihan A, Bruce I, Coakley D, Lawlor BA. The clock drawing test in primary care: sensitivity in dementia detection and specificity against normal and depressed elderly. Int J Geriatr Psychiatry. 2001;16(10):935-940. doi:10.1002/gps.445
22. Kaufer DI, Williams CS, Braaten AJ, Gill K, Zimmerman S, Sloane PD. Cognitive screening for dementia and mild cognitive impairment in assisted living: comparison of 3 tests. J Am Med Dir Assoc. 2008;9(8):586-593. doi:10.1016/j.jamda.2008.05.006
23. McDowell I, Kristjansson B, Hill GB, Hébert R. Community screening for dementia: the Mini Mental State Exam (MMSE) and Modified Mini-Mental State Exam (3MS) compared. J Clin Epidemiol. 1997;50(4):377-383. doi:10.1016/S0895-4356(97)00060-7
24. Tariq SH, Tumosa N, Chibnall JT, Perry MH III, Morley JE. Comparison of the Saint Louis University mental status examination and the mini-mental state examination for detecting dementia and mild neurocognitive disorder—a pilot study. Am J Geriatr Psychiatry. 2006;14(11):900-910. doi:10.1097/01.JGP.0000221510.33817.86
25. Saxton J, Morrow L, Eschman A, Archer G, Luther J, Zuccolotto A. Computer assessment of mild cognitive impairment. Postgrad Med. 2009;121(2):177-185. doi:10.3810/pgm.2009.03.1990
26. Fuchs A, Wiese B, Altiner A, Wollny A, Pentzek M. Cued recall and other cognitive tasks to facilitate dementia recognition in primary care. J Am Geriatr Soc. 2012;60(1):130-135. doi:10.1111/j.1532-5415.2011.03765.x
27. Kay DW, Henderson AS, Scott R, Wilson J, Rickwood D, Grayson DA. Dementia and depression among the elderly living in the Hobart community: the effect of the diagnostic criteria on the prevalence rates. Psychol Med. 1985;15(4):771-788. doi:10.1017/S0033291700005006
28. Cruz-Orduña I, Bellón JM, Torrero P, et al. Detecting MCI and dementia in primary care: efficiency of the MMS, the FAQ and the IQCODE. Fam Pract. 2012;29(4):401-406. doi:10.1093/fampra/cmr114
29. Fong TG, Jones RN, Rudolph JL, et al. Development and validation of a brief cognitive assessment tool: the Sweet 16. Arch Intern Med. 2011;171(5):432-437. doi:10.1001/archinternmed.2010.423
30. Mao HF, Chang LH, Tsai AY, et al. Diagnostic accuracy of Instrumental Activities of Daily Living for dementia in community-dwelling older adults. Age Ageing. 2018;47(4):551-557. doi:10.1093/ageing/afy021
31. Juva K, Mäkelä M, Erkinjuntti T, et al. Functional assessment scales in detecting dementia. Age Ageing. 1997;26(5):393-400. doi:10.1093/ageing/26.5.393
32. Jorm AF, Broe GA, Creasy H, et al. Further data on the validity of the informant questionnaire on cognitive decline in the elderly (IQCODE). Int J Geriatr Psychiatry. 1996;11(2):131-139. doi:10.1002/(SICI)1099-1166(199602)11:2<131::AID-GPS294>3.0.CO;2-5
33. Brodaty H, Pond D, Kemp NM, et al. The GPCOG: a new screening test for dementia designed for general practice. J Am Geriatr Soc. 2002;50(3):530-534. doi:10.1046/j.1532-5415.2002.50122.x
34. Solomon PR, Brush M, Calvo V, et al. Identifying dementia in the primary care practice. Int Psychogeriatr. 2000;12(4):483-493. doi:10.1017/S1041610200006608
35. Hsu JL, Fan YC, Huang YL, et al. Improved predictive ability of the Montreal Cognitive Assessment for diagnosing dementia in a community-based study. Alzheimers Res Ther. 2015;7(1):69. doi:10.1186/s13195-015-0156-8
36. Borson S, Scanlan JM, Watanabe J, Tu SP, Lessig M. Improving identification of cognitive impairment in primary care. Int J Geriatr Psychiatry. 2006;21(4):349-355. doi:10.1002/gps.1470
37. Ayalon L. The IQCODE versus a single-item informant measure to discriminate between cognitively intact individuals and individuals with dementia or cognitive impairment. J Geriatr Psychiatry Neurol. 2011;24(3):168-173. doi:10.1177/0891988711418506
38. Ehreke L, Luppa M, Luck T, et al; AgeCoDe group. Is the clock drawing test appropriate for screening for mild cognitive impairment? results of the German study on Ageing, Cognition and Dementia in Primary Care Patients (AgeCoDe). Dement Geriatr Cogn Disord. 2009;28(4):365-372. doi:10.1159/000253484
39. Stein J, Luppa M, Kaduszkiewicz H, et al. Is the Short Form of the Mini-Mental State Examination (MMSE) a better screening instrument for dementia in older primary care patients than the original MMSE? results of the German study on Ageing, Cognition, and Dementia in Primary Care Patients (AgeCoDe). Psychol Assess. 2015;27(3):895-904. doi:10.1037/pas0000076
40. Vannier-Nitenberg C, Dauphinot V, Bongue B, et al. Performance of cognitive tests, individually and combined, for the detection of cognitive disorders amongst community-dwelling elderly people with memory complaints: the EVATEM study. Eur J Neurol. 2016;23(3):554-561. doi:10.1111/ene.12888
41. Ranson JM, Kuźma E, Hamilton W, Muniz-Terrera G, Langa KM, Llewellyn DJ. Predictors of dementia misclassification when using brief cognitive assessments. Neurol Clin Pract. 2019;9(2):109-117. doi:10.1212/CPJ.0000000000000566
42. Donnelly K, Donnelly JP, Cory E. Primary care screening for cognitive impairment in elderly veterans. Am J Alzheimers Dis Other Demen. 2008;23(3):218-226. doi:10.1177/1533317508315932
43. Markwick A, Zamboni G, de Jager CA. Profiles of cognitive subtest impairment in the Montreal Cognitive Assessment (MoCA) in a research cohort with normal Mini-Mental State Examination (MMSE) scores. J Clin Exp Neuropsychol. 2012;34(7):750-757. doi:10.1080/13803395.2012.672966
44. Wolf-Klein GP, Silverstone FA, Levy AP, Brod MS. Screening for Alzheimer’s disease by clock drawing. J Am Geriatr Soc. 1989;37(8):730-734. doi:10.1111/j.1532-5415.1989.tb02234.x
45. Kuslansky G, Buschke H, Katz M, Sliwinski M, Lipton RB. Screening for Alzheimer’s disease: the memory impairment screen versus the conventional three-word memory test. J Am Geriatr Soc. 2002;50(6):1086-1091. doi:10.1046/j.1532-5415.2002.50265.x
46. Rait G, Morley M, Burns A, Baldwin R, Chew-Graham C, St Leger AS. Screening for cognitive impairment in older African-Caribbeans. Psychol Med. 2000;30(4):957-963. doi:10.1017/S0033291799002305
47. Holsinger T, Plassman BL, Stechuchak KM, Burke JR, Coffman CJ, Williams JW Jr. Screening for cognitive impairment: comparing the performance of four instruments in primary care. J Am Geriatr Soc. 2012;60(6):1027-1036. doi:10.1111/j.1532-5415.2012.03967.x
48. Lipton RB, Katz MJ, Kuslansky G, et al. Screening for dementia by telephone using the memory impairment screen. J Am Geriatr Soc. 2003;51(10):1382-1390. doi:10.1046/j.1532-5415.2003.51455.x
49. Swearer JM, Drachman DA, Li L, Kane KJ, Dessureau B, Tabloski P. Screening for dementia in “real world” settings: the cognitive assessment screening test: CAST. Clin Neuropsychol. 2002;16(2):128-135. doi:10.1076/clin.16.2.128.13235
50. Cullen B, Fahy S, Cunningham CJ, et al. Screening for dementia in an Irish community sample using MMSE: a comparison of norm-adjusted versus fixed cut-points. Int J Geriatr Psychiatry. 2005;20(4):371-376. doi:10.1002/gps.1291
51. Waite LM, Broe GA, Casey B, et al. Screening for dementia using an informant interview. Neuropsychol Dev Cogn B Aging Neuropsychol Cogn. 1998;5(3):194-202. doi:10.1076/anec.5.3.194.614
52. Buschke H, Kuslansky G, Katz M, et al. Screening for dementia with the memory impairment screen. Neurology. 1999;52(2):231-238. doi:10.1212/WNL.52.2.231
53. Morales JM, Bermejo F, Romero M, Del-Ser T. Screening of dementia in community-dwelling elderly through informant report. Int J Geriatr Psychiatry. 1997;12(8):808-816. doi:10.1002/(SICI) 1099-1166(199708)12:8<808::AID-GPS644>3.0.CO;2-5
54. Lam LC, Tam CW, Lui VW, et al. Screening of mild cognitive impairment in Chinese older adults—a multistage validation of the Chinese abbreviated mild cognitive impairment test. Neuroepidemiology. 2008;30(1):6-12. doi:10.1159/000113300
55. Fillenbaum G, Heyman A, Williams K, Prosnitz B, Burchett B. Sensitivity and specificity of standardized screens of cognitive impairment and dementia among elderly black and white community residents. J Clin Epidemiol. 1990;43(7):651-660. doi:10.1016/0895-4356(90)90035-N
56. Kahle-Wrobleski K, Corrada MM, Li B, Kawas CH. Sensitivity and specificity of the mini-mental state examination for identifying dementia in the oldest-old: the 90+ study. J Am Geriatr Soc. 2007;55(2):284-289. doi:10.1111/j.1532-5415.2007.01049. x
57. Erkinjuntti T, Sulkava R, Wikström J, Autio L. Short Portable Mental Status Questionnaire as a screening test for dementia and delirium among the elderly. J Am Geriatr Soc. 1987;35(5):412-416. doi:10.1111/j.1532-5415.1987.tb04662.x
58. Hooijer C, Dinkgreve M, Jonker C, Lindeboom J, Kay DWK. Short screening tests for dementia in the elderly population, I: a comparison between AMTS, MMSE, MSQ and SPMSQ. Int J Geriatr Psychiatry. 1992;7(8):559-571. doi:10.1002/gps.930070805
59. Callahan CM, Unverzagt FW, Hui SL, Perkins AJ, Hendrie HC. Six-item screener to identify cognitive impairment among potential subjects for clinical research. Med Care. 2002;40(9):771-781. doi:10.1097/00005650-200209000-00007
60. Manly JJ, Schupf N, Stern Y, Brickman AM, Tang MX, Mayeux R. Telephone-based identification of mild cognitive impairment and dementia in a multicultural cohort. Arch Neurol. 2011;68(5):607-614. doi:10.1001/archneurol.2011.88
61. Cook SE, Marsiske M, McCoy KJ. The use of the Modified Telephone Interview for Cognitive Status (TICS-M) in the detection of amnestic mild cognitive impairment. J Geriatr Psychiatry Neurol. 2009;22(2):103-109. doi:10.1177/0891988708328214
62. Jeong SK, Cho KH, Kim JM. The usefulness of the Korean version of modified Mini-Mental State Examination (K-mMMSE) for dementia screening in community dwelling elderly people. BMC Public Health. 2004;4:31. doi:10.1186/1471-2458-4-31
63. Rideaux T, Beaudreau SA, Fernandez S, O’Hara R. Utility of the abbreviated Fuld Object Memory Evaluation and MMSE for detection of dementia and cognitive impairment not dementia in diverse ethnic groups. J Alzheimers Dis. 2012;31(2):371-386. doi:10.3233/JAD-2012-112180
64. Tokuhara KG, Valcour VG, Masaki KH, Blanchette PL. Utility of the Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE) for dementia in a Japanese-American population. Hawaii Med J. 2006;65(3):72-75.
65. Rait G, Burns A, Baldwin R, Morley M, Chew-Graham C, St Leger AS. Validating screening instruments for cognitive impairment in older South Asians in the United Kingdom. Int J Geriatr Psychiatry. 2000;15(1):54-62. doi:10.1002/(SICI)1099-1166(200001)15:1<54::AID-GPS77>3.0.CO;2-C
66. Ball LJ, Ogden A, Mandi D, Birge SJ. The validation of a mailed health survey for screening of dementia of the Alzheimer’s type. J Am Geriatr Soc. 2001;49(6):798-802. doi:10.1046/j.1532-5415.2001.49159.x
67. del Ser T, Sánchez-Sánchez F, García de Yébenes MJ, Otero A, Munoz DG. Validation of the seven-minute screen neurocognitive battery for the diagnosis of dementia in a Spanish population-based sample. Dement Geriatr Cogn Disord. 2006;22(5-6):454-464. doi:10.1159/000095858
68. Vercambre MN, Cuvelier H, Gayon YA, et al. Validation study of a French version of the modified telephone interview for cognitive status (F-TICS-m) in elderly women. Int J Geriatr Psychiatry. 2010;25(11):1142-1149. doi:10.1002/gps.2447
69. Heun R, Papassotiropoulos A, Jennssen F. The validity of psychometric instruments for detection of dementia in the elderly general population. Int J Geriatr Psychiatry. 1998;13(6):368-380. doi:10.1002/(SICI)1099-1166(199806)13:6<368::AIDGPS775> 3.0.CO;2-9
70. Ozer S, Noonan K, Burke M, et al. The validity of the Memory Alteration Test and the Test Your Memory test for community-based identification of amnestic mild cognitive impairment. Alzheimers Dement. 2016;12(9):987-995. doi:10.1016/j.jalz.2016 03.014
71. Gagnon M, Letenneur L, Dartigues JF, et al. Validity of the Mini-Mental State Examination as a screening instrument for cognitive impairment and dementia in French elderly community residents. Neuroepidemiology. 1990;9(3):143-150. doi:10.1159/000110764
72. Cummings-Vaughn LA, Chavakula NN, Malmstrom TK, Tumosa N, Morley JE, Cruz-Oliver DM. Veterans Affairs Saint Louis University Mental Status examination compared with the Montreal Cognitive Assessment and the Short Test of Mental Status. J Am Geriatr Soc. 2014;62(7):1341-1346. doi:10.1111/jgs.12874
73. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34(7):939-944. doi:10.1212/WNL.34.7.939
74. Román GC, Tatemichi TK, Erkinjuntti T, et al. Vascular dementia: diagnostic criteria for research studies. Report of the NINDS-AIREN International Workshop. Neurology. 1993;43(2):250-260. doi:10.1212/WNL.43.2.250
75. Winblad B, Palmer K, Kivipelto M, et al. Mild cognitive impairment—beyond controversies, towards a consensus: report of the International Working Group on Mild Cognitive Impairment. J Intern Med. 2004;256(3):240-246. doi:10.1111/j.1365-2796.2004.01380.x
76. Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern Med. 2004;256(3):183-194. doi:10.1111/j.1365-2796.2004.01388.x
77. Agid Y, Dubois B, Anand R, et al. Efficacy and tolerability of rivastigmine in patients with dementia of the Alzheimer type. Curr Ther Res Clin Exp. 1998;59:837-845. doi:10.1016/S0011-393X (98)85048-0
78. Aisen PS, Schafer KA, Grundman M, et al; Alzheimer’s Disease Cooperative Study. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;289(21):2819-2826. doi:10.1001/jama. 289.21.2819
79. Aisen PS, Schneider LS, Sano M, et al; Alzheimer Disease Cooperative Study. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA. 2008;300(15):1774-1783. doi:10.1001/jama.300.15.1774
80. Amieva H, Robert PH, Grandoulier AS, et al. Group and individual cognitive therapies in Alzheimer’s disease: the ETNA3 randomized trial. Int Psychogeriatr. 2016;28(5):707-717. doi:10.1017/S1041610215001830
81. Auchus AP, Brashear HR, Salloway S, Korczyn AD, De Deyn PP, Gassmann-Mayer C; GAL-INT-26 Study Group. Galantamine treatment of vascular dementia: a randomized trial. Neurology. 2007;69(5):448-458. doi:10.1212/01.wnl.0000266625.31615.f6
82. Bae S, Lee S, Lee S, et al. The effect of a multicomponent intervention to promote community activity on cognitive function in older adults with mild cognitive impairment: a randomized controlled trial. Complement Ther Med. 2019;42:164-169. doi:10.1016/j.ctim.2018.11.011
83. Bakchine S, Loft H. Memantine treatment in patients with mild to moderate Alzheimer’s disease: results of a randomised, double-blind, placebo-controlled 6-month study [republished from J Alzheimers Dis. 2007;11(4):471-479]. J Alzheimers Dis. 2008;13(1):97-107. doi:10.3233/JAD-2008-13110
84. Baker LD, Frank LL, Foster-Schubert K, et al. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch Neurol. 2010;67 (1):71-79. doi:10.1001/archneurol.2009.307
85. Ballard C, Sauter M, Scheltens P, et al. Efficacy, safety and tolerability of rivastigmine capsules in patients with probable vascular dementia: the VantagE study. Curr Med Res Opin. 2008;24(9):2561-2574. doi:10.1185/03007990802328142
86. Barnes CJ, Markham C. A pilot study to evaluate the effectiveness of an individualized and cognitive behavioural communication intervention for informal carers of people with dementia: the Talking Sense programme. Int J Lang Commun Disord. 2018;53(3):615-627. doi:10.1111/1460-6984.12375
87. Bass DM, Clark PA, Looman WJ, McCarthy CA, Eckert S. The Cleveland Alzheimer’s managed care demonstration: outcomes after 12 months of implementation. Gerontologist. 2003;43(1):73-85. doi:10.1093/geront/43.1.73
88. Bellantonio S, Kenny AM, Fortinsky RH, et al. Efficacy of a geriatrics team intervention for residents in dementia-specific assisted living facilities: effect on unanticipated transitions. J Am Geriatr Soc. 2008;56(3):523-528. doi:10.1111/j.1532-5415.2007.01591.x
89. Belle SH, Burgio L, Burns R, et al; Resources for Enhancing Alzheimer’s Caregiver Health (REACH) II Investigators. Enhancing the quality of life of dementia caregivers from different ethnic or racial groups: a randomized, controlled trial. Ann Intern Med. 2006;145(10):727-738. doi:10.7326/0003-4819-145-10-200611210-00005
90. Belleville S, Hudon C, Bier N, et al. MEMO+: efficacy, durability and effect of cognitive training and psychosocial intervention in individuals with mild cognitive impairment. J Am Geriatr Soc. 2018;66(4):655-663. doi:10.1111/jgs.15192
91. Bergamaschi S, Arcara G, Calza A, Villani D, Orgeta V, Mondini S. One-year repeated cycles of cognitive training (CT) for Alzheimer’s disease. Aging Clin Exp Res. 2013;25(4):421-426. doi:10.1007/s40520-013-0065-2
92. Berwig M, Heinrich S, Spahlholz J, Hallensleben N, Brähler E, Gertz HJ. Individualized support for informal caregivers of people with dementia—effectiveness of the German adaptation of REACH II. BMC Geriatr. 2017;17(1):286. doi:10.1186/s12877-017-0678-y
93. Black S, Román GC, Geldmacher DS, et al; Donepezil 307 Vascular Dementia Study Group. Efficacy and tolerability of donepezil in vascular dementia: positive results of a 24-week, multicenter, international, randomized, placebo-controlled clinical trial. Stroke. 2003;34(10):2323-2330. doi:10.1161/01.STR.0000091396.95360.E1
94. Blumenthal JA, Smith PJ, Mabe S, et al. Lifestyle and neurocognition in older adults with cognitive impairments: a randomized trial. Neurology. 2019;92(3):e212-e223. doi:10.1212/WNL.0000000000006784
95. Brennan PF, Moore SM, Smyth KA. The effects of a special computer network on caregivers of persons with Alzheimer’s disease. Nurs Res. 1995; 44(3):166-172. doi:10.1097/00006199-199505000-00007
96. Brodaty H, Corey-Bloom J, Potocnik FC, Truyen L, Gold M, Damaraju CR. Galantamine prolonged-release formulation in the treatment of mild to moderate Alzheimer’s disease. Dement Geriatr Cogn Disord. 2005;20(2-3):120-132. doi:10.1159/000086613
97. Bruvik FK, Allore HG, Ranhoff AH, Engedal K. The effect of psychosocial support intervention on depression in patients with dementia and their family caregivers: an assessor-blinded randomized controlled trial. Dement Geriatr Cogn Dis Extra. 2013;3(1):386-397. doi:10.1159/000355912
98. Burgener SC, Yang Y, Gilbert R, Marsh-Yant S. The effects of a multimodal intervention on outcomes of persons with early-stage dementia. Am J Alzheimers Dis Other Demen. 2008;23(4):382-394. doi:10.1177/1533317508317527
99. Burgio L, Stevens A, Guy D, Roth DL, HaleyWE. Impact of two psychosocial interventions on white and African American family caregivers of individuals with dementia. Gerontologist. 2003;43(4):568-579. doi:10.1093/geront/43.4.568
100. Burns A, Rossor M, Hecker J, et al. The effects of donepezil in Alzheimer’s disease—results from a multinational trial. Dement Geriatr Cogn Disord. 1999;10(3):237-244. doi:10.1159/000017126
101. Buschert VC, Friese U, Teipel SJ, et al. Effects of a newly developed cognitive intervention in amnestic mild cognitive impairment and mild Alzheimer’s disease: a pilot study. J Alzheimers Dis. 2011;25(4):679-694. doi:10.3233/JAD-2011-100999
102. Cahn-Weiner DA, Malloy PF, Rebok GW, Ott BR. Results of a randomized placebo-controlled study of memory training for mildly impaired Alzheimer’s disease patients. Appl Neuropsychol. 2003;10(4):215-223. doi:10.1207/s15324826an1004_3
103. Callahan CM, Boustani MA, Unverzagt FW, et al. Effectiveness of collaborative care for older adults with Alzheimer disease in primary care: a randomized controlled trial. JAMA. 2006;295(18):2148-2157. doi:10.1001/jama.295.18.2148
104. Cavallo M, Hunter EM, van der Hiele K, Angilletta C. Computerized structured cognitive training in patients affected by early-stage Alzheimer’s disease is feasible and effective: a randomized controlled study. Arch Clin Neuropsychol. 2016;31(8):868-876. doi:10.1093/arclin/acw072
105. Chang BL. Cognitive-behavioral intervention for homebound caregivers of persons with dementia. Nurs Res. 1999;48(3):173-182. doi:10.1097/00006199-199905000-00007
106. Chapman SB, Weiner MF, Rackley A, Hynan LS, Zientz J. Effects of cognitive-communication stimulation for Alzheimer’s disease patients treated with donepezil. J Speech Lang Hear Res. 2004;47(5):1149-1163. doi:10.1044/1092-4388(2004/085)
107. Charlesworth G, Shepstone L, Wilson E, et al. Befriending carers of people with dementia: randomised controlled trial. BMJ. 2008;336(7656):1295-1297. doi:10.1136/bmj.39549.548831.AE
108. Chien WT, Lee IY. Randomized controlled trial of a dementia care programme for families of home-resided older people with dementia. J Adv Nurs. 2011;67(4):774-787. doi:10.1111/j.1365-2648.2010.05537.x
109. Chien WT, Lee YM. A disease management program for families of persons in Hong Kong with dementia. Psychiatr Serv. 2008;59(4):433-436. doi:10.1176/ps.2008.59.4.433
110. Choi SH, Park KW, Na DL, et al; Expect Study Group. Tolerability and efficacy of memantine add-on therapy to rivastigmine transdermal patches in mild to moderate Alzheimer’s disease: a multicenter, randomized, open-label, parallel-group study. Curr Med Res Opin. 2011;27(7):1375-1383. doi:10.1185/03007995.2011.582484
111. Chu H, Yang CY, Liao YH, et al. The effects of a support group on dementia caregivers’ burden and depression. J Aging Health. 2011;23(2):228-241. doi: 10.1177/0898264310381522
112. Chu P, Edwards J, Levin R, Thomson J. The use of clinical case management for early state Alzheimer’s patients and their families. Am J Alzheimers Dis Other Demen. 2000;15(5):284-290. doi:10.1177/153331750001500506
113. Connell CM, Janevic MR. Effects of a telephone-based exercise intervention for dementia caregiving wives: a randomized controlled trial. J Appl Gerontol. 2009;28(2):171-194. doi:10.1177/0733464808326951
114. Connelly PJ, Prentice NP, Cousland G, Bonham J. A randomised double-blind placebo-controlled trial of folic acid supplementation of cholinesterase inhibitors in Alzheimer’s disease. Int J Geriatr Psychiatry. 2008;23(2):155-160. doi:10.1002/gps.1856
115. Coon DW, Thompson L, Steffen A, Sorocco K, Gallagher-Thompson D. Anger and depression management: psychoeducational skill training interventions for women caregivers of a relative with dementia. Gerontologist. 2003;43(5):678-689. doi:10.1093/geront/43.5.678
116. Corey-Bloom J, Anand R, Veach J. A randomized trial evaluating the efficacy and safety of ENA 713 (rivastigmine tartrate), a new acetylcholinesterase inhibitor, in patients with mild to moderately severe Alzheimer’s disease. Int J Geriatr Psychopharmacol. 1998;1(2):55-65.
117. Cove J, Jacobi N, Donovan H, Orrell M, Stott J, Spector A. Effectiveness of weekly cognitive stimulation therapy for people with dementia and the additional impact of enhancing cognitive stimulation therapy with a carer training program. Clin Interv Aging. 2014;9:2143-2150. doi:10.2147/CIA.S66232
118. Cristancho-Lacroix V, Wrobel J, Cantegreil-Kallen I, Dub T, Rouquette A, Rigaud AS. A web-based psychoeducational program for informal caregivers of patients with Alzheimer’s disease: a pilot randomized controlled trial. J Med Internet Res. 2015;17(5):e117. doi:10.2196/jmir.3717
119. Dawson NT. Examining the Effects of a Moderate-Intensity Home-Based Functional Exercise Intervention on Cognition and Function in Individuals With Dementia [dissertation]. Cleveland, OH: Cleveland State University; 2015. OhioLINK website. https://rave.ohiolink.edu/etdc/view?acc_num=csu1435768988. Accessed December 27, 2019.
120. de Jager CA, Oulhaj A, Jacoby R, Refsum H, Smith AD. Cognitive and clinical outcomes of homocysteine-lowering B-vitamin treatment in mild cognitive impairment: a randomized controlled trial. Int J Geriatr Psychiatry. 2012;27(6):592-600. doi:10.1002/gps.2758
121. de Jong D, Jansen R, Hoefnagels W, et al. No effect of one-year treatment with indomethacin on Alzheimer’s disease progression: a randomized controlled trial. PLoS One. 2008;3(1):e1475. doi:10.1371/journal.pone.0001475
122. de Rotrou J, Cantegreil I, Faucounau V, et al. Do patients diagnosed with Alzheimer’s disease benefit from a psycho-educational programme for family caregivers? a randomised controlled study. Int J Geriatr Psychiatry. 2011;26(8):833-842. doi:10.1002/gps.2611
123. Doi T, Verghese J, Makizako H, et al. Effects of cognitive leisure activity on cognition in mild cognitive impairment: results of a randomized controlled trial. J Am Med Dir Assoc. 2017;18(8):686-691. doi:10.1016/j.jamda.2017.02.013
124. Doody RS, Ferris SH, Salloway S, et al. Donepezil treatment of patients with MCI: a 48-week randomized, placebo-controlled trial. Neurology. 2009;72(18):1555-1561. doi:10.1212/01.wnl.0000344650.95823.03
125. Ducharme FC, Lévesque LL, Lachance LM, et al. “Learning to become a family caregiver” efficacy of an intervention program for caregivers following diagnosis of dementia in a relative. Gerontologist. 2011;51(4):484-494. doi:10.1093/geront/gnr014
126. Duggleby W, Ploeg J, McAiney C, et al. Web-based intervention for family carers of persons with dementia and multiple chronic conditions (My Tools 4 Care): pragmatic randomized controlled trial. J Med Internet Res. 2018;20(6):e10484. doi:10.2196/10484
127. Dysken MW, Sano M, Asthana S, et al. Effect of vitamin E and memantine on functional decline in Alzheimer disease: the TEAM-AD VA cooperative randomized trial. JAMA. 2014;311(1):33-44. doi:10.1001/jama.2013.282834
128. Eloniemi-Sulkava U, Notkola IL, Hentinen M, Kivelä SL, Sivenius J, Sulkava R. Effects of supporting community-living demented patients and their caregivers: a randomized trial. J Am Geriatr Soc. 2001;49(10):1282-1287. doi:10.1046/j.1532-5415.2001.49255.x
129. Eloniemi-Sulkava U, Saarenheimo M, Laakkonen ML, et al. Family care as collaboration: effectiveness of a multicomponent support program for elderly couples with dementia: randomized controlled intervention study. J Am Geriatr Soc. 2009;57(12):2200-2208. doi:10.1111/j.1532-5415.2009.02564.x
130. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer’s disease combined with cerebrovascular disease: a randomised trial. Lancet. 2002;359(9314):1283-1290. doi:10.1016/S0140-6736(02)08267-3
131. Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E; Donepezil MSAD Study Investigators Group. A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer’s disease [published correction appears in Neurology. 2001;57(11):2153]. Neurology. 2001;57(4):613-620. doi:10.1212/WNL.57.4.613
132. Feldman HH, Doody RS, Kivipelto M, et al; LEADe Investigators. Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: LEADe. Neurology. 2010;74(12):956-964. doi:10.1212/WNL.0b013e3181d6476a
133. Feldman HH, Lane R; Study 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(10):1056-1063. doi:10.1136/jnnp.2006.099424
134. Ferris S, Schneider L, Farmer M, Kay G, Crook T. A double-blind, placebo-controlled trial of memantine in age-associated memory impairment (memantine in AAMI). Int J Geriatr Psychiatry. 2007;22(5):448-455. doi:10.1002/gps.1711
135. Fiatarone Singh MA, Gates N, Saigal N, et al. The Study of Mental and Resistance Training (SMART) study—resistance training and/or cognitive training in mild cognitive impairment: a randomized, double-blind, double-sham controlled trial. J Am Med Dir Assoc. 2014;15(12):873-880. doi:10.1016/j.jamda.2014.09.010
136. Finkel S, Czaja SJ, Schulz R, Martinovich Z, Harris C, Pezzuto D. E-care: a telecommunications technology intervention for family caregivers of dementia patients. Am J Geriatr Psychiatry. 2007;15(5):443-448. doi:10.1097/JGP.0b013e3180437d87
137. Fortinsky RH, Kulldorff M, Kleppinger A, Kenyon-Pesce L. Dementia care consultation for family caregivers: collaborative model linking an Alzheimer’s association chapter with primary care physicians. Aging Ment Health. 2009;13(2):162-170. doi:10.1080/13607860902746160
138. Freund-Levi Y, Eriksdotter-Jönhagen M, Cederholm T, et 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(10):1402-1408. doi:10.1001/archneur.63.10.1402
139. FungWY, Chien WT. The effectiveness of a mutual support group for family caregivers of a relative with dementia. Arch Psychiatr Nurs. 2002;16(3):134-144. doi:10.1053/apnu.2002.32951
140. Gallagher-Thompson D, Coon DW, Solano N, Ambler C, Rabinowitz Y, Thompson LW. Change in indices of distress among Latino and Anglo female caregivers of elderly relatives with dementia: site-specific results from the REACH national collaborative study. Gerontologist. 2003;43(4):580-591. doi:10.1093/geront/43.4.580
141. Gallagher-Thompson D, Gray HL, Dupart T, Jimenez D, Thompson LW. Effectiveness of cognitive/behavioral small group intervention for reduction of depression and stress in non-Hispanic White and Hispanic/Latino women dementia family caregivers: outcomes and mediators of change. J Ration Emot Cogn Behav Ther. 2008;26(4):286-303. doi:10.1007/s10942-008-0087-4
142. Gallagher-Thompson D,Wang PC, Liu W, et al. Effectiveness of a psychoeducational skill training DVD program to reduce stress in Chinese American dementia caregivers: results of a preliminary study. Aging Ment Health. 2010;14(3):263-273. doi:10.1080/13607860903420989
143. Garand L, Rinaldo DE, Alberth MM, et al. Effects of problem solving therapy on mental health outcomes in family caregivers of persons with a new diagnosis of mild cognitive impairment or early dementia: a randomized controlled trial. Am J Geriatr Psychiatry. 2014;22(8):771-781. doi:10.1016/j.jagp.2013.07.007
144. Gaugler JE, Reese M, Mittelman MS. Effects of the NYU caregiver intervention-adult child on residential care placement. Gerontologist. 2013;53(6):985-997. doi:10.1093/geront/gns193
145. Gitlin LN, Arthur P, Piersol C, et al. Targeting behavioral symptoms and functional decline in dementia: a randomized clinical trial. J Am Geriatr Soc. 2018;66(2):339-345. doi:10.1111/jgs.15194
146. Gitlin LN, Corcoran M, Winter L, Boyce A, Hauck WW. A randomized, controlled trial of a home environmental intervention: effect on efficacy and upset in caregivers and on daily function of persons with dementia. Gerontologist. 2001;41(1):4-14. doi:10.1093/geront/41.1.4
147. Gitlin LN, Winter L, Burke J, Chernett N, Dennis MP, Hauck WW. Tailored activities to manage neuropsychiatric behaviors in persons with dementia and reduce caregiver burden: a randomized pilot study. Am J Geriatr Psychiatry. 2008;16(3):229-239. doi:10.1097/01.JGP.0000300629.35408.94
148. Gitlin LN, Winter L, Corcoran M, Dennis MP, Schinfeld S, Hauck WW. Effects of the home environmental skill-building program on the caregiver-care recipient dyad: 6-month outcomes from the Philadelphia REACH Initiative. Gerontologist. 2003;43(4):532-546. doi:10.1093/geront/43.4.532
149. Gitlin LN, Winter L, Dennis MP, Hodgson N, Hauck WW. A biobehavioral home-based intervention and the well-being of patients with dementia and their caregivers: the COPE randomized trial. JAMA. 2010;304(9):983-991. doi:10.1001/jama.2010.1253
150. Gitlin LN, Winter L, Dennis MP, Hodgson N, Hauck WW. Targeting and managing behavioral symptoms in individuals with dementia: a randomized trial of a nonpharmacological intervention. J Am Geriatr Soc. 2010;58(8):1465-1474. doi:10.1111/j.1532-5415.2010.02971.x
151. Graff MJ, Vernooij-Dassen MJ, Thijssen M, Dekker J, Hoefnagels WH, Rikkert MG. Community based occupational therapy for patients with dementia and their care givers: randomised controlled trial. BMJ. 2006;333(7580):1196. doi:10.1136/bmj.39001.688843.BE
152. Greenaway MC, Duncan NL, Smith GE. The memory support system for mild cognitive impairment: randomized trial of a cognitive rehabilitation intervention. Int J Geriatr Psychiatry. 2013;28(4):402-409. doi:10.1002/gps.3838
153. Hager K, Baseman AS, Nye JS, et al. Effects of galantamine in a 2-year, randomized, placebo-controlled study in Alzheimer’s disease. Neuropsychiatr Dis Treat. 2014;10:391-401.
154. Hébert R, Lévesque L, Vézina J, et al. Efficacy of a psychoeducative group program for caregivers of demented persons living at home: a randomized controlled trial. J Gerontol B Psychol Sci Soc Sci. 2003;58(1):S58-S67. doi:10.1093/geronb/58.1.S58
155. Henderson VW, Ala T, Sainani KL, et al. Raloxifene for women with Alzheimer disease: a randomized controlled pilot trial. Neurology. 2015;85(22):1937-1944. doi:10.1212/WNL.0000000000002171
156. Henderson VW, Paganini-Hill A, Miller BL, et al. Estrogen for Alzheimer’s disease in women: randomized, double-blind, placebo-controlled trial. Neurology. 2000;54(2):295-301. doi:10.1212/WNL.54.2.295
157. Hepburn KW, Lewis M, Narayan S, et al. Partners in Caregiving: a psychoeducation program affecting dementia family caregivers’ distress and caregiving outlook. Clin Gerontol. 2005;29(1):53-69. doi:10.1300/J018v29n01_05
158. Herrera C, Chambon C, Michel BF, Paban V, Alescio-Lautier B. Positive effects of computer-based cognitive training in adults with mild cognitive impairment. Neuropsychologia. 2012;50(8):1871-1881. doi:10.1016/j.neuropsychologia.2012.04.012
159. Herrmann N, Gauthier S, Boneva N, Lemming OM; 10158 Investigators. A randomized, double-blind, placebo-controlled trial of memantine in a behaviorally enriched sample of patients with moderate-to-severe Alzheimer’s disease. Int Psychogeriatr. 2013;25(6):919-927. doi:10.1017/S1041610213000239
160. Hirano A, Suzuki Y, Kuzuya M, Onishi J, Ban N, Umegaki H. Influence of regular exercise on subjective sense of burden and physical symptoms in community-dwelling caregivers of dementia patients: a randomized controlled trial. Arch Gerontol Geriatr. 2011;53(2):e158-e163. doi:10.1016/j.archger.2010.08.004
161. Ho RTH, Fong TCT, Chan WC, et al. Psychophysiological effects of Dance Movement Therapy and physical exercise on older adults with mild dementia: a randomized controlled trial. [published online November 28, 2018]. J Gerontol B Psychol Sci Soc Sci. 2018. doi:10.1093/geronb/gby145
162. Hoffmann K, Sobol NA, Frederiksen KS, et al. Moderate-to-high intensity physical exercise in patients with alzheimer’s disease: a randomized controlled trial. J Alzheimers Dis. 2016;50(2):443-453. doi:10.3233/JAD-150817
163. Holmes C, Wilkinson D, Dean C, et al. The efficacy of donepezil in the treatment of neuropsychiatric symptoms in Alzheimer disease. Neurology. 2004;63(2):214-219. doi:10.1212/01.WNL.0000129990.32253.7B
164. Holthoff VA, Marschner K, Scharf M, et al. Effects of physical activity training in patients with Alzheimer’s dementia: results of a pilot RCT study. PLoS One. 2015;10(4):e0121478. doi:10.1371/journal.pone.0121478
165. Hong SG, Kim JH, Jun TW. Effects of 12-week resistance exercise on electroencephalogram patterns and cognitive function in the elderly with mild cognitive impairment: a randomized controlled trial. Clin J Sport Med. 2018;28(6):500-508. doi:10.1097/JSM.0000000000000476
166. Hyer L, Scott C, Atkinson MM, et al. Cognitive training program to improve working memory in older adults with MCI. Clin Gerontol. 2016;39(5):410-427. doi:10.1080/07317115.2015.1120257
167. Ikeda M, Mori E, Matsuo K, Nakagawa M, Kosaka K. Donepezil for dementia with Lewy bodies: a randomized, placebo-controlled, confirmatory phase III trial. Alzheimers Res Ther. 2015;7(1):4. doi:10.1186/s13195-014-0083-0
168. Jansen AP, van Hout HP, Nijpels G, et al. Effectiveness of case management among older adults with early symptoms of dementia and their primary informal caregivers: a randomized clinical trial. Int J Nurs Stud. 2011;48(8):933-943. doi:10.1016/j.ijnurstu.2011.02.004
169. Jelcic N, Cagnin A, Meneghello F, Turolla A, Ermani M, Dam M. Effects of lexical-semantic treatment on memory in early Alzheimer disease: an observer-blinded randomized controlled trial. Neurorehabil Neural Repair. 2012;26(8):949-956. doi:10.1177/1545968312440146
170. Jeong JH, Na HR, Choi SH, et al. Group- and home-based cognitive intervention for patients with mild cognitive impairment: a randomized controlled trial. Psychother Psychosom. 2016;85(4):198-207. doi:10.1159/000442261
171. Jha A, Jan F, Gale T, Newman C. Effectiveness of a recovery-orientated psychiatric intervention package on the wellbeing of people with early dementia: a preliminary randomised controlled trial. Int J Geriatr Psychiatry. 2013;28(6):589-596. doi:10.1002/gps.3863
172. Joling KJ, van Marwijk HW, Smit F, et al. Does a family meetings intervention prevent depression and anxiety in family caregivers of dementia patients? a randomized trial. PLoS One. 2012;7(1):e30936. doi:10.1371/journal.pone.0030936
173. Judge KS, Yarry SJ, Looman WJ, Bass DM. Improved strain and psychosocial outcomes for caregivers of individuals with dementia: findings from Project ANSWERS. Gerontologist. 2013;53(2):280-292. doi:10.1093/geront/gns076
174. Kallio EL, Öhman H, Hietanen M, et al. Effects of cognitive training on cognition and quality of life of older persons with dementia. J Am Geriatr Soc. 2018;66(4):664-670. doi:10.1111/jgs.15196
175. Karssemeijer EGA, Aaronson JA, Bossers WJR, Donders R, Olde Rikkert MGM, Kessels RPC. The quest for synergy between physical exercise and cognitive stimulation via exergaming in people with dementia: a randomized controlled trial. Alzheimers Res Ther. 2019;11(1):3. doi:10.1186/s13195-018-0454-z
176. King AC, Baumann K, O’Sullivan P, Wilcox S, Castro C. Effects of moderate-intensity exercise on physiological, behavioral, and emotional responses to family caregiving: a randomized controlled trial. J Gerontol A Biol Sci Med Sci. 2002;57(1):M26-M36. doi:10.1093/gerona/57.1.M26
177. Kinsella GJ, Mullaly E, Rand E, et al. Early intervention for mild cognitive impairment: a randomised controlled trial. J Neurol Neurosurg Psychiatry. 2009;80(7):730-736. doi:10.1136/jnnp.2008.148346
178. Koivisto AM, Hallikainen I, Välimäki T, et al. Early psychosocial intervention does not delay institutionalization in persons with mild Alzheimer disease and has impact on neither disease progression nor caregivers’ well-being: ALSOVA 3-year follow-up. Int J Geriatr Psychiatry. 2016;31 (3):273-283. doi:10.1002/gps.4321
179. Krishnan KR, Charles HC, Doraiswamy PM, et 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(11):2003-2011. doi:10.1176/appi.ajp.160.11.2003
180. Kurz A, Thöne-Otto A, Cramer B, et al. CORDIAL: cognitive rehabilitation and cognitive-behavioral treatment for early dementia in Alzheimer disease: a multicenter, randomized, controlled trial. Alzheimer Dis Assoc Disord. 2012;26(3):246-253. doi:10.1097/WAD.0b013e318231e46e
181. Kurz A, Wagenpfeil S, Hallauer J, Schneider-Schelte H, Jansen S; AENEAS Study. Evaluation of a brief educational program for dementia carers: the AENEAS study. Int J Geriatr Psychiatry. 2010;25(8):861-869. doi:10.1002/gps.2428
182. Kwok T, Lee J, Law CB, et al. A randomized placebo controlled trial of homocysteine lowering to reduce cognitive decline in older demented people. Clin Nutr. 2011;30(3):297-302. doi:10.1016/j.clnu.2010.12.004
183. Kwok T, Wong B, Ip I, Chui K, Young D, Ho F. Telephone-delivered psychoeducational intervention for Hong Kong Chinese dementia caregivers: a single-blinded randomized controlled trial. Clin Interv Aging. 2013;8:1191-1197. doi:10.2147/CIA.S48264
184. Laakkonen ML, Kautiainen H, Hölttä E, et al. Effects of self-management groups for people with dementia and their spouses—randomized controlled trial. J Am Geriatr Soc. 2016;64(4):752-760. doi:10.1111/jgs.14055
185. Lam LC, Chau RC, Wong BM, et al. Interim follow-up of a randomized controlled trial comparing Chinese style mind body (Tai Chi) and stretching exercises on cognitive function in subjects at risk of progressive cognitive decline. Int J Geriatr Psychiatry. 2011;26(7):733-740. doi:10.1002/gps.2602
186. Lam LC, Lee JS, Chung JC, Lau A, Woo J, Kwok TC. A randomized controlled trial to examine the effectiveness of case management model for community dwelling older persons with mild dementia in Hong Kong. Int J Geriatr Psychiatry. 2010;25(4):395-402. doi:10.1002/gps.2352
187. Lamb SE, Sheehan B, Atherton N, et al; DAPA Trial Investigators. Dementia And Physical Activity (DAPA) trial of moderate to high intensity exercise training for people with dementia: randomised controlled trial. BMJ. 2018;361:k1675. doi:10.1136/bmj.k1675
188. Lautenschlager NT, Cox KL, Flicker L, et al. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial [published correction appears in JAMA. 2009;301(3):276]. JAMA. 2008;300(9):1027-1037. doi:10.1001/jama.300.9.1027
189. Lazarou I, Parastatidis T, Tsolaki A, et al. International Ballroom Dancing Against Neurodegeneration: a randomized controlled trial in Greek community-dwelling elders with mild cognitive impairment. Am J Alzheimers Dis Other Demen. 2017;32(8):489-499. doi:10.1177/1533317517725813
190. Leach MJ, Francis A, Ziaian T. Transcendental Meditation for the improvement of health and wellbeing in community-dwelling dementia caregivers [TRANSCENDENT]: a randomised wait-list controlled trial. BMC Complement Altern Med. 2015;15:145. doi:10.1186/s12906-015-0666-8
191. Liu-Ambrose T, Best JR, Davis JC, et al. Aerobic exercise and vascular cognitive impairment: a randomized controlled trial. Neurology. 2016;87(20):2082-2090. doi:10.1212/WNL.0000000000003332
192. Livingston G, Barber J, Rapaport P, et al. Clinical effectiveness of a manual based coping strategy programme (START, STrAtegies for RelaTives) in promoting the mental health of carers of family members with dementia: pragmatic randomised controlled trial. BMJ. 2013;347:f6276. doi:10.1136/bmj.f6276
193. Logiudice D, Waltrowicz W, Brown K, Burrows C, Ames D, Flicker L. Do memory clinics improve the quality of life of carers? a randomized pilot trial. Int J Geriatr Psychiatry. 1999;14(8):626-632. doi:10.1002/(SICI)1099-1166(199908)14:8<626::AIDGPS990>3.0.CO;2-5
194. Losada A, Márquez-González M, Romero-Moreno R. Mechanisms of action of a psychological intervention for dementia caregivers: effects of behavioral activation and modification of dysfunctional thoughts. Int J Geriatr Psychiatry. 2011;26(11):1119-1127.
195. Lu PH, Masterman DA, Mulnard R, et al. Effects of testosterone on cognition and mood in male patients with mild Alzheimer disease and healthy elderly men. Arch Neurol. 2006;63(2):177-185. doi:10.1001/archneur.63.2.nct50002
196. Marriott A, Donaldson C, Tarrier N, Burns A. Effectiveness of cognitive-behavioural family intervention in reducing the burden of care in carers of patients with Alzheimer’s disease. Br J Psychiatry. 2000;176:557-562. doi:10.1192/bjp.176.6.557
197. Marshall A, Spreadbury J, Cheston R, et al. A pilot randomised controlled trial to compare changes in quality of life for participants with early diagnosis dementia who attend a “Living Well with Dementia” group compared to waiting-list control. Aging Ment Health. 2015;19(6):526-535. doi:10.1080/13607863.2014.954527
198. Martín-Carrasco M, Domínguez-Panchón AI, González-Fraile E, Muñoz-Hermoso P, Ballesteros J; EDUCA Group. Effectiveness of a psychoeducational intervention group program in the reduction of the burden experienced by caregivers of patients with dementia: the EDUCA-II randomized trial. Alzheimer Dis Assoc Disord. 2014;28(1):79-87. doi:10.1097/WAD.0000000000000003
199. Martín-Carrasco M, Martín MF, Valero CP, et al. Effectiveness of a psychoeducational intervention program in the reduction of caregiver burden in Alzheimer’s disease patients’ caregivers. Int J Geriatr Psychiatry. 2009;24(5):489-499. doi:10.1002/gps.2142
200. Martin-Cook K, Davis BA, Hynan LS, Weiner MF. A randomized, controlled study of an Alzheimer’s caregiver skills training program. Am J Alzheimers Dis Other Demen. 2005;20(4):204-210. doi:10.1177/153331750502000411
201. Martindale-Adams J, Nichols LO, Burns R, Graney MJ, Zuber J. A trial of dementia caregiver telephone support. Can J Nurs Res. 2013;45(4):30-48. doi:10.1177/084456211304500404
202. Mavandadi S,Wright EM, Graydon MM, Oslin DW, Wray LO. A randomized pilot trial of a telephone-based collaborative care management program for caregivers of individuals with dementia. Psychol Serv. 2017;14(1):102-111. doi:10.1037/ser0000118
203. Mazza M, Capuano A, Bria P, Mazza S. 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(9):981-985. doi:10.1111/j.1468-1331.2006.01409.x
204. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):2031-2036. doi:10.1016/S0140-6736(00)03399-7
205. Meeuwsen EJ, Melis RJ, Van Der Aa GC, et al. Effectiveness of dementia follow-up care by memory clinics or general practitioners: randomised controlled trial. BMJ. 2012;344:e3086. doi:10.1136/bmj.e3086
206. Menn P, Holle R, Kunz S, et al. Dementia care in the general practice setting: a cluster randomized trial on the effectiveness and cost impact of three management strategies. Value Health. 2012;15(6):851-859. doi:10.1016/j.jval.2012.06.007
207. Mittelman MS, Roth DL, Coon DW, Haley WE. Sustained benefit of supportive intervention for depressive symptoms in caregivers of patients with Alzheimer’s disease. Am J Psychiatry. 2004;161(5):850-856. doi:10.1176/appi.ajp.161.5.850
208. Mohs RC, Doody RS, Morris JC, et al; “312” Study Group. A 1-year, placebo-controlled preservation of function survival study of donepezil in AD patients. Neurology. 2001;57(3):481-488. doi:10.1212/WNL.57.3.481
209. Mok V, Wong A, Ho S, Leung T, Lam WW, Wong KS. Rivastigmine in Chinese patients with subcortical vascular dementia. Neuropsychiatr Dis Treat. 2007;3(6):943-948.
210. Moonen JE, Foster-Dingley JC, de Ruijter W, et al. Effect of discontinuation of antihypertensive treatment in elderly people on cognitive functioning—the DANTE Study Leiden: a randomized clinical trial [published correction appears in JAMA Intern Med. 2016;176(2):284]. JAMA Intern Med. 2015;175(10):1622-1630. doi:10.1001/jamainternmed.2015.4103
211. Mori E, Ikeda M, Kosaka K; Donepezil-DLB Study Investigators. Donepezil for dementia with Lewy bodies: a randomized, placebo-controlled trial. Ann Neurol. 2012;72(1):41-52. doi:10.1002/ana.23557
212. Morris JK, Vidoni ED, Johnson DK, et al. Aerobic exercise for Alzheimer’s disease: a randomized controlled pilot trial. PLoS One. 2017;12(2):e0170547. doi:10.1371/journal.pone.0170547
213. Mulnard RA, Cotman CW, Kawas C, et al. Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer’s Disease Cooperative Study [published correction appears in JAMA. 2000;284(20):2597]. JAMA. 2000;283(8):1007-1015. doi:10.1001/jama.283.8.1007
214. Nourhashemi F, Andrieu S, Gillette-Guyonnet S, et al; PLASA Group. Effectiveness of a specific care plan in patients with Alzheimer’s disease: cluster randomised trial (PLASA study). BMJ. 2010;340:c2466. doi:10.1136/bmj.c2466
215. Nousia A, Siokas V, Aretouli E, et al. Beneficial effect of multidomain cognitive training on the neuropsychological performance of patients with early-stage Alzheimer’s disease. Neural Plast. 2018;2018:2845176. doi:10.1155/2018/2845176
216. Núñez-Naveira L, Alonso-Búa B, de Labra C, et al. UnderstAID, an ICT platform to help informal caregivers of people with dementia: a pilot randomized controlled study. Biomed Res Int. 2016;2016:5726465. doi:10.1155/2016/5726465
217. Olazarán J, Muñiz R, Reisberg B, et al. Benefits of cognitive-motor intervention in MCI and mild to moderate Alzheimer disease. Neurology. 2004; 63(12):2348-2353. doi:10.1212/01.WNL.0000147478.03911.28
218. Orgogozo JM, Rigaud AS, Stöffler A, Möbius HJ, Forette F. Efficacy and safety of memantine in patients with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke. 2002;33(7):1834-1839. doi:10.1161/01.STR.0000020094.08790.49
219. Orrell M, Aguirre E, Spector A, et al. Maintenance cognitive stimulation therapy for dementia: single-blind, multicentre, pragmatic randomised controlled trial. Br J Psychiatry. 2014;204(6):454-461. doi:10.1192/bjp.bp.113.137414
220. Orrell M, Yates L, Leung P, et al. The impact of individual Cognitive Stimulation Therapy (iCST) on cognition, quality of life, caregiver health, and family relationships in dementia: a randomised controlled trial. PLoS Med. 2017;14(3):e1002269. doi:10.1371/journal.pmed.1002269
221. Ostwald SK, Hepburn KW, Caron Caron W, Burns T, Mantell R. Reducing caregiver burden: a randomized psychoeducational intervention for caregivers of persons with dementia. Gerontologist. 1999;39(3):299-309. doi:10.1093/geront/39.3.299
222. Pantoni L, Poggesi A, Diciotti S, et al. Effect of attention training in mild cognitive impairment patients with subcortical vascular changes: the RehAtt study. J Alzheimers Dis. 2017;60(2):615-624. doi:10.3233/JAD-170428
223. Pasqualetti P, Bonomini C, Dal Forno G, et al. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clin Exp Res. 2009;21(2):102-110. doi:10.1007/BF03325217
224. Peskind ER, Potkin SG, Pomara N, et al. Memantine treatment in mild to moderate Alzheimer disease: a 24-week randomized, controlled trial. Am J Geriatr Psychiatry. 2006;14 (8):704-715. doi:10.1097/01.JGP.0000224350.82719.83
225. Peters O, FuentesM, Joachim LK, et al. Combined treatment with memantine and galantamine-CR compared with galantamine-CR only in antidementia drug naïve patients with mild-to-moderate Alzheimer’s disease. Alzheimers Dement (NY). 2015;1(3):198-204. doi:10.1016/j.trci.2015.10.001
226. Petersen RC, Thomas RG, Grundman M, et al; Alzheimer’s Disease Cooperative Study Group. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352(23):2379-2388. doi:10.1056/NEJMoa050151
227. Phillips MA, Childs CE, Calder PC, Rogers PJ. No effect of omega-3 fatty acid supplementation on cognition and mood in individuals with cognitive impairment and probable Alzheimer’s disease: a randomised controlled trial. Int J Mol Sci. 2015;16(10):24600-24613. doi:10.3390/ijms161024600
228. Pillemer K, Jill SJ. Peer support for Alzheimer’s caregivers: is it enough to make a difference? Res Aging. 2002;24(2):171-192. doi:10.1177/0164027502242001
229. Pitkälä KH, Pöysti MM, Laakkonen ML, et al. Effects of the Finnish Alzheimer Disease Exercise Trial (FINALEX): a randomized controlled trial. JAMA Intern Med. 2013;173(10):894-901. doi:10.1001/jamainternmed.2013.359
230. Porsteinsson AP, Grossberg GT, Mintzer J, Olin JT; Memantine 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(1):83-89. doi:10.2174/156720508783884576
231. Prick AE, de Lange J, Twisk J, Pot AM. The effects of a multi-component dyadic intervention on the psychological distress of family caregivers providing care to people with dementia: a randomized controlled trial. Int Psychogeriatr. 2015;27(12):2031-2044. doi:10.1017/S104161021500071X
232. Quayhagen MP, Quayhagen M, Corbeil RR, Roth PA, Rodgers JA. A dyadic remediation program for care recipients with dementia. Nurs Res. 1995;44(3):153-159. doi:10.1097/00006199-199505000-00005
233. Quinn C, Toms G, Jones C, et al. A pilot randomized controlled trial of a self-management group intervention for people with early-stage dementia (the SMART study). Int Psychogeriatr. 2016;28(5):787-800. doi:10.1017/ S1041610215002094
234. Quinn JF, Raman R, Thomas RG, et al. Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: a randomized trial. JAMA. 2010;304(17):1903-1911. doi:10.1001/jama.2010.1510
235. Rapp S, Brenes G, Marsh AP. Memory enhancement training for older adults with mild cognitive impairment: a preliminary study. Aging Ment Health. 2002;6(1):5-11. doi:10.1080/13607860120101077
236. Raskind MA, Peskind ER, Wessel T, Yuan W; Galantamine USA-1 Study Group. Galantamine in AD: a 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology. 2000;54 (12):2261-2268. doi:10.1212/WNL.54.12.2261
237. Richard E, Kuiper R, Dijkgraaf MG, Van Gool WA; Evaluation of Vascular Care in Alzheimer’s Disease. Vascular care in patients with Alzheimer’s disease with cerebrovascular lesions—a randomized clinical trial. J Am Geriatr Soc. 2009;57(5):797-805. doi:10.1111/j.1532-5415.2009.02217.x
238. Roberts J, Browne G, Milne C, et al. Problem-solving counseling for caregivers of the cognitively impaired: effective for whom? Nurs Res. 1999;48(3):162-172. doi:10.1097/00006199-199905000-00006
239. Rockwood K, Fay S, Song X, MacKnight C, Gorman M; Video-Imaging Synthesis of Treating Alzheimer’s Disease (VISTA) Investigators. Attainment of treatment goals by people with Alzheimer’s disease receiving galantamine: a randomized controlled trial. CMAJ. 2006;174(8):1099-1105. doi:10.1503/cmaj.051432
240. Rockwood K, Mintzer J, Truyen L, Wessel T, Wilkinson D. Effects of a flexible galantamine dose in Alzheimer’s disease: a randomised, controlled trial. J Neurol Neurosurg Psychiatry. 2001;71(5):589-595. doi:10.1136/jnnp.71.5.589
241. Rogers SL, Doody RS, Mohs RC, Friedhoff LT; Donepezil Study Group. Donepezil improves cognition and global function in Alzheimer disease: a 15-week, double-blind, placebo-controlled study. Arch Intern Med. 1998;158(9):1021-1031. doi:10.1001/archinte.158.9.1021
242. Rogers SL, Friedhoff LT; Donepezil Study Group. The efficacy and safety of donepezil in patients with Alzheimer’s disease: results of a US multicentre, randomized, double-blind, placebo-controlled trial. Dementia. 1996;7(6):293-303.
243. Rösler M, Anand R, Cicin-Sain A, et al. Efficacy and safety of rivastigmine in patients with Alzheimer’s disease: international randomised controlled trial [published correction appears in BMJ. 2001;322(7300):1456]. BMJ. 1999;318(7184): 633-638. doi:10.1136/BMJ.318.7184.633
244. Rovner BW, Casten RJ, Hegel MT, Leiby B. Preventing cognitive decline in black individuals with mild cognitive impairment: a randomized clinical trial. JAMA Neurol. 2018;75(12):1487-1493. doi:10.1001/jamaneurol.2018.2513
245. Salloway S, Ferris S, Kluger A, et al; Donepezil 401 Study Group. Efficacy of donepezil in mild cognitive impairment: a randomized placebo-controlled trial. Neurology. 2004;63(4): 651-657. doi:10.1212/01.WNL.0000134664.80320.92
246. Samus QM, Johnston D, Black BS, et al. A multidimensional home-based care coordination intervention for elders with memory disorders: the Maximizing Independence at Home (MIND) pilot randomized trial. Am J Geriatr Psychiatry. 2014;22(4):398-414. doi:10.1016/j.jagp.2013.12.175
247. Sano M, Bell KL, Galasko D, et al. A randomized, double-blind, placebo-controlled trial of simvastatin to treat Alzheimer disease. Neurology. 2011;77(6):556-563. doi:10.1212/WNL. 0b013e318228bf11
248. Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease: the Alzheimer’s Disease Cooperative Study. N Engl J Med. 1997;336(17):1216-1222. doi:10.1056/ NEJM199704243361704
249. Saxton J, Hofbauer RK, Woodward M, et al. Memantine and functional communication in Alzheimer’s disease: results of a 12-week, international, randomized clinical trial. J Alzheimers Dis. 2012;28(1):109-118. doi:10.3233/JAD-2011-110947
250. Schoenmakers B, Buntinx F, Delepeleire J. Supporting family carers of community-dwelling elder with cognitive decline: a randomized controlled trial. Int J Family Med. 2010;2010:184152. doi:10.1155/2010/184152
251. Schwenk M, Zieschang T, Oster P, Hauer K. Dual-task performances can be improved in patients with dementia: a randomized controlled trial. Neurology. 2010;74(24):1961-1968. doi:10.1212/WNL.0b013e3181e39696
252. Seltzer B, Zolnouni P, Nunez M, et al; Donepezil “402” Study Group. Efficacy of donepezil in early-stage Alzheimer disease: a randomized placebo-controlled trial. Arch Neurol. 2004;61(12):1852-1856. doi:10.1001/archneur.61.12.1852
253. Shimada H, Makizako H, Doi T, et al. Effects of combined physical and cognitive exercises on cognition and mobility in patients with mild cognitive impairment: a randomized clinical trial. J Am Med Dir Assoc. 2018;19(7):584-591. doi:10.1016/j.jamda.2017.09.019
254. Shinto L, Quinn J, Montine T, et 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(1):111-120. doi:10.3233/JAD-130722
255. Simons M, Schwärzler F, Lütjohann D, et al. Treatment with simvastatin in normocholesterolemic patients with Alzheimer’s disease: a 26-week randomized, placebo-controlled, double-blind trial. Ann Neurol. 2002;52(3):346-350. doi:10.1002/ana.10292
256. Sinn N, Milte CM, Street SJ, et al. Effects of n-3 fatty acids, EPA v. DHA, on depressive symptoms, quality of life, memory and executive function in older adults with mild cognitive impairment: a 6-month randomised controlled trial. Br J Nutr. 2012;107(11):1682-1693. doi:10.1017/S0007114511004788
257. Siu MY, Lee DTF. Effects of tai chi on cognition and instrumental activities of daily living in community dwelling older people with mild cognitive impairment. BMC Geriatr. 2018;18(1):37. doi:10.1186/s12877-018-0720-8
258. Soininen H,West C, Robbins J, Niculescu L. Long-term efficacy and safety of celecoxib in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2007;23(1):8-21. doi:10.1159/000096588
259. Spalding-Wilson KN, Guzmán-Vélez E, Angelica J, Wiggs K, Savransky A, Tranel D. A novel two-day intervention reduces stress in caregivers of persons with dementia. Alzheimers Dement (NY). 2018;4:450-460. doi:10.1016/j.trci.2018.08.004
260. Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for the treatment of mild to moderate Alzheimer disease: preliminary results. Arch Neurol. 2005;62(5):753-757. doi:10.1001/archneur.62.5.753
261. Spijker A, Wollersheim H, Teerenstra S, et al. Systematic care for caregivers of patients with dementia: a multicenter, cluster-randomized, controlled trial. Am J Geriatr Psychiatry. 2011;19(6):521-531. doi:10.1097/JGP.0b013e3182110599
262. Steffen AM, Gant JR. A telehealth behavioral coaching intervention for neurocognitive disorder family carers. Int J Geriatr Psychiatry. 2016;31(2): 195-203. doi:10.1002/gps.4312
263. Straubmeier M, Behrndt E-M, Seidl H, Özbe D, Luttenberger K, Graessel E. Non-pharmacological treatment in people with cognitive impairment. Dtsch Arztebl Int. 2017;114(48):815-821. doi:10.3238/arztebl.2017.0815
264. Sun Y, Lu CJ, Chien KL, Chen ST, Chen RC. Efficacy of multivitamin supplementation containing vitamins B6 and B12 and folic acid as adjunctive treatment with a cholinesterase inhibitor in Alzheimer’s disease: a 26-week, randomized, double-blind, placebo-controlled study in Taiwanese patients. Clin Ther. 2007;29(10):2204-2214. doi:10.1016/j.clinthera.2007.10.012
265. Suzuki T, Shimada H, Makizako H, et al. Effects of multicomponent exercise on cognitive function in older adults with amnestic mild cognitive impairment: a randomized controlled trial. BMC Neurol. 2012;12(1):128. doi:10.1186/1471-2377-12-128
266. Tariot PN, Solomon PR, Morris JC, Kershaw P, Lilienfeld S, Ding C; Galantamine USA-10 Study Group. A 5-month, randomized, placebo-controlled trial of galantamine in AD. Neurology. 2000;54(12):2269-2276. doi:10.1212/WNL.54.12.2269
267. Teri L, Gibbons LE, McCurry SM, et al. Exercise plus behavioral management in patients with Alzheimer disease: a randomized controlled trial. JAMA. 2003;290(15):2015-2022. doi:10.1001/jama.290.15.2015
268. Teri L, McCurry SM, Logsdon R, Gibbons LE. Training community consultants to help family members improve dementia care: a randomized controlled trial. Gerontologist. 2005;45(6):802-811. doi:10.1093/geront/45.6.802
269. Thyrian JR, Hertel J, Wucherer D, et al. Effectiveness and safety of dementia care management in primary care: a randomized clinical trial. JAMA Psychiatry. 2017;74(10):996-1004. doi:10.1001/jamapsychiatry.2017.2124
270. Train the Brain Consortium. Randomized trial on the effects of a combined physical/cognitive training in aged MCI subjects: the Train the Brain study. Sci Rep. 2017;7:39471. doi:10.1038/srep39471
271. Tremont G, Davis JD, Papandonatos GD, et al. Psychosocial telephone intervention for dementia caregivers: a randomized, controlled trial. Alzheimers Dement. 2015;11(5):541-548. doi:10.1016/j.jalz.2014.05.1752
272. Troyer AK, Murphy KJ, Anderson ND, Moscovitch M, Craik FI. Changing everyday memory behaviour in amnestic mild cognitive impairment: a randomised controlled trial. Neuropsychol Rehabil. 2008;18(1):65-88. doi:10.1080/09602010701409684
273. Tsantali E, Economidis D, Rigopoulou S. Testing the benefits of cognitive training vs. cognitive stimulation in mild Alzheimer’s disease: a randomised controlled trial. Brain Impair. 2017;18(2):188-196. doi:10.1017/BrImp.2017.6
274. Tsolaki M, Kounti F, Agogiatou C, et al. Effectiveness of nonpharmacological approaches in patients with mild cognitive impairment. Neurodegener Dis. 2011;8(3):138-145. doi:10.1159/000320575
275. Tune L, Tiseo PJ, Ieni J, et 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(2):169-177. doi:10.1097/00019442-200303000-00007
276. Ulstein ID, Sandvik L, Wyller TB, Engedal K. A one-year randomized controlled psychosocial intervention study among family carers of dementia patients—effects on patients and carers. Dement Geriatr Cogn Disord. 2007;24(6):469-475. doi:10.1159/000110740
277. Valen-Sendstad A, Engedal K, Stray-Pedersen B, et al; ADACT Study Group. Effects of hormone therapy on depressive symptoms and cognitive functions in women with Alzheimer disease: a 12 month randomized, double-blind, placebo-controlled study of low-dose estradiol and norethisterone. Am J Geriatr Psychiatry. 2010;18(1):11-20. doi:10.1097/JGP.0b013e3181beaaf4
278. Venturelli M, Lanza M, Muti E, Schena F. Positive effects of physical training in activity of daily living-dependent older adults. Exp Aging Res. 2010;36(2):190-205. doi:10.1080/03610731003613771
279. Vickrey BG, Mittman BS, Connor KI, et al. The effect of a disease management intervention on quality and outcomes of dementia care: a randomized, controlled trial. Ann Intern Med. 2006;145(10):713-726. doi:10.7326/0003-4819-145-10-200611210-00004
280. Vidovich MR, Lautenschlager NT, Flicker L, Clare L, McCaul K, Almeida OP. The PACE study: a randomized clinical trial of cognitive activity strategy training for older people with mild cognitive impairment. Am J Geriatr Psychiatry. 2015;23(4):360-372. doi:10.1016/j.jagp.2014.04.002
281. Voigt-Radloff S, Graff M, Leonhart R, et al. A multicentre RCT on community occupational therapy in Alzheimer’s disease: 10 sessions are not better than one consultation. BMJ Open. 2011;1(1):e000096. doi:10.1136/bmjopen-2011-000096
282. Vreugdenhil A, Cannell J, Davies A, Razay G. A community-based exercise programme to improve functional ability in people with Alzheimer’s disease: a randomized controlled trial. Scand J Caring Sci. 2012;26(1):12-19. doi:10.1111/j.1471-6712.2011.00895.x
283. Waldorff FB, Buss DV, Eckermann A, et al. Efficacy of psychosocial intervention in patients with mild Alzheimer’s disease: the multicentre, rater blinded, randomised Danish Alzheimer Intervention Study (DAISY). BMJ. 2012;345:e4693. doi:10.1136/bmj.e4693
284. Wang L-Q, Chien W-T. Randomised controlled trial of a family-led mutual support programme for people with dementia. J Clin Nurs. 2011;20(15-16):2362-2366. doi:10.1111/j.1365-2702.2011.03746.x
285. Wang PN, Liao SQ, Liu RS, et al. Effects of estrogen on cognition, mood, and cerebral blood flow in AD: a controlled study. Neurology. 2000;54(11):2061-2066. doi:10.1212/WNL.54.11.2061
286. Wilcock G, Möbius HJ, Stöffler A; MMM500 Group. A double-blind, placebo-controlled multicentre study of memantine in mild to moderate vascular dementia (MMM500). Int Clin Psychopharmacol. 2002;17(6):297-305. doi:10.1097/00004850-200211000-00005
287. Wilcock GK, Lilienfeld S, Gaens E. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer’s disease: multicentre randomised controlled trial [published correction appears in BMJ. 2001;322(7283):405]. BMJ. 2000;321(7274):1445-1449. doi:10.1136/bmj.321.7274.1445
288. Wilkinson D, Doody R, Helme R, et al; Donepezil 308 Study Group. Donepezil in vascular dementia: a randomized, placebo-controlled study. Neurology. 2003;61(4):479-486. doi:10.1212/01.WNL.0000078943.50032.FC
289. Wilkinson D, Fox NC, Barkhof F, Phul R, Lemming O, Scheltens P. Memantine and brain atrophy in Alzheimer’s disease: a 1-year randomized controlled trial. J Alzheimers Dis. 2012;29(2):459-469. doi:10.3233/JAD-2011-111616
290. Wilkinson D, Murray J. Galantamine: a randomized, double-blind, dose comparison in patients with Alzheimer’s disease. Int J Geriatr Psychiatry. 2001;16(9):852-857. doi:10.1002/gps.409
291. Williams VP, Bishop-Fitzpatrick L, Lane JD, et al. Video-based coping skills to reduce health risk and improve psychological and physical well-being in Alzheimer’s disease family caregivers. Psychosom Med. 2010;72(9):897-904. doi:10.1097/PSY.0b013e3181fc2d09
292. Wilz G, Reder M, Meichsner F, Soellner R. The Tele.TAnDem Intervention: telephone-based CBT for family caregivers of people with dementia. Gerontologist. 2018;58(2):e118-e129. doi:10.1093/geront/gnx183
293. Wilz G, Soellner R. Evaluation of a short-term telephone-based cognitive behavioral intervention for dementia family caregivers. Clin Gerontol. 2016;39(1):25-47. doi:10.1080/07317115.2015.1101631
294. Winblad B, Cummings J, Andreasen N, et 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(5):456-467. doi:10.1002/gps.1788
295. Winblad B, Engedal K, Soininen H, et al; Donepezil Nordic Study Group. A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology. 2001;57(3):489-495. doi:10.1212/WNL.57.3.489
296. Winter L, Gitlin LN. Evaluation of a telephone-based support group intervention for female caregivers of community-dwelling individuals with dementia. Am J Alzheimers Dis Other Demen. 2006;21(6):391-397. doi:10.1177/1533317506291371
297. Wolfs CAG, Kessels A, Dirksen CD, Severens JL, Verhey FRJ. Integrated multidisciplinary diagnostic approach for dementia care: randomised controlled trial. Br J Psychiatry. 2008;192(4):300-305. doi:10.1192/bjp.bp.107.035204
298. Wright LK, Litaker M, Laraia MT, DeAndrade S. Continuum of care for Alzheimer’s disease: a nurse education and counseling program. Issues Ment Health Nurs. 2001;22(3):231-252. doi:10.1080/ 01612840152053084
299. Xiao LD, De Bellis A, Kyriazopoulos H, Draper B, Ullah S. The effect of a personalized dementia care intervention for caregivers from Australian minority groups. Am J Alzheimers Dis Other Demen. 2016;31(1):57-67. doi:10.1177/1533317515578256
300. Yurko-Mauro K, McCarthy D, Rom D, et al; MIDAS Investigators. Beneficial effects of docosahexaenoic acid on cognition in age-related cognitive decline. Alzheimers Dement. 2010;6(6):456-464. doi:10.1016/j.jalz.2010.01.013
301. Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern Med. 2009;169(9):867-873. doi:10.1001/archinternmed.2009.43
302. Hernandez RK, Farwell W, Cantor MD, Lawler EV. Cholinesterase inhibitors and incidence of bradycardia in patients with dementia in the Veterans Affairs New England Healthcare System. J Am Geriatr Soc. 2009;57(11):1997-2003. doi:10.1111/j.1532-5415.2009.02488.x
303. Thavorn K, Gomes T, Camacho X, Yao Z, Juurlink D, Mamdani M. Upper gastrointestinal bleeding in elderly adults with dementia receiving cholinesterase inhibitors: a population-based cohort study. J Am Geriatr Soc. 2014;62(2):382-384. doi:10.1111/jgs.12670
304. Psychological Assessment Resources (PAR). MMSE: Mini-Mental State Examination. PAR website. https://www.parinc.com/products/pkey/237. January 2020. Accessed January 16, 2020.
305. Prince M, Bryce R, Ferri C; Alzheimer’s Disease International.World Alzheimer Report 2011: the Benefits of Early Diagnosis and Intervention. https://www.alz.co.uk/research/WorldAlzheimerReport2011.pdf. Published 2011. Accessed December 5, 2018.
306. Martin S, Kelly S, Khan A, et al. Attitudes and preferences towards screening for dementia: a systematic review of the literature. BMC Geriatr. 2015;15:66. doi:10.1186/s12877-015-0064-6
307. Borson S, Frank L, Bayley PJ, et al. Improving dementia care: the role of screening and detection of cognitive impairment. Alzheimers Dement. 2013; 9(2):151-159. doi:10.1016/j.jalz.2012.08.008
308. Le Couteur DG, Doust J, Creasey H, Brayne C. Political drive to screen for pre-dementia: not evidence based and ignores the harms of diagnosis. BMJ. 2013;347:f5125. doi:10.1136/bmj.f5125
309. Bunn F, Goodman C, Sworn K, et al. Psychosocial factors that shape patient and carer experiences of dementia diagnosis and treatment: a systematic review of qualitative studies. PLoS Med. 2012;9(10):e1001331. doi:10.1371/journal.pmed.1001331
310. Boustani MA, Justiss MD, Frame A, et al. Caregiver and noncaregiver attitudes toward dementia screening. J Am Geriatr Soc. 2011;59(4):681-686. doi:10.1111/j.1532-5415.2011.03327.x
311. Holsinger T, Boustani M, Abbot D, Williams JW. Acceptability of dementia screening in primary care patients. Int J Geriatr Psychiatry. 2011;26(4):373-379. doi:10.1002/gps.2536

Return to Table of Contents

This figure is the analytic framework that depicts the five Key Questions to be addressed in the systematic review. The figure illustrates how screening for cognitive impairment in adults aged 65 years or older may result in improved intermediate outcomes (decision making) and health outcomes (patient, family/caregiver, and societal outcomes) (Key Question 1). There is also a question related to the accuracy of screening instruments used to detect cognitive impairment (Key Question 2) and potential harms of screening (Key Question 3). Additionally, the figure illustrates how pharmacologic and nonpharmacologic interventions for early dementia may have an impact on intermediate outcomes (decision making) and health outcomes (patient, family/caregiver, and societal outcomes) (Key Question 4) and whether these interventions result in any harms (Key Question 5).

Evidence reviews for the US Preventive Services Task Force (USPSTF) use an analytic framework to visually display the key questions that the review will address to allow the USPSTF to evaluate the effectiveness and safety of a preventive service. The questions are depicted by linkages that relate interventions and outcomes. A dashed line indicates a health outcome that immediately follows an intermediate outcome. Details available in the USPSTF Procedure Manual.6 MCI indicates mild cognitive impairment. 
a Outcomes included (1) patient-related outcomes (eg, health-related quality of life, incident dementia, cognitive function, physical function, global function, dementia-related symptoms/behaviors, institutionalizations, safety); (2) caregiver outcomes (eg, health-related quality of life, global stress/distress, caregiver burden, depression, anxiety); and (3) societal outcomes (safety [eg, automobile crashes]).

Return to Table of Contents

This figure is a flow chart that summarizes the search and selection of articles for the review. There were 21,018 citations identified through literature databases. An additional 71 citations were identified from outside sources such as reference lists and suggestions from peer reviewers, 365 citations were from the most recent (2014) USPSTF review. After duplicates were removed, 11,644 unique citations were screened at the title/abstract stage. The full-text of 966 citations were examined for inclusion for one or more of the Key Questions. The following number of studies were included for Key Question 1 (k=0), Key Question 2 (k=59),  Key Question 3 (k=0), Key Question 4 (k=224), and Key Question 5 (k=98).

KQ indicates key question.
a Articles could be assessed for more than 1 KQ.
b Reasons for exclusion: Aim: Study aim not relevant. Setting: Study was not conducted in a country relevant to US practice or study was conducted in intermediate care facility or otherwise unrepresentative setting. Outcomes: Study did not report relevant outcomes. Population: Study population not relevant (age <65 years; exclusively populations with mental health illnesses or chronic disease; severe dementia; professional caregivers; otherwise not representative community-dwelling population). Intervention: Study used an excluded intervention or screening approach or intervention aim irrelevant. Study design: Not an included study design; comparative effectiveness; follow-up less than 3 months (does not apply to harms); case-control design (KQ2 only); cohort or case-control (with n<1000) (KQ5 only). Quality: Study did not meet criteria for fair or good quality. Publication type: Ancillary study to excluded primary study.

Return to Table of Contents

Intervention Type Outcome Pooled Mean Difference, Change
(95% CI)a,b
No. Analyzed I2 Tau
RCTs Participants
FDA Medications
Donepezil GCF (ADAS-Cog) −2.13 (−3.32 to −0.94) 6 1981 64.4 0.90
GCF (MMSE) 1.24 (0.81 to 1.67) 12 3192 65.3 0.57
GF (continuous) −0.24 (−0.39 to −0.09) 8 3302 70.7 0.15
GF (dichotomous)c 1.33 (1.07 to 1.66) 9 2440 77.4 0.23
Severe adverse events 1.18 (0.99 to 1.40) 12 4045 0.0 0
Withdrawals 1.88 (1.54 to 2.29) 13 4124 8.8 0
Galantamine GCF (ADAS-Cog) −2.13 (−2.94 to −1.32) 9 3786 65.9 0.84
GCF (MMSE) NA 1 1765 NA NA
GF (continuous) NA 1 126 NA NA
GF (dichotomous)c 1.21 (1.11 to 1.31) 8 3486 56.2 0.07
Severe adverse events 1.06 (0.88 to 1.29) 7 4987 0.0 0
Withdrawals 1.98 (1.52 to 2.57) 10 6147 51.1 0.28
Rivastigmine GCF (ADAS-Cog) −2.43 (−4.10 to −0.75) 5 2618 81.9 1.21
GCF (MMSE) 0.88 (0.28 to 1.49) 6 2415 44.9 0.39
GF (continuous) −0.14 (−0.43 to 0.15) 6 2535 85.7 0.25
GF (dichotomous)c 1.49 (1.13 to 1.98) 5 1934 61.4 0.16
Severe adverse events 1.15 (0.87 to 1.52) 6 2619 10.4 0
Withdrawals 2.21 (1.43 to 3.42) 8 3131 57.0 0.38
Memantine GCF (ADAS-Cog) −0.88 (−1.65 to −0.11) 8 2609 78.1 0.69
GCF (MMSE) 0.36 (−0.31 to 1.04) 5 1217 33.2 0.27
GF (continuous) −0.14 (−0.33 to 0.05) 5 1396 32.9 0.09
GF (dichotomous)c 1.15 (0.49 to 2.69) 2 545 0.0 0
Severe adverse events 0.88 (0.77 to 1.01) 10 3350 0.0 0
Withdrawals 1.26 (0.94 to 1.70) 9 3288 0.0 0
Nonpharmaceutical Patient-Level
Cognitive stimulation and training GCF (ADAS-Cog) −0.66 (−1.60 to 0.29) 8 842 0 0
GCF (MMSE) 1.33 (0.29 to 2.37) 15 1384 91.1 1.91
Exercise interventions GCF (ADAS-Cog) −1.05 (−3.49 to 1.10) 6 1071 77.4 1.62
GCF (MMSE) 1.17 (0.45 to 1.90) 10 1168 81.3 0.98
Multicomponent and other interventions GCF (ADAS-Cog) −1.66 (−10.03 to 6.72) 2 167 56.5 0.72
GCF (MMSE) 0.26 (−0.54 to 1.00) 8 1238 30.3 0.55
Caregiver
Psychoeducation interventions Caregiver burden −0.24 (−0.36 to −0.13) 27 2776 50.2 0.20
Caregiver depression −0.26 (−0.39 to −0.13) 37 4555 76.9 0.35
Care or case management Caregiver burden −0.54 (−0.96 to −0.12) 8 1215 82.9 0.45
Caregiver depression −0.13 (−0.39 to 0.12) 4 668 0.0 0
Other caregiver or caregiver-patient dyad interventions Caregiver burden −0.30 (−2.26 to 1.36) 5 459 89.6 1.36
Caregiver depression −0.00 (−0.34 to 0.34) 5 645 53.7 0.20

Abbreviations: ADAS-Cog, Alzheimer Disease Assessment Scale–Cognitive Subscale; FDA, US Food and Drug Administration; GCF, global cognitive function; GF, global function; MMSE, Mini-Mental State Examination; NA, not applicable.
a For dichotomous outcomes, this represents an RR.
b For analyses with fewer than 10 studies, the restricted maximum likelihood method was used to calculate the confidence interval.
c Global function measure compares improving or maintaining global function vs a decline in global function as assessed by measures such as the Clinician’s Interview-Based Impression of Change Plus Informant Input (CIBIC+).

Return to Table of Contents

Instrument or Treatment, Study Designs,
Observations
Summary of Findings Consistency and Precisiona Other Limitation Strength of Evidencea Applicability
KQ1. Benefits of Screening
1 RCT
(n = 4005)
No evidence of a difference in health-related QOL at 1, 6, and 12 mo between participants randomized to screening vs no screening as well as no significant difference in health care utilization and advanced planning at 12 mo NA High rate of missing data for all outcomes at all time points given attrition and data quality issues (42% missing data at 12 mo for primary outcome) Low evidence of no benefit Mean age of participants within the 1 RCT was 74.2 y, and the majority were women (66%) and white (67%)

More than one-third of primary care–eligible older adults declined participation in the study, and 66% of those who screened positive refused further diagnostic assessment

KQ2: Accuracy of Screening
Very brief instruments (31 cross-sectional studies [n = 22,359]) 25 Instruments

To detect dementia, sensitivity was usually at ≥0.75 and specificity at ≥0.80

Across all very brief instruments, the detection of MCI was less consistent, with a wide range in sensitivity and specificity

Reasonably consistent and precise (dementia); inconsistent and imprecise (MCI) Large number of instruments with little replication Moderate evidence of adequate sensitivity and specificity Broad inclusion of older adult populations with a wide range of underlying dementia and MCI
Brief instruments (48 cross-sectional studies [n = 29,950]) 20 Instruments

For the MMSE, to detect dementia, 15 studies (n = 12,796) resulted in a pooled sensitivity of 0.89 (95% CI, 0.85 to 0.92) and a specificity of 0.89 (95% CI, 0.85 to 0.93)

For other brief instruments reported in ≥1 study, sensitivity ranged from 0.74 to 1.0 and specificity ranged from 0.65 to 0.96

Across all brief instruments, the detection of MCI was less consistent, with a wide range in sensitivity and specificity

Reasonably consistent and precise (dementia); inconsistent and imprecise (MCI) Large number of instruments with little replication, except for the MMSE Moderate evidence of adequate sensitivity and specificity Broad inclusion of older adult populations with a wide range of underlying dementia and MCI

Administration time less useful for primary care screening

Longer, self-administered instruments (8 cross-sectional studies [n = 2271]) 4 Instruments

Only the IQCODE was assessed in ≥1 study, with sensitivity to detect dementia ranging from 0.80 to 0.88 and specificity ranging from 0.51 to 0.91

To detect MCI, sensitivity ranged from 0.71 to 0.82 and specificity ranged from 0.69 to 0.92

Reasonably consistent (dementia and MCI); precise (dementia and MCI) Few instruments, little replication Moderate evidence of adequate sensitivity and specificity Broad inclusion of older adult populations with a wide range of underlying dementia and MCI
KQ3: Harms of Screening
1 RCT
(n = 4005)
No evidence of a difference in symptoms of depression or anxiety between those in the screening vs no screening group at 1-, 6-, and 12-mo follow-up NA High rate of missing data for all outcomes at all time points given attrition and data quality issues (42% missing data at 12 mo for primary outcome) Low evidence of no harm Mean age of participants within the 1 RCT was 74.2 y, and the majority were women (66%) and white (67%)

More than one-third of primary care eligible older adults declined participation in the study, and 66% of those who screened positive refused further diagnostic assessment

KQ4: Benefits of Interventionsa
AChEIs and memantine (48 RCTs [n = 22,431]) Medications may improve measures of global cognitive function in short term, but magnitude of differences between drug vs placebo groups was small

Pooled results indicate differences in change ranging from approximately 1 to 2.5 points in favor of drug groups on the ADAS-Cog-11 (range, 0-70)

Donepezil: MD, –2.13 (95% CI, –0.94 to –3.32); 6 studies (n = 1981); I2 = 64.4%
Galantamine: MD, –2.13 (95% CI, –1.32 to –2.94); 9 studies (n = 3786); I2 = 65.9
Rivastigmine: –2.43 (95% CI, –0.75 to –4.10); 5 studies (n = 2618); I2 = 81.9%
Memantine: –0.88 (95% CI, –0.11 to –1.65); 8 studies (n = 2609); I2 = 78.1%

Using accepted thresholds of clinical benefit, the average benefit across patients was not clinically significant

AChEIs and memantine increased the likelihood of improving or maintaining patients’ global function (eg, using a CIBIC+) by 15% (for memantine) to 50% (for rivastigmine) in the short term (pooled 95% CI range, 0.49 to 2.69)

Pooled change in global function found small effect sizes (SMDs ranging from 0.14 to 0.46)

Other important measures such as mental health and neuropsychiatric symptoms and rates of institutionalization were rarely reported; no RCTs included measures of QOL

Reasonably consistent; precise Evidence of a small-studies effect for the pooled result for global cognitive function measured by the MMSE for donepezil, indicating the possibility of publication bias

Few studies included follow-up longer than 6 mo

Moderate evidence of a small benefit Older adults with dementia (mainly Alzheimer disease), particularly among those with moderate vs mild forms

Unclear representation of ethnic minorities and those of varying education levels

Doses of medications applicable to common use

Other medications and supplements (29 RCTs [n = 6489]) No evidence that antihypertensives, vitamins or omega-3 fatty acids, gonadal steroids, HMG-CoA reductase inhibitors, or NSAIDS are beneficial for any cognitive, functional, or other outcome at 3 mo to 4 y of follow-up Reasonably consistent; imprecise Small studies often with differential attrition between groups

Lack of consistency in formulations and dosages of agents used

Low evidence of no benefit Older adults with mild to moderate dementia

Unclear representation of ethnic minorities and those of varying education levels

Nonpharmacologic patient-level interventions (61 RCTs [n = 7847]) No clear benefit of cognitive stimulation, training, or rehabilitation; exercise interventions; multicomponent interventions; and other interventions on global and domain-specific cognitive function compared with controls at 3 mo to 2 y follow-up among persons with MCI or dementia

Effect estimates generally favored intervention groups, but the magnitude of effects was inconsistent across RCTs and represented very wide confidence intervals

Measures related to physical function, QOL, and mental and neuropsychiatric symptoms were only reported by one-half or less of the studies for each intervention group, and few found robust differences between groups

Reasonably consistent; imprecise Small studies of limited duration

Types of outcomes, specific measures, and duration of follow-up was highly variable across studies

Low evidence of small to no benefit Broad range of older adults with MCI and mild and moderate dementia

Very sparse reporting of clinical characteristics of the included patients such as race/ethnicity and education

Virtually no data on effect modification by important clinical differences

Many complex interventions may not be widely available in the United States

Caregiver and caregiver-patient dyad interventions (88 RCTs [n = 14,880]) Consistent benefit of psychoeducation and care and case management interventions on caregiver burden and depression outcomes; however, effect sizes were mostly small and are of unclear clinical significance

For caregiver burden, the standardized pooled effect was –0.24 (95% CI, –0.36 to –0.13); 27 studies (n = 2776); I2 = 50.2% for psychoeducation interventions and –0.54 (95% CI, –0.85 to –0.22); 8 studies (n = 1215); I2 = 82.9% for care and case management interventions

Other outcomes such as caregiver or patient QOL, rates or time to institutionalization, patient mental health and neuropsychiatric symptoms, and patient functional ability were sparsely reported across the RCTs with no consistent evidence of benefit

Decision-making and preparation for meeting dementia-related needs were only reported by 1 RCT each, with neither finding statistically significant benefit of the interventions vs control conditions on overall scores for these measure

Reasonably consistent; precise Little evidence of longer-term effects;inconsistency in outcomes and specific measures across studies, with many providing little data on precise scales used Moderate evidence of small benefit Generally applicable to caregivers of persons with moderate dementia

Many complex interventions may not be widely available in the United States

KQ5: Harms of Intervention
AChEIs and memantine (48 RCTs [n = 22,431]; 3 observational studies [n = 190,076]) Adverse effects from medications were common

Withdrawal or discontinuation was more common with AChEIs (13% withdrawing for donepezil and rivastigmine, 14% for galantamine) than placebo (8%)

Memantine appeared to be better tolerated, with no significant difference in withdrawal rates (8%) compared with placebo (8%)

In total, there did not appear to be a difference in total SAEs for these medications across studies with limited duration of follow-up; however, individual studies, including observational evidence, reported increased rates of bradycardia and, relatedly, of syncope, falls, and need for pacemaker placement among those exposed vs unexposed to AChEIs

Reasonably consistent; precise The definitions of SAEs, which likely vary, were rarely described in the included studies Moderate evidence of harm Mostly represented patients with moderate dementia
Other medications and supplements (21 RCTs [n = 5688]) Across interventions, harms were not clearly significantly increased in intervention vs control groups Reasonably consistent; precise Small studies often with differential attrition between groups

Lack of consistency in formulations and dosages of agents used

Low evidence of no harm Older adults with mild to moderate dementia

Unclear representation of ethnic minorities and those of varying education levels

Nonpharmacologic patient-level interventions (12 RCTs [n = 2370]) Little evidence of harms from good quality studies. Evidence of greater musculoskeletal problems among persons taking part in exercise interventions vs comparators

One trial reported 1 case of atrial fibrillation among 1 patient during an exercise session

Reasonably consistent; precise Sparse reporting of harms

RCTs of exercise interventions more likely to report monitoring harms than cognitive training or other interventions

Low evidence of no harmb Applicable to patients with mild to moderate dementia and MCI
Caregiver and caregiver-patient dyad interventions (4 RCTs [n = 486]) No harms evident NA Sparse reporting of harms for patients or caregivers Low evidence of no harmb Generally applicable to caregivers of persons with moderate dementia

Abbreviations: AChEI, acetylcholinesterase inhibitor; ADAS-Cog, Alzheimer Disease Assessment Scale–Cognitive Subscale; CIBIC+, Clinician’s Interview-Based Impression of Change Plus Informant Input; HMG-CoA, 3-hydroxy-3-methyl-glutaryl-CoA reductase; IQCODE, Informant Questionnaire on Cognitive Decline in the Elderly; KQ, key question; MCI, mild cognitive impairment; MD, mean difference; MMSE, Mini-Mental State Examination; NA, not applicable; NR, not reported; NSAID, nonsteroidal anti-inflammatory drug; QOL, quality of life; RCT, randomized clinical trial; SAE, serious adverse event; SMD, standardized mean difference.
a For KQ4, consistency, precision, and strength of evidence, assessments were based on primary outcomes within each body of evidence. For AChEIs and memantine, assessments were for cognitive function and global function outcomes. For other medications and supplements, assessments were for cognitive function. For nonpharmacologic patient-level interventions, assessments were for cognitive function, physical function, and neuropsychiatric symptoms. For caregiver and caregiver-patient dyad interventions, assessments were for caregiver burden and depression outcomes.
b No hypothesized serious harms of nonpharmacologic patient or caregiver interventions. Thus, despite few studies reporting this outcome, there is low confidence that the finding of no harm in these RCTs reflects this body of evidence.

Return to Table of Contents