archived

Evidence Summary

Dementia in Older Adults: Screening, 2003

June 03, 2003

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 Malaz Boustani, M.D., M.P.H.;a Britt Peterson, M.D., M.P.H.;b Laura Hanson, M.D., M.P.H.;c Russell Harris, M.D., M.P.H.;d  Kathleen N. Lohr, Ph.D.e

The authors of this article are responsible for its contents, including any clinical or treatment recommendations. No statement in this article should be construed as an official position from the Agency for Healthcare Research and Quality, or the U.S. Department of Health and Human Services.

Select for copyright information. The USPSTF recommendations based on this review are online.

[a] Boustani: Indiana University Center for Aging Research, Indiana University School of Medicine, Indianapolis, IN.
[b] Peterson: Department of Psychiatry, University of New Mexico School of Medicine Albuquerque, NM.
[c] Hanson: Program on Aging, University of North Carolina School of Medicine, Chapel Hill, NC.
[d] Harris: Cecil G. Sheps Center for Health Services Research, University of North Carolina School of Medicine, Chapel Hill, NC.
[e] Lohr: Department of Health Policy and Administration, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, and RTI International, Research Triangle Park, NC.

 

Return to Table of Contents

Background: Dementia is a large and growing problem but is often not diagnosed in its earlier stages. Screening and earlier treatment could reduce the burden of suffering of this syndrome. Purpose: To review the evidence of benefits and harms of screening for and earlier treatment of dementia.

Data Sources: MEDLINE, PsycINFO, EMBASE, the Cochrane Library, experts, and bibliographies of reviews.

Study Selection: The authors developed eight key questions representing a logical chain between screening and improved health outcomes, along with eligibility criteria for admissible evidence for each question. Admissible evidence was obtained by searching the data sources.

Data Extraction: Two reviewers abstracted relevant information using standardized abstraction forms and graded article quality according to U.S. Preventive Services Task Force criteria. Data Synthesis: No randomized, controlled trial of screening for dementia has been completed. Brief screening tools can detect some persons with early dementia (positive predictive value <50%). Six to 12 months of treatment with cholinesterase inhibitors modestly slows the decline of cognitive and global clinical change scores in some patients with mild to moderate Alzheimer disease. Function is minimally affected, and fewer than 20% of patients stop taking cholinesterase inhibitors because of side effects. Only limited evidence indicates that any other pharmacologic or nonpharmacologic intervention slows decline in persons with early dementia. Although intensive multicomponent caregiver interventions may delay nursing home placement of patients who have caregivers, the relevance of this finding for persons who do not yet have caregivers is uncertain. Other potential benefits and harms of screening have not been studied.

Conclusions: Screening tests can detect undiagnosed dementia. In persons with mild to moderate clinically detected Alzheimer disease, cholinesterase inhibitors are somewhat effective in slowing cognitive decline. The effect of cholinesterase inhibitors or other treatments on persons with dementia detected by screening is uncertain.

Return to Table of Contents

Dementia is an acquired syndrome of decline in memory and at least one other cognitive domain, such as language, visuo-spatial, or executive function, sufficient to interfere with social or occupational functioning in an alert person.1 Multiple diseases can cause the dementia syndrome (hereafter called dementia). Alzheimer's disease and cerebrovascular ischemia (vascular dementia) are the 2 most common causes; some cases involve both of these etiologies. Although some potentially reversible conditions, such as hypothyroidism or vitamin B12 deficiency, are often considered to be causes of dementia, no more than 1.5% of cases of mild to moderate dementia are fully reversible.2

Age is the best studied and the strongest risk factor for dementia. Risk factors for Alzheimer's disease, other than age, include having a first-degree relative with a history of Alzheimer's disease and the apolipoprotein E ε4 genotype.3-5 Cardiovascular risk factors such as hypertension are associated with an increased risk of both Alzheimer's disease and vascular dementia.5-7

The aging of the U.S. population has been accompanied by a dramatic increase in the prevalence of dementia. Three percent to 11% of persons older than 65 years of age and 25% to 47% of those older than 85 years of age have dementia.8 –13 In 1997, the number of persons with Alzheimer disease in the United States was estimated to be 2.3 million, more than 90% of whom were 60 years of age and older.14

Dementia causes a high burden of suffering for patients, their families, and society.15-21 For patients, it leads to increased dependency and complicates other comorbid conditions. For families, it leads to anxiety, depression, and increased time spent caring for a loved one. The annual societal cost of dementia is approximately $100 billion, from health care and related costs as well as lost wages for patients and family caregivers.10,16,22

Clinicians using routine history and physical examination do not readily diagnose dementia during clinic visits. More than 50 percent of people with dementia have never been diagnosed by a physician, including many with mild but some with moderate dementia.23-27 This raises the possibility that screening tests might be able to identify people with undiagnosed dementia, and therefore permit patients and their families to receive care at an earlier stage in the disease process. Given the low prevalence of reversible causes, a recommendation for screening will depend on evidence of the additional benefits of earlier treatment for persons with irreversible causes, primarily Alzheimer's disease and vascular dementia.

For dementia screening to lead to improved health outcomes, primary care providers would need to have a brief, accurate screening test to apply during routine office visits. A positive screening test would then result in a diagnostic interview and clinical examination based on the Diagnostic and Statistical Manual of Mental Disorders, fourth edition (DSM IV) that could be performed by either the primary care physician or a specialist such as a geriatrician or neurologist. Finally, knowledge of the dementia diagnosis at this early stage would lead to improved health outcomes through more effective treatment. Ideal evidence to support these hypotheses would come from a randomized, controlled trial (RCT) of screening and earlier intervention, with long-term follow-up for both adverse and beneficial effects of screening.

The 1996 Guide to Clinical Preventive Services from the U.S. Preventive Services Task Force (USPSTF) found insufficient evidence to recommend for or against screening for dementia.28 Since the last USPSTF review, studies have been published concerning screening tests as well as both pharmacologic and caregiver interventions. Given the new evidence and the growing importance of this condition, the RTI-University of North Carolina Evidence-based Practice Center (RTI-UNC EPC) conducted a systematic review of the literature regarding the benefits and the harms of screening primary care populations to detect undiagnosed cases of dementia. Our review did not consider the issue of screening to detect people with cognitive impairment, termed mild cognitive impairment, who do not meet the criteria for dementia. Experts do not agree about the definition of MCI and other reviews have not found evidence of effective treatment for people with this problem.29,30 

Return to Table of Contents

Using USPSTF methods,31 we developed an analytic framework and eight key questions to guide our systematic review of the evidence for dementia screening. We developed eligibility criteria for selecting the evidence relevant to answer the key questions. We used the eligibility criteria to develop search terms and searched MEDLINE, PsycINFO, EMBASE, and the Cochrane Library databases for systematic reviews and high-quality studies relevant to each question. We limited all searches to reviews and studies published in English between 1 January 1994, and 1 September  2002, and that contained information relevant to a primary care population. 

We searched first for studies of screening that provided direct evidence that screening improves cognitive, social, or physical function; number of hospitalizations, institutionalizations, or health care visits; behavioral problems; caregiver burden; accidental injuries such as falls or automobile crashes; or patients' overall health-related quality of life. Because we found no direct evidence connecting screening and improved health outcomes, we searched for indirect evidence of the benefit of screening, including the prevalence of undiagnosed dementia; the accuracy of screening tests; the efficacy of early pharmacologic and nonpharmacologic treatment for people with Alzheimer's disease and vascular dementia; caregiver intervention for people with dementia; and the efficacy of interventions targeted to caregivers. We also searched for evidence of the adverse effects of screening and treatment. 

At least 2 authors reviewed abstracts and articles to identify those that met the eligibility criteria and then abstracted relevant information using standardized abstraction forms. We graded the quality of the included articles using criteria developed by the USPSTF Methods Work Group.31 In all cases, we accepted either single studies or systematic reviews that addressed the key question, met eligibility criteria, and were rated of at least fair quality. Table 1 lists these criteria and the number of articles found meeting them for each question. (For more information about methods, go to the Appendix.)

This evidence report was developed by the RTI–University of North Carolina Evidence-based Practice Center under contract to the Agency for Healthcare Research and Quality. Staff of the funding agency and members of the USPSTF contributed to the study design, reviewed draft and final manuscripts, and made editing suggestions.

Return to Table of Contents

The best evidence for or against screening for dementia would be derived from a well-designed RCT of screening with health outcomes. No such trial has been completed. In the absence of such a trial, evidence for or against screening comes from studies of the prevalence of undiagnosed dementia, the accuracy of screening instruments, the efficacy of treatments for people with dementia detected by screening, and the harms of screening and treatment.

How Common Is Undiagnosed Dementia?

Three studies in primary care populations aged 65 years and older compared the frequency of dementia by standard diagnostic tests with medical record notation of dementia or cognitive impairment26,27,32 or by independent questionnaire of the physician.27 Among all primary care patients aged 65 and older, 3.2% to 12% met criteria for dementia but had no dementia documentation (Table 2) or knowledge by the physician. A population-based study found that the prevalence of undiagnosed dementia among individuals aged 65 years and older was 1.8%.33 Another population-based study found that about half of reliable relatives of men with mild dementia failed to recognize a problem with thinking or memory.81

Patients who had not received a dementia diagnosis accounted for 50% to 66% of all cases of dementia in the primary care samples studied. Most missed cases were mild to moderate. In one small study, 78.6% of persons with mild dementia (11 of 14), 71.4% of persons with moderate dementia (5 of 7), and 20% of persons with severe dementia (1 of 5) had no documentation of a dementia diagnosis in the medical record.27 New screening in primary care practice could therefore potentially double the number of patients who receive a diagnosis of dementia. Most newly detected cases would be mild to moderate.

How Accurate Are the Screening Tests?

Three methodological problems make it difficult to assess the accuracy of screening tests for dementia. First, research has examined the accuracy of a large number of screening instruments to a limited degree. Few instruments have been examined in more than 2 or 3 small studies. Second, investigators used a variety of reference standards for the diagnosis of dementia. Because functions such as cognition are continuous, the reference standard must set the point at which the diagnosis of dementia can be made. Where this point is set makes a large difference in evaluating screening tests.82 Although research has yet to determine the optimal point for diagnosing dementia, the DSM-IV criteria are widely accepted in the United States and will be used as the standard in this review.83 Third, the populations in the studies of screening instruments varied greatly. Many studies included participants with severe dementia or people from memory clinics; who are not the focus of screening. Few studies have provided information on the accuracy of screening tests for detecting people with mild dementia. Thus, the evidence about test accuracy from these studies may be most appropriately extrapolated to detect people with moderate dementia; the accuracy of these tests in detecting people with mild dementia may be lower. Although we examined many studies of screening tests, we included only studies that examined instruments that are feasible for use in primary care settings, used DSM-IV (or similar) criteria as the reference standard, and provided information about the test's characteristics among people who were not known by themselves or others to have symptoms of dementia.

Most screening tests for dementia can be divided into cognitive tests of patients and functional assessments using both patients and others as informants. In 1996, the Agency for Health Care Policy and Research (now the Agency for Healthcare Research and Quality) published a systematic review and meta-analysis of studies that evaluated dementia screening tools.37 The review found 1 informant-based functional status instrument and 4 patient-based cognitive assessment tools, including the Mini-Mental Status Examination [MMSE], the most commonly used and studied test, which had acceptable accuracy (Table 3). Since that review, eight additional studies have examined the MMSE with similar findings.39-43, 45,84,85 The sensitivity of the five tests examined in the study by the Agency for Health Care Policy and Research ranged approximately from 69% to 90%; specificity depended on the cut-point used for an abnormal test result ( (Table 3). Using a cutpoint to attain high sensitivity necessarily lowers specificity. The positive predictive value of a screening test in a population with a prevalence of undiagnosed dementia of 10% is approximately 40% to 50% (Table 3). 

Several other patient-based cognitive screening tests have been tested either in single studies or in several studies that do not meet our criteria. Tests such as the Short Portable Mental Status Questionnaire,86,87 the Clock Drawing Test,88-92 the Modified Mini-Mental Status Test,40,93 the Mini-Cog,94 the Hopkins Verbal Learning Test,95,96 and the 7-Minute Screen97,98 are promising but need further testing in primary care populations. A new approach uses the telephone to examine cognitive function.97,99 

A weakness of the MMSE is its varying accuracy in patients of different ages, education levels, and ethnicities.84,100-103 The MMSE is most accurate for white people with at least a high school education. One way of compensating for this is to change the cutpoint for an abnormal test result for different ages and education levels.104 Whether this approach or other patient-based cognitive screening tools will be able to overcome this problem remains unclear. 

The Functional Activities Questionnaire was the informant-based functional assessment tool examined by the Agency for Health Care Policy and Research study (Table 3). Few other high quality studies have focused on functional assessments to screen for dementia. One promising screening test that warrants further study is the Informant Questionnaire on Cognitive Decline in the Elderly, which assesses change in both cognitive function as well as activities of daily living.42

In summary, some patient-based cognitive screening tests are available with reasonable accuracy for mild to moderate dementia, and research continues on other instruments and approaches. Although the MMSE is the most widely used and studied screening test for dementia, it requires specific adjustment for age and education. Tests of functional assessment have promise but need further study.

Because the apolipoprotein E ε4 allele is found approximately three times more frequently among people with Alzheimer's disease than among those without this condition, some groups express interest in using this test to screen for dementia.105 Many people with Alzheimer's disease, however, do not have the apolipoprotein E ε4 allele, and many with the allele never develop dementia.5 The value of this test in screening for dementia has yet to be demonstrated.

How Effective Are Pharmacologic Interventions for People with Mild to Moderate Dementia?

To be effective, interventions for people with dementia should ideally improve functional status to a degree discernible by caregivers or health care providers. Interpreting whether the change in an index of function in a clinical trial meets this test requires an understanding of measurement instruments for function as well as the natural history of Alzheimer's disease.  In most clinical research on Alzheimer's disease, function is measured by 1 or both of two scales: the Alzheimer's Disease Assessment Scale for Cognition (ADAS-Cog), a 70-point scale that measures cognitive status, and the Clinician's Interview Based Impression of Change, a scale that also includes caregiver input (CIBIC). The CIBIC includes information from both patient and caregiver interviews conducted by an experienced and independent clinician; the clinician rates the patient on a 7-point scale: 1, very much better; 4, no change; and 7, very much worse. Both the ADAS-Cog and the CIBIC scales are stable and sensitive to clinical change over time.106

The natural history of Alzheimer's disease is one of progressive decline; cognitive, physical, and social functions gradually deteriorate. Thus, "improvement" from an intervention for Alzheimer's disease means slowing the rate of decline. The rate of decline in Alzheimer's disease is not linear, however.67 People with mild dementia experience an average rate of decline of 5 or fewer ADAS-Cog points (≤2 MMSE points) per year. Thus, for people with mild dementia, a slowing of decline by 2 to 3 ADAS-Cog points over a year could mean a delay of up to 7 months in the progress of the disease.

By contrast, those individuals with moderate dementia (ADAS-Cog >15 but <55) experience an average decline in cognition of 7 to 11 ADAS-Cog points (2 to 4 MMSE points) annually.107-110 Thus, for people with moderate dementia, a slowing of decline by 2 to 3 ADAS-Cog points in a year could mean a delay of 2 to 5 months in the progress of the disease. An average difference between intervention and control groups of 2 to 3 ADAS-Cog points could be attributable either to this amount of delay in disease progress for all people in the intervention group or to a longer delay in a subset of people while others have no delay. Clinically, a difference of 2 to 3 points on the ADAS-Cog110 is equivalent, for example, to a person remembering who came to dinner the previous evening or performing familiar tasks, such as dressing.111 

Cholinesterase Inhibitors

Four systematic reviews72-75 and five additional RCTs76-80 compared one of the four U.S. Food and Drug Administration-approved cholinesterase inhibitors to placebo among people with mild to moderate Alzheimer's disease, apparently detected clinically. All nine studies were well conducted (Table 4). With few exceptions, all of these studies with at least 6 months of follow-up found a statistically significant difference between drug and placebo groups favoring the cholinesterase inhibitor ranging from 2.12 to 3.4 points on the ADAS-Cog scale. This difference manifests as a reduced rate of cognitive decline in people taking cholinesterase inhibitors compared with those taking placebo. 

In addition to their effects on cognition, cholinesterase inhibitors also stabilized or slightly improved clinician global impression of change as measured by the CIBIC after 6 to 12 months of treatment. In many studies, the proportion of patients whose condition had stabilized was an absolute 10 to 20 percent higher in the cholinesterase group than in the placebo group. The evidence is mixed, however, about the effects of cholinesterase inhibitors on functional measures such as instrumental activities of daily living (i.e., ability to use the telephone, mode of transportation, responsibility for medication, and ability to handle finances). In general, the studies show little or no effect on functional decline after 6 months of treatment and a small but statistically significant difference from placebo after 12 months of treatment.34-36,38,44 The most positive study was a 12-month RCT of donepezil treatment in 431 community-dwelling person with mild to moderate Alzheimer disease. The investigators found a median time to clinically evident decline of 6.9 months in the placebo group and 11.9 months in the donepezil group.90,100 

Research has found no clinically important differences between people taking cholinesterase inhibitors and those taking placebo in the development of behavioral and psychological symptoms, although not all trials measure this important health outcome. No well-conducted RCT of cholinesterase inhibitors has maintained a placebo-controlled, blinded control group for more than 12 months of treatment, and studies rarely addressed other important health outcomes such as utilization of health care services, injuries, and caregiver burden.

Ginkgo Biloba, Selegiline, Vitamin E, and Estrogen

The evidence is weak that drugs other than cholinesterase inhibitors have important benefits for people with Alzheimer's disease. Several RCTs have examined the effects of ginkgo biloba on cognitive function in people with mild to moderate dementia. Two meta-analyses of these studies, including one that examined only the 4 highest quality studies, found an approximate 3 percent difference in cognitive scales between Ginkgo biloba and placebo groups.16,46

A recent Cochrane review and meta-analysis of 15 placebo-controlled studies found that selegiline, a selective monoamine oxidase inhibitor, produced no clinically important differences in cognition, function, mood, behavior, or global clinical ratings when compared with placebo.47 In a 2-year RCT of the effect of vitamin E in people with moderate Alzheimer's disease, investigators found that it had no effect on cognition but limited evidence that it delayed institutionalization.48 No other well-conducted RCT has examined the effects of vitamin E. Two recent well-conducted RCTs examined estrogen therapy for women with mild to moderate dementia and found no evidence of clinical benefit.49,50 

Pharmacologic Treatment for Vascular Dementia 

The category of vascular dementia is heterogeneous, and the natural history of the disorder is not well understood.112 Some people who meet criteria for vascular dementia exhibit a progressive functional decline similar to that of Alzheimer's disease. Although antihypertensive treatment reduces the development of stroke and dementia, the evidence is limited that a similar course of treatment for people with mild to moderate vascular dementia delays progression of the disease.51 One systematic review found that aspirin had no effect on cognitive symptoms in people with vascular dementia.52 One RCT found that nimodipine (a calcium channel blocker) had no effect on the cognitive and the global symptoms of patients with vascular dementia;51, and 1 RCT found that galantamine at least stabilized CIBIC score and delayed cognitive deterioration among patients with vascular dementia as well as patients with Alzheimer disease and cerebrovascular
disease.113

Neuroleptics

Even people with mild dementia have a high prevalence of neuropsychiatric symptoms.114 A potential benefit of early detection of dementia is that these associated problem behaviors could be recognized and treated earlier with such drugs as neuroleptics. Although RCTs have examined these agents in people with more severe dementia, no study has examined this treatment in patients with mild to moderate dementia who would be detected by screening.

Antidepressants

Many people with mild to moderate dementia are depressed.114 Two RCTs provide evidence that antidepressants are effective for depressive symptoms among community-dwelling elderly persons with mild to moderate Alzheimer's disease. One RCT with a crossover design showed that 6 weeks of therapy with clomipramine (a tricyclic antidepressant) reduced depressive symptoms.34 In a recent study, Lyketsos and colleagues38 found that sertraline effectively treated depression in individuals with both Alzheimer's disease and major depression. 

Although research demonstrates a positive effect of antidepressants on depression, it is not clear that these drugs modify the progression of dementia. No high-quality trial has examined the effect of antidepressants on health outcomes such as cognition, functional status, health-related quality of life, or clinician global impression of change.

How Effective Are Nonpharmacologic Interventions for People with Mild to Moderate Dementia and Their Caregivers?

Nonpharmacologic interventions for dementia include behavioral training, caregiver education, and supportive services. Nonpharmacologic interventions may be directed at either patients or their caregivers. Numerous studies have targeted patients with severe dementia; no study has involved people with mild to moderate dementia. Caregiver interventions are complex and varied but usually include 1 or more of the following components: support groups, individual or family counseling, skills training, or education. Interventions targeted to caregivers are usually studied for benefit to patients as well as benefit to caregivers themselves. 

One systematic review53 and five well-conducted RCTs54-58,115 have examined interventions directed at caregivers of people with mild to moderate dementia (Table 5). The systematic review found no significant differences in caregiver burden between intervention and control groups and concluded that little or no evidence exists that interventions to support caregivers of people with Alzheimer's disease yield quantifiable benefit.53 One other RCT had similar findings.115 The other four studies involved multi-component interventions, and all had some positive results. Two of these focused on caregiver outcomes and found modest benefits.57,58 Two other studies found that intensive, comprehensive caregiver interventions enabled caregivers to maintain the affected persons at home for a substantially longer period of time (between 11 and 19 months) than those who did not receive the intervention.59,116 

Thus, several types of interventions targeted to caregivers showed positive impact on varied outcomes. Because of the heterogeneity of the interventions, it is difficult to determine the optimal components of these interventions. All of these studies were conducted among clinically detected patients who required caregivers. The extent to which such interventions would be useful for family members of people with milder degrees of dementia, detected by screening, is not clear.

Does Earlier Knowledge of a Dementia Diagnosis Improve Patient and Family Planning for Future Medical Care and Safety?

Individuals identified with early dementia by screening may have the opportunity to discuss the nature of the syndrome, its prognosis, and future planning with regard to health care, safety, and finances. They may be able to formulate advance directives; choose a person to exercise power of attorney for financial and personal care decision making; consent to participate in research; and contemplate issues such as motor vehicle driving, self-neglect, financial victimization, and housing relocation. Screening may also permit earlier and more effective treatment of co-existing conditions by improving medication adherence and avoiding drug interactions. No high-quality study has been done to verify, quantify, or refute these potential benefits. 

What are the Adverse Effects of Screening and Early Treatment of Dementia?

The harms of dementia screening have not been systematically studied. Potential harms include risk of depression and anxiety, the time and cost of screening, and possible labeling effects. Because 50 percent or more of patients with a positive screening test will not meet criteria for dementia, any screening program must be linked to a source of diagnostic interviews. Although these interviews could be done by primary care physicians, relatively few nonspecialist physicians engage in such interviews as a standard part of practice. Patients and families may face delays after a positive screen before they can obtain referral for a reference standard diagnostic work-up, potentially leading to increased anxiety and worry. 

Screening will also result in the detection of persons with MCI, a newly recognized condition that increases the risk of developing dementia.117 No well-conducted RCT, however, provides information about an effective treatment for such individuals. Labeling such persons with a disease could potentially cause unnecessary anxiety. Once a patient receives a diagnosis of dementia, he or she is unlikely to qualify for long-term care insurance or acceptance into a continuous-care retirement community. Despite these potential hardships, however, surveys of elderly patients and caregivers of patients with Alzheimer disease show that most persons want to be told of a dementia diagnosis.118, 119

The potential harms of treatment apply primarily to drugs, both cholinesterase inhibitors and others. Common side effects experienced by people taking cholinesterase inhibitors are nausea, vomiting, weight loss, and diarrhea. In the trials of galantamine, the dropout rate attributable to adverse events in participants ranged from 2% to 15% higher in the drug group than the placebo group. In the trials of rivastigmine, the adverse effect rate was from 5% to 20% higher and about 8% higher, respectively, in patients receiving the drugs. Tacrine has significant gastrointestinal and hepatic side effects, and the odds ratio for adverse event–related dropout among persons who took this drug was 5.7 (95% CI, 4.1 to 7.9). However, in general, fewer than 20% of patients stop taking cholinesterase inhibitors because of side effects. In RCTs of other drugs, dropout rates did not differ significantly between persons who took Ginkgo biloba, selegiline, or vitamin E and those who took placebo. 

Return to Table of Contents

The prevalence of dementia increases rapidly in the seventh and eighth decades of life; the condition affects 25 to 47 percent of people older than 85. Patients suffer from progressive cognitive and functional dependence, psychotic and depressive symptoms, and injuries. The burden of disease also extends to caregivers, who have high rates of emotional and financial stress and depression. Among all primary care patients older than 65 who have dementia, approximately one-half are undiagnosed. 

No randomized trial has evaluated the efficacy of dementia screening in primary care. The MMSE is the best-studied brief screening tool for dementia, but it has limited specificity when the cutpoint is set for higher sensitivity, and scores must be adjusted for age and educational attainment. Other patient-based cognitive screening tests have similar characteristics. Although many of these tests take 10 minutes or less to administer, a screening program would require additional time for diagnostic interviews and patient and family counseling.

The most important problem with the evidence for screening for dementia is the uncertainty about the effectiveness of treatment for people whose disease would be detected by screening. Cholinesterase inhibitor treatment of people clinically detected with mild to moderate Alzheimer's disease provides minimal impact on functional status but a modest and consistent tendency in some people to stabilize cognition and the clinician's global impression of change. The literature makes it difficult to define clearly how many people with early dementia benefit from this treatment, and by how much. The degree to which this evidence can be extrapolated to the situation of people detected by screening is uncertain. Other pharmacologic treatments have not been adequately studied.

Limited evidence indicates that intensive, multi-component interventions to support caregivers may delay nursing home placement for people with Alzheimer's disease, but they have demonstrated few direct benefits for either patients or caregivers. The relevance of this finding for the families of people with screening-detected dementia, who presumably do not yet have caregivers, is unclear. Additional benefits of screening, including individual and family planning and better decisions about health care interventions for other conditions, have not been studied.

Important gaps remain in our knowledge about screening for and early treatment of dementia. An RCT of screening for dementia in primary care with prospective evaluation of multiple health outcomes would provide the best evidence of benefits and harms. A trial of screening is justifiable given the high prevalence of undiagnosed dementia among primary care patients over age 65 and the efficacy of cholinesterase inhibitor treatment for clinically detected mild to moderate Alzheimer's disease. Such a trial should also monitor costs and harms and include the effects of screening and treatment on cognition, function, health care utilization, health-related quality of life, and caregiver burden.  The MMSE has been criticized for limited specificity and the need to adjust scoring for age and educational attainment. Future research should examine other promising brief screening tools that may be less education dependent, testing their positive and negative predictive value in primary care.

Although caregiver burden, increased use of health care, problem behaviors, psychiatric symptoms, and accidental injury are common in early dementia, little research to date has dealt with treatments to address these important aspects of the syndrome. The value of nonpharmacologic as well as pharmacologic interventions in the situation of early dementia merits further study. Outcome measures should be reported in order to clarify how many people benefit from these interventions and by how much. Because of the progressive nature of dementia, outcome measures that incorporate time, such as the time to decline or survival analyses, are most appropriate. 

Although many uncertainties remain, the concept of detecting dementia at an early stage to allow interventions is a good one. Interventions are the only means to modify the otherwise certain decline of people with dementia. The idea of screening for dementia is worth pursuing with further research.

Return to Table of Contents

Source: Boustani M, Peterson B, Hanson L, Harris R, Lohr K. Screening for Dementia in Primary Care: A summary of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med June 3, 2003;138(11):927-37.

Acknowledgements: Dr. Boustani would like to thank the Program on Aging, John A. Hartford Foundation, New York City and the American Federation for Aging Research for their career development support. We thank our liaisons from the U.S. Preventive Services Task Force—Cynthia Mulrow, M.D., M.Sc., of the University of Texas Health Sciences Center, San Antonio, TX; and Albert Siu, M.D., M.S.P.H., of Mount Sinai Medical Center, New York, NY—for their assistance on the full systematic evidence review from which this article has been developed. We acknowledge David Atkins, M.D., M.P.H., Director, AHRQ Clinical Prevention Program, for his advice and counsel throughout this project. Finally, we thank our RTI-UNC EPC colleagues Sonya Sutton, B.S.P.H., and Loraine Monroe (both of RTI) and Carol Krasnov and Audrina Bunton (both of the University of North Carolina) for their assistance in this project and production of this article.

Grant Support: This study was conducted by the RTI–University of North Carolina Evidence-based Practice Center under contract to the Agency for Healthcare Research and Quality, Rockville, Maryland (contract no. 290-97-0011, task order 3). Dr. Boustani has received career development support from the Program on Aging, John A. Hartford Foundation, and the American Federation for Aging Research.

Potential Financial Conflicts of Interest: Grants received: M. Boustani (Pfizer, Inc.).

Return to Table of Contents

1. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington, DC: American Psychiatric Assoc; 1994.
2. Boustani M, Peterson B, Hanson L, Harris R, Krasnov C. Screening for Dementia. A Systematic Evidence Review. Available at http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=es20. Rockville, MD: Agency for Healthcare Research and Quality; 2002.
3. Lautenschlager NT, Cupples LA, Rao VS, Auerbach SA, Becker R, Burke J, et al. Risk of dementia among relatives of Alzheimer's disease patients in the MIRAGE study: What is in store for the oldest old? Neurology 1996;46:641-50.[PMID: 8618660]
4. Blacker D, Tanzi R. The genetics of Alzheimer disease: current status and future prospects. Arch Neurol 1998;55:294-96. [PMID: 9520001]
5. Lai F, Williams R. A prospective study of Alzheimer disease in Down syndrome. Arch Neurol 1989;46:849-53. [PMID: 2527024]
6. Longstreth W Jr, Bernick C, Manolio T, Bryan N, Jungreis C, Price T. Lacunar infarcts defined by magnetic resonance imaging of 3660 elderly people: the Cardiovascular Health Study. Arch Neurol 1998;55:1217-25. [PMID: 9740116]
7. Hofman A, Ott A, Breteler MM, Bots ML, Slooter AJ, van Harskamp F, et al. Atherosclerosis, apolipoprotein E, and prevalence of dementia and Alzheimer's disease in the Rotterdam Study. Lancet 1997;349:151-54. [PMID: 9111537]
8. U.S. Office of Technology Assessment. Losing a Million Minds: Confronting the Tragedy of AD and Other Dementias. Washington, DC: US Government Printing Office; 1987.
9. Evans DA, Funkenstein HH, Albert MS, Scherr PA, Cook NR, Chown MJ, et al. Prevalence of Alzheimer's disease in a community population of older persons. Higher than previously reported. JAMA 1989;262:2551-56. [PMID: 2810583]
10. Evans D, Smith L, Scherr P, Albert M, Funkenstein H, Hebert L. Risk of death from Alzheimer's disease in a community population of older persons. Am J Epidemiol 1991;134:403-12. [PMID: 1877601]
11. U.S. General Accounting Office. Alzheimer's Disease: Estimates of Prevalence in the United States. Washington, DC: U.S. General Accounting Office; 1998. Publication HEHS 98-16.
12. Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW, Feldman H, et al. The recognition, assessment and management of dementing disorders: conclusions from the Canadian Consensus Conference on Dementia. CMAJ 1999;160(Suppl 12):S1-S15. [PMID: 10410645]
13. Canadian study of health and aging: study methods and prevalence of dementia. CMAJ 1994;150:899-913. [PMID: 8131123]
14) Brookmeyer R, Gray S, Kawas C. Projections of Alzheimer's disease in the United States and the public health impact of delaying disease onset. Am J Public Health 1998;88:1337-42. [PMID: 9736873]
15. Brodaty H, Clarke J, Ganguli M, Grek A, Jorm AF, Khachaturian, et al. Screening for cognitive impairment in general practice: toward a consensus. Alzheimer Dis Assoc Disord 1998;12:1-13. [PMID: 9539404]
16. Ernst RL, Hay JW. The US economic and social costs of Alzheimer's disease revisited. Am J Public Health 1994;84:1261-64. [PMID: 8059882]
17. Dunkin J, Anderson-Hanley C. Dementia caregiver burden: a review of the literature and guidelines for assessment and intervention. Neurology 1998;51(Suppl 1):S53-S60. [PMID: 967476
18. Grafström M, Fratiglioni L, Sandman P, Winblad B. Health and social consequences for relatives of demented and non-demented elderly. A population-based study. J Clin Epidemiol 1992;45:861-70.[PMID: 1624968]
19. Schulz R, O'Brien A, Bookwala J, Fleissner K. Psychiatric and physical morbidity effects of dementia caregiving: prevalence, correlates, and causes. Gerontologist 1995;35:771-91. [PMID: 8557205]
20. Hoyert DL, Kochanek KD, Murphy SL. Deaths: final data for 1997. Natl Vital Stat Rep 1999;47:1-104. [PMID: 10410536]
21. Gold DP, Reis MF, Markiewicz D, Andres D. When home caregiving ends: a longitudinal study of outcomes for caregivers of relatives with dementia. J Am Geriatr Soc 1995;43:10-16. [PMID: 7806732]
22. Arno PS, Levine C, Memmott MM. The economic value of informal caregiving. Health Aff (Millwood) 1999;18:182-8. [PMID: 10091447]
23. O'Connor D, Pollitt P, Hyde J, Brook C, Reiss B, Roth M. Do general practitioners miss dementia in elderly patients? Br Med J 1988;297:1107-10. [PMID: 3143447]
24. Lagaay A, van der Meij J, Hijmans W. Validation of medical history taking as part of a population based survey in subjects aged 85 and over. Br Med J 1992; 304:1091-92. [PMID: 1586823]
25. Cooper B, Bickel H, Schaufele M. Early development and progression of dementing illness in the elderly: a general-practice based study. Psycholog Med 1996;26:411-19. [PMID: 8685297
26. Olafsdottir M, Skoog I, Marcusson J. Detection of dementia in primary care: the Linkoping study. Dement Geriatr Cogn Disord 2000;11:223-29. [PMID: 10867449]
27. Valcour V, Masaki K, Curb J, Blanchette P. The detection of dementia in the primary care setting. Arch Intern Med 2000;160:2964-68. [PMID:
11041904]
28. U.S. Preventive Services Task Force. Guide to Clinical Preventive Services, 2nd ed. Alexandria, VA: International Medical Publishing;; 1996:531-40.
29. Petersen R, Stevens J, Ganguli M, Tangalos E, Cummings J, DeKosky S. Practice parameter: Early detection of dementia: Mild cognitive impairment (an evidence-based review): Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2001;56:1133-42. [PMID: 11342677]
30. Patterson CJ, Gass DA. Screening for cognitive impairment and dementia in the elderly. Can J Neurol Sci 2001;28(Suppl 1):S42-S51.[PMID: 11237310]
31. Harris RP, Helfand M, Woolf SH, Lohr KN, Mulrow CD, Teutsch SM, et al. Current methods of the U.S. Preventive Services Task Force: A review of the process. Am J Prev Med 2001;2:21-35. [PMID: 11306229]
32. Eefsting J, Boersma F, Van den Brink W, Van Tilburg W. Differences in prevalence of dementia based on community survey and general practitioner recognition. Psycholog Med 1996;26:1223-30. [PMID: 8931168]
33. Sternberg SA, Wolfson C, Baumgarten M. Undetected dementia in community-dwelling older people: the Canadian Study of Health and Aging. J Am Geriatr Soc 2000;48:1430-34. [PMID: 11083319
34. Petracca G, Teson A, Chemerinski E, Leiguarda R, Starkstein S. A double-blind placebo-controlled study of clomipramine in depressed patients with Alzheimer's disease. J Neuropsychiatry Clin Neurosci 1996;8:270-75. [PMID: 8854297]
35. Auchus A, Bissey-Black C. Pilot study of haloperidol, fluoxetine, and placebo for agitation in Alzheimer's disease. J Neuropsychiatry Clin Neurosci 1997;9:591-93.[PMID: 9447502]
36. Devanand DP, Marder K, Michaels KS, Sackeim HA, Bell K, Sullivan MA, et al. A randomized, placebo-controlled dose-comparison trial of haloperidol for psychosis and disruptive behaviors in Alzheimer’s disease. Am J Psychiatry. 1998;155:1512-20. [PMID: 9812111]
37. Costa PT Jr, Williams TF, Somerfield M, Albert MS, Nutters NM, Folstein M, et. al. Recognition and Initial Assessment of Alzheimer’s Disease and Related Dementias. Clinical Practice Guideline no. 19. Rockville, MD: U.S. Department of Health and Human Services, Public Health Service, Agency for Health Care Policy and Research; 1996. AHCPR publication no. 97-0702.
38. Lyketsos CG, Sheppard JM, Steele CD, Kopunek S, Steinberg M, Baker AS,, et al. Randomized, placebo-controlled, double-blind clinical trial of sertraline in the treatment of depression complicating Alzheimer's disease: initial results from the Depression in Alzheimer's Disease study. Am J Psychiatry 2000;157:1686-9. [PMID: 11007727]
39. Jitapunkul S, Lailert C, Worakul P, Srikiatkhachorn A, Ebrahim S. Chula Mental Test: A screening test for elderly people in less developed countries. Int J Geriatr Psychiatry 1996;11:714-20.
40. McDowell I, Kristjansson B, Hill GB, Hebert R. Community screening for dementia: the Mini Mental State Exam (MMSE) and Modified Mini-Mental State Exam (3MS) compared. J Clin Epidemiol 1997;50:377-83. [PMID: 9179095]
41. Lindeboom J , Launer LJ, Schmand BA, Hooyer C, Jonker C. Effects of adjustment on the case-finding potential of cognitive tests. J Clin Epidemiol 1996;49:691-95. [PMID: 8656232]
42. Law S, Wolfson C. Validation of a French version of an informant-based questionnaire as a screening test for Alzheimer's disease. Br J Psychiatry 1995;167:541-44. [PMID: 8829727]
43. Braekhus A, Laake K, Engedal K. A low, 'normal' score on the Mini-Mental State Examination predicts development of dementia after three years. J Am Geriatr Soc 1995;43:656-61. [PMID: 7775725]
44. Teri L, Logsdon RG, Peskind E, Raskind M, Weiner MF, Tractenberg RE, et al. Treatment of agitation in AD: a randomized, placebo-controlled clinical trial. Neurology. 2000;55:1271-8. [PMID: 110877]
45. Solomon PR, Brush M, Calvo V, Adams F, DeVeaux RD, Pendlebury WW, et al. Identifying dementia in the primary care practice. Int Psychogeriatr 2000;12:483-93. [PMID: 11263715]
46. Oken B, Storzbach D, Kaye J. The efficacy of Ginkgo biloba on cognitive function in Alzheimer disease. Arch Neurol 1998;55:1409-15. [PMID: 9823823]
47. Birks J, Flicker L. Selegiline for Alzheimer's disease (Cochrane Review). Cochrane Database Syst Rev 2000;CD000442.[PMID: 10796544]
48. 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;336:1216-22. [PMID: 9110909]
49. 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. JAMA 2000;283:1007-15. [PMID: 10697060]
50. Asthana S, Baker LD, Craft S, Stanczyk FZ, Veith RC, Raskind MA, et al. High-dose estradiol improves cognition for women with AD: results of a randomized study. Neurology 2001;57:605-12. [PMID: 11524467]
51.Pantoni L, Rossi R, Inzitari D, Bianchi C, Beneke M, Erkinjuntti T, et al. Efficacy and safety of nimodipine in subcortical vascular dementia: a subgroup analysis of the Scandinavian Multi-Infarct Dementia Trial. J Neurol Sci 2000;175:124-34.[PMID: 10831773]
52. Williams P, Rands G, Orrel M, Spector A. Aspirin for vascular dementia. Cochrane Database Syst Rev 2000;CD001296. [PMID: 11034710]
53. Thompson C, Briggs M. Support for carers of people with Alzheimer’s type dementia. Cochrane Database Syst Rev. 2000:CD000454. [PMID: 10796545]
54. Hebert R, Leclerc G, Bravo G, Girouard D, Lefrancois R. Efficacy of a support group programme for caregivers of demented patients in the community: a randomized controlled trial. Arch Gerontol Geriatr 1994;18:1-14.
55. Hebert R, Girouard D, Leclerc G, Bravo G, Lefrancois R. The impact of a support group programme for care-givers on the institutionalization of demented patients. Arch Gerontol Geriatr 1995;20:129-34.
56. Mittelman MS, Ferris SH, Shulman E, Steinberg G, Levin B. A family intervention to delay nursing home placement of patients with Alzheimer disease. A randomized controlled trial. JAMA 1996;276:1725-31. [PMID: 8940320]
57. McCurry SM, Logsdon RG, Vitiello MV, Teri L. Successful behavioral treatment for reported sleep problems in elderly caregivers of dementia patients: a controlled study. J Gerontol B Psychol Sci Soc Sci 1998;53:P122-9. [PMID: 9520929]
58. 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;176:557-62. [PMID: 10974962]
59. Brodaty H, Gresham M, Luscombe G. The Prince Henry Hospital dementia caregivers' training programme. Int J Geriatr Psychiatry 1997;12:183-92. [PMID: 9097211]
60. Barberger-Gateau P, Fabrigoule C, Helmer C, Rouch I, Dartigues J. Functional impairment in instrumental activities of daily living: an early clinical sign of dementia? J Am Geriatr Soc 1999;47:456-62. [PMID: 10203122
61. Heun R. Validity of family history diagnosis for dementia. Genet Epidemiol 1999;17:151-4. [PMID: 10414558]
62. Lanctot KL, Best TS, Mittmann N, Liu BA, Oh PI, Einarson T, et al. Efficacy and safety of neuroleptics in behavioral disorders associated with dementia. J Clin Psychiatry 1998;59:550-61. [PMID: 9818639]
63. Davidson M, Weiser M, Soares K. Novel antipsychotics in the treatment of psychosis and aggression associated with dementia: A meta-analysis of randomized controlled clinical trials. Int Psychogeriatr 2000;12(Suppl 11):271-77.
64. Qizilbash N, Birks J, Lopez Arrieta J, Lewington S, Szeto S. Tacrine for Alzheimer’s disease. Cochrane Database Syst Rev. 2000:CD000202. [PMID: 10908463]
65. Kirchner V, Kelly CA, Harvey RJ. Thioridazine for dementia. Cochrane Database Syst Rev. 2000:CD000464. [PMID: 10796547]
66. Burns A, Jacoby R, Levy R. Psychiatric phenomena in Alzheimer's disease. I: Disorders of thought content. Br J Psychiatry 1990;157:72-76, 92-94. [PMID:
2397365]
67. Maurer K, Ihl R, Dierks T, Frolich L. Clinical efficacy of Ginkgo biloba special extract EGb 761 in dementia of the Alzheimer type. J Psychiatr Res 1997;31:645-55. [PMID: 94475
68. Le Bars P, Katz M, Berman N, Itil T, Freedman A, Schatzberg A. A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA 1997;278:1327-32. [PMID: 9343463]
69. Aisen P, Davis K, Berg J, et al. A randomized controlled trial of prednisone in Alzheimer's disease. Alzheimer's Disease Cooperative Study. Neurology 2000;54:588-93. [PMID: 10680787]
70. Scharf S, Mander A, Ugoni A, Vajda F, Christophidis N. A double-blind, placebo-controlled trial of diclofenac/misoprostol in Alzheimer's disease. Neurology 1999;53:197-201. [PMID: 10408559]
71. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet 2000;356:2031-36. [PMID: 11145488]
72. Birks J, Iakovidou V, Tsolaki M. Rivastigmine for Alzheimer's disease. Cochrane Database Syst Rev. 2000:CD001191. [PMID: 11034705]
73. Birks J, Melzer D, Beppu H. Donepezil for mild and moderate Alzheimer's disease. Cochrane Database Syst Rev 2000;CD001190. [PMID: 11034704]
74. Qizilbash N, Whitehead A, Higgins J, Wilcock G, Schneider L, Farlow M. Cholinesterase inhibition for Alzheimer disease: a meta-analysis of the tacrine trials. Dementia Trialists' Collaboration. JAMA 1998;280:1777-82. [PMID: 9842955]
75. Olin J, Schneider L. Galantamine for Alzheimer's disease. Cochrane Database Syst Rev 2002;CD001747. [PMID: 12137632
76. Greenberg SM, Tennis MK, Brown LB, Gomez-Isla T, Hayden DL, Schoenfeld DA, et al. Donepezil therapy in clinical practice: a randomized crossover study. Arch Neurol 2000;57:94-99. [PMID: 10634454]
77. Burns A, Rossor M, Hecker J, Gauthier S, Petit H, Moller HJ, et al. The effects of donepezil in Alzheimer's disease—results from a multinational trial. Dement Geriatr Cogn Disord 1999;10:237-44. [PMID: 10325453]
78. Rosler M, Anand R, Cicin-Sain A, Gauthier S, Agid Y, Dal-Bianco P, et al. Efficacy and safety of rivastigmine in patients with Alzheimer's disease: international randomised controlled trial. BMJ 1999;318:633-38. [PMID: 10066203]
79. Mohs RC, Doody RS, Morris JC, Ieni JR, Rogers SL, Perdomo CA, et al. A 1-year, placebo-controlled preservation of function survival study of donepezil in AD patients. Neurology 2001;57:481-88. [PMID: 11502917]
80. Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G, Wimo A, et al. A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology 2001;57:489-95. [PMID: 11502918]
81. Ross GW, Abbott RD, Petrovitch H, et al. Frequency and characteristics of silent dementia among elderly Japanese-American men. The Honolulu-Asia Aging Study. JAMA 1997;277:800-5. [PMID: 8854297
82. Erkinjuntti T, Ostbye T, Steenhuis R, Hachinski V. The effect of different diagnostic criteria on the prevalence of dementia. N Engl J Med 1997;337:1667-74. [PMID: 9447502]
83. Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom J, Relkin N,, et al. Practice parameter: diagnosis of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2001;56:1143-53. [PMID: 11342678]
84. Wilder D, Cross P, Chen J, Gurland B, Lantigua R, Teresi J,, et al. Operating characteristics of brief screens for dementia in a multicultural population. Am J Geriatric Psychiatry 1995;3:96-107.
85. 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;13:368-80. [PMID: 9658272]
86. Pfeiffer E. A short portable mental status questionnaire for the assessment of organic brain deficit in elderly patients. J Am Geriatr Soc 1975;23:433-41. [PMID: 1159263]
87. Callahan CM, Hendrie HC, Tierney WM. Documentation and evaluation of cognitive impairment in elderly primary care patients. Ann Intern Med 1995;122:422-9. [PMID: 785699
88. Schulman K. Clock-drawing: is it the ideal cognitive screening test? Int J Geriatr Psychiatry 2000;15:548-61. [PMID: 10861923]
89. 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;16:935-40.[PMID: 11607936]
90. Scanlan JM, Brush M, Quijano C, Borson S. Comparing clock tests for dementia screening: naíve judgments vs formal systems—what is optimal? Int J Geriatr Psychiatry 2002;17:14-21. [PMID: 11802225]
91. Schramm U, Berger G, Muller R, Kratzsch T, Peters J, Frolich L. Psychometric properties of Clock Drawing Test and MMSE or Short Performance Test (SKT) in dementia screening in a memory clinic population. Int J Geriatr Psychiatry 2002;17:254-60. [PMID: 11921154]
92. Powlishta KK, Von Dras DD, Stanford A, et al. The clock drawing test is a poor screen for very mild dementia. Neurology 2002;59:898-903. [PMID: 12297574]
93. Teng EL, Chui HC. The Modified Mini-Mental State (3MS) examination. J Clin Psychiatry 1987;48:314-18. [PMID: 3611032]
94. Scanlan J, Borson S. The Mini-Cog: receiver operating characteristics with expert and naive raters. Int J Geriatr Psychiatry 2001;16:216-22. [PMID: 11241728
95. Frank RM, Byrne GJ. The clinical utility of the Hopkins Verbal Learning Test as a screening test for mild dementia. Int J Geriatr Psychiatry 2000;15:317-24. [PMID: 10767731]
96. Hogervorst E, Combrinck M, Lapuerta P, Rue J, Swales K, Budge M. The Hopkins Verbal Learning Test and screening for dementia. Dement Geriatr Cogn Disord 2002;13:13-20. [PMID: 11731710]
97. Jarvenpaa T, Rinne JO, Raiha I, Koskenvuo M, Lopponen M, Hinkka S, et al. Characteristics of two telephone screens for cognitive impairment. Dement Geriatr Cogn Disord 2002;13:149-55. [PMID: 11893836]
98. Lawrence J, Davidoff D, Katt-Lloyd D, Auerbach M, Hennen J. A pilot program of improved methods for community-based screening for dementia. Am J Geriatric Psychiatry 2001;9:205-11. [PMID: 11481127]
99. Mundt JC, Ferber KL, Rizzo M, Greist JH. Computer-automated dementia screening using a touch-tone telephone. Arch Intern Med 2001;161:2481-7. [PMID: 11700161]
100. Belle SH, Seaberg EC, Ganguli M, Ratcliff G, DeKosky S, Kuller LH. Effect of education and gender adjustment on the sensitivity and specificity of a cognitive screening battery for dementia: results from the MoVIES Project. Monongahela Valley Independent Elders Survey. Neuroepidemiology 1996;15:321-9. [PMID: 8930945]
101. Uhlmann RF, Larson EB. Effect of education on the Mini-Mental State Examination as a screening test for dementia. J Am Geriatr Soc 1991;39:876-80. [PMID: 1885862]
102. Fratiglioni L, Jorm AF, Grut M, Viitanen M, Holmen K, Ahlbom A, et al. Predicting dementia from the Mini-Mental State Examination in an elderly population: the role of education. J Clin Epidemiol 1993;46:281-7. [PMID: 8455053]
103. Freidl W, Schmidt R, Stronegger WJ, Irmler A, Reinhart B, Koch M. Mini mental state examination: influence of sociodemographic, environmental and behavioral factors and vascular risk factors. J Clin Epidemiol 1996;49:73-8. [PMID: 8598514]
104. Crum RM, Anthony JC, Bassett SS, Folstein MF. Population-based norms for the Mini-Mental State Examination by age and educational level. JAMA 1993;269:2386-91. [PMID: 8479064]
105. Apolipoprotein E genotyping in Alzheimer's disease. National Institute on Aging/Alzheimer's Association Working Group. Lancet 1996;347:1091-5. [PMID: 8602063]
106. Demers L, Oremus M, Perrault A, Wolfson C. Review of outcome measurement instruments in Alzheimer's disease drug trials: introduction. J Geriatr Psychiatry Neurol 2000;13:161-69. [PMID: 11128056]
107. Stern RG, Mohs RC, Davidson M, Schmeidler J, Silverman J, Kramer-Ginsberg E, et al. A longitudinal study of Alzheimer's disease: measurement, rate, and predictors of cognitive deterioration. Am J Psychiatry 1994;151:390-96. [PMID: 8109647]
108. Kramer-Ginsberg E, Mohs RC, Aryan M, Lobel D, Silverman J, Davidson M, et al. Clinical predictors of course for Alzheimer patients in a longitudinal
study: a preliminary report. Psychopharmacol Bull 1988;24:458-62. [PMID: 3153508]
109. Brooks JO 3rd, Yesavage JA, Taylor J, et al. Cognitive decline in Alzheimer's disease: elaborating on the nature of the longitudinal factor structure of the Mini-Mental State Examination. Int Psychogeriatr 1993;5:135-46. [PMID: 8292767]
110. Salmon DP, Thal LJ, Butters N, Heindel WC. Longitudinal evaluation of dementia of the Alzheimer type: a comparison of 3 standardized mental status examinations. Neurology 1990;40:1225-30. [PMID: 2381530]
111. Peripheral and Central Nervous System Drugs Advisory Committee Meeting. 17 July 1989. Document no. 227. Rockville, MD: Department of Health and Human Services, Public Health Service, U.S. Food and Drug Administration; 1989.
112. Roman GC. Vascular dementia revisited: diagnosis, pathogenesis, treatment, and prevention. Med Clin North Am 2002;86:477-99. [PMID: 12168556]
113. Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer's disease combined with cerebrovascular disease: a randomised trial. Lancet 2002;359:1283-90. [PMID: 11965273]
114. Lyketsos CG, Lopez O, Jones B, Fitzpatrick AL, Breitner J, DeKosky S. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the cardiovascular health study. JAMA 2002;288:1475-83. [PMID: 12243634]
115. Tappen RM. The effect of skill training on functional abilities of nursing home residents with dementia. Res Nurs Health 1994;17:159-65. [PMID: 8184127]
116. Mittelman MS, Ferris SH, Steinberg G, Shulman E, Mackell JA, Ambinder A, et al. An intervention that delays institutionalization of Alzheimer’s disease patients: treatment of spouse-caregivers. Gerontologist. 1993;33:730-40. [PMID: 8314099]
117. Morris J, Storandt M, Miller J, et al. Mild cognitive impairment represents early-stage Alzheimer disease. Arch Neurol 2001;58:397-405. [PMID: 11255443]
118. Drickamer MA, Lachs MS. Should patients with Alzheimer's disease be told their diagnosis? N Engl J Med 1992;326:947-51. [PMID: 1542346]
119. Jha A, Tabet N, Orrell M. To tell or not to tell-comparison of older patients' reaction to their diagnosis of dementia and depression. Int J Geriatr Psychiatry 2001;16:879-85. [PMID: 1157176]

Return to Table of Contents

The authors thank their liaisons from the U.S. Preventive Services Task Force—Cynthia Mulrow, MD, MSc, The University of Texas Health Science Center, San Antonio, Texas, and Albert Siu, MD, MSPH, Mount Sinai Medical Center, New York, New York—for their assistance with the full systematic evidence review. They also thank David Atkins, MD, MPH, Director, Agency for Healthcare Research and Quality Clinical Prevention Program, for his advice and counsel and Sonya Sutton, BSPH, and Loraine Monroe (both of RTI) and Carol Krasnov and Audrina Bunton (both of the University of North Carolina) for their assistance.

Return to Table of Contents

Grant Support: This study was conducted by the RTI–University of North Carolina Evidence-based Practice Center under contract to the Agency for Healthcare Research and Quality, Rockville, Maryland (contract no. 290-97-0011, task order 3). Dr. Boustani has received career development support from the Program on Aging, John A. Hartford Foundation, and the American Federation for Aging Research.

Potential Financial Conflicts of Interest: Grants received: M. Boustani (Pfizer, Inc.).

Return to Table of Contents

Key Question Eligibility Criteria Systematic Reviews (Reference) Additional Studies Not in Systematic Reviews (Reference)
All Published 1 January 1994–1 September 2002
English language.
Human subjects age ≥ 60 y
MEDLINE, PsycINFO,
EMBASE, Cochrane Library.
  n
1. Direct evidence of screening RCT of screening.
Health outcomes.
0 0
2. Prevalence of undiagnosed dementia Systematic reviews.
Cross-sectional prevalence.
Community or primary care setting.
Appropriate reference standard.
0 426,27,32,33
3. Accuracy of screening tests Systematic reviews; prospective cohorts.
Cross-sectional prevalence.
Community or primary care setting.
Appropriate reference standard.
137 938-43,45, 60, 61, 84
4. Efficacy of pharmacologic treatment Systematic reviews, RCTs.
Mild to moderate dementia.
Health outcomes.
12[[46, 47, 52,
62–66, 72–75]
1934–36, 38, 44, 48–51,67–71, 76–80
5. Efficacy of nonpharmacologic interventions
For patients



For caregivers



Systematic reviews, RCTs.
Mild to moderate dementia.
Health outcomes.

Systematic reviews, RCTs.
Mild to moderate dementia.
Health outcomes.



0



1[[53]

 



0



654–59

6. Efficacy of interventions for planning Systematic reviews, RCTs.
Mild to moderate dementia.
Health outcomes.
0 0
7. Harms of screening Systematic reviews, RCTs.
Mild to moderate dementia.
Psychological or other health outcomes.
0 0
8. Harms of treatment Systematic reviews, RCTs.
Mild to moderate dementia.
Health outcomes.
472,72-75 576-80

Note: RCT indicates randomized, controlled trial.
†Two articles were combined because they both report the results of one study.

Return to Table of Contents

Study, Year (Reference) Setting Age of Patient
Sample, y
Reference Standard Prevalence of
Undiagnosed Dementia
in All Patients, %
Olafsdottir, et al., 200026 Primary health center, Sweden > 70 years DSM-III-R 12.0
Eefsting, et al., 199632 Community and general practices, Netherlands ≥ 65 years DSM-III 3.2
Valcour, et al., 200027 General internal medicine clinic, Hawaii ≥ 65 years DSM-III-R 5.7
Sternberg, et al., 200033 General, randomly sampled community, Canada ≥ 65 years DSM-III-R 1.8

Note: DSM-III indicates Diagnostic and Statistical Manual, third edition; DSM-III-R, Diagnostic and Statistical Manual, third edition: revised.

Return to Table of Contents

Instrument (Reference) Sensitivity, % Specificity, % Positive
predictive value, %
Negative
predictive value, %
MMSE37,39-43,84,45 71-92 56-96 15-72 95-99
FAQ37 90 90 50 99
BIMC37,84 90 65-90 22-50 98-99
BOMC37 69 90 43 96
STMS37 81 90 47 98

Note:BIMC = Blessed Information Memory Concentration; BOMC = Blessed Orientation Memory Concentration; FAQ = Functional Activities Questionnaire; MMSE = Mini-Mental Status Examination; STMS = Short Test of Mental Status.
† Based on a dementia prevalence of 10%.

Return to Table of Contents

Drug (Reference) Patients, n Effect on Cognitive Function as Assessed by the Mean Difference between Drug and Placebo Groups on the ADAS-Cog†‡ Range of Odds Ratio for Having Stable Global Function or Better after Drug Treatment Compared with Placebo, as Measured by the CIBIC Effect on Physical Function as Assessed by IADLs
Tacrine74 1984 1.36-2.78 1.18-2.11 Inconclusive
Donepezil73,76,77,79,80 1980 2.12-3.01 2.04-2.63 Inconclusive
Rivastigmine72,78 4095 2.28-2.4 1.4-2.36 Inconclusive
Galantamine75 1674 3.00-3.4 1.71-1.94 Inconclusive

Note: ADAS-Cog = Alzheimer’s Disease Assessment Scale for Cognition; CIBC = Clinician’s Interview-Based Impression of Change; IADL = instrumental activity of daily living.
All between-group differences were statistically significant.
The values represent the range of differences in the ADAS-Cog between the treatment and placebo groups.

Return to Table of Contents

Study, Year (Reference) Patients, n Patient Outcomes Caregiver Outcomes Time to Nursing Home Placement
Hebert, et al., 199454
45 Not significant Not significant Not significant
Hebert, et al., 199555        
Mittleman, et al., 199656 206 Not tested Not tested Significant
Brodaty, et al., 199759 96 Not tested Not tested Significant
McCurry, et al., 199857 36 Not significant Significant Not tested
Marriott, et al., 200058 42 Not significant Significant Not tested

* Significance and non-significance were measured statistically.
Cognitive, functional, and behavioral symptoms.
Included caregiver burden, depressive symptoms, and reaction to patients’ behavioral problems in the trials by Hebert et al.; caregiver burden, depressive symptoms, sleep problems, and reaction to patients’ behavioral problems in the trial by McCurry et al.; and caregiver burden and depressive symptoms in the trial by Marriott et al.

Return to Table of Contents

The RTI-UNC Evidence-based Practice Center, together with members of the U.S. Preventive Services Task Force (USPSTF), sought to clarify issues concerning screening for the dementia syndrome (hereafter, dementia) by performing a systematic review of the relevant scientific literature on this topic.

Analytic Framework

The systematic evidence review examined the evidence for screening for dementia. This summary of the evidence utilizes the information from the systematic evidence review, including the analytic framework. Appendix Figure 1 presents the analytic framework that we used to guide our literature search. The analytic framework describes the logical chain that must be supported by evidence to link screening to improved health outcomes. Each arrow in the analytic framework represents a key question (Appendix Table 1), and we searched systematically for evidence concerning each key question in the analytic framework.

The analytic framework begins on the left side of the figure with a primary care population at risk for dementia and moves to the right. The first key question (represented by the overarching arrow) examines direct evidence that screening improves health outcomes. The health outcomes of interest are improved function (including cognitive, social, and physical); reduced hospitalizations, institutionalizations, and health care visits; fewer behavioral problems; reduced caregiver stress; fewer injuries; and improved health-related quality of life. Since we found no such overview studies, we continued to examine the indirect evidence in the following key questions, represented as linkages in the analytic framework.

Key questions 2 and 3 examine the yield of screening, involving both the prevalence of undiagnosed dementia in the population and the accuracy and reliability of various screening tests. Farther to the right in the analytic framework, Key question 4 examines the efficacy of various pharmacologic treatments, including treatment with antihypertensives or aspirin to prevent the progression of vascular dementia; treatment of people with Alzheimer's disease with cholinesterase inhibitors; and treatment of people with Alzheimer's disease with other drugs, neuroleptics, or antidepressants. Key question 5 examines the effectiveness of nonpharmacologic interventions for patients or caregivers; Key question 6 involves the effect of knowledge about the diagnosis of dementia on family and individual planning.

The critical issue is the efficacy of these treatments among people who would be detected by screening. Many studies examine treatment for people with clinically detected dementia; these are useful only insofar as they allow extrapolation to the efficacy of treatment at detection by screening. Furthermore, arrows 4 through 6 (and key questions 4-6) actually imply that the issue of interest is the added efficacy of initiating treatment after screening detection as opposed to initiation after clinical detection.

At the far right in the analytic framework are the health outcomes. In the end, the indirect evidence must allow a reasonable estimation of the magnitude of benefit in these outcomes that is attributable to screening. At the bottom of the analytic framework is linkage and key question 7, the issue of the harms of screening (for example, labeling), and key question 8, concerning the harms of treatment (for example, side effects).

Eligibility Criteria for Admissible Evidence

The EPC staff and Task Force liaisons developed eligibility criteria for selecting the evidence relevant to answer the key questions (Appendix Table 2). For key question 1, we required a well-conducted RCT of screening of adequate size and length to estimate health outcomes with reasonable accuracy. For key questions 2 and 3, we required either cross-sectional or cohort studies in which screening tests were performed on a primary care or general unselected population compared with an acceptable reference standard. For key questions 4 through 6, we accepted RCTs of treatments with health outcomes that provided information about the severity of participants with dementia. For key questions 7-8, we required RCTs of screened (or treated) versus non-screened (or non-treated) groups.

Literature Search Strategy, Results, and Review of Abstracts/Articles

The analytic framework and key questions guided our literature searches. We examined the critical literature described in the previous USPSTF review of this topic (published in 1996)28 and used our eligibility criteria to develop search terms. We used the search terms to search MEDLINE and the Cochrane Library for English-language articles that met our inclusion criteria and were published between 1 January 1994, and 1 September 2002. We also examined the bibliographies of pertinent articles and contacted experts for other references. When we found that a key question could best be answered by older literature, we also examined these studies. When we found that a systematic review used acceptable methods and that its studies met our criteria, we used the review instead of the individual studies. The search strategy and results are shown in Appendix Table 2 and Appendix Figures 1-8. All searches started with the term dementia and then proceeded by adding other terms as appropriate.

The first author and at least one other coauthor reviewed all abstracts found through our searches to determine which met our eligibility criteria. When either reviewer thought that an abstract might meet criteria, the article was copied for full review. The first author and at least one other coauthor reviewed each full article. Those that met eligibility criteria after full review (and discussion, when necessary) were abstracted. We critically appraised each study using criteria developed by the USPSTF.31 If we found a study or systematic review that met criteria but that contained a methodological flaw that invalidated its findings, we excluded it from further review. Abstracted articles or systematic reviews that met eligibility criteria and had no fatal flaws were entered into predesigned evidence tables (go to Appendix B in the systematic evidence review "Screening for the Dementia Syndrom"e, on the USPSTF Web site [https://www.uspreventiveservicestaskforce.org]).

Development of the Systematic Evidence Review and Summary of the Evidence Article

The authors presented an initial work plan including a provisional analytic framework and key questions to the entire Task Force, and also presented interim reports at subsequent meetings. The Task Force discussed and made important contributions to the review on several occasions. The two Task Force liaisons participated in every phase of the review, including conference calls to discuss critical parts of the evidence.

A draft systematic evidence review was presented to the Task Force and then sent for broad peer review. The peer review included individual experts in the field, representatives of relevant professional organizations, and representatives of appropriate federal agencies. We made revisions to the evidence review as appropriate after receiving peer review comments. The Task Force reviewed all information and voted on a recommendation. We then put the systematic evidence review into final form and from it developed the manuscript for journal publication.

Return to Table of Contents

  1. Is there direct evidence from a randomized controlled trial (RCT) of screening that screening for dementia improves health outcomes?
  2. How common is undiagnosed dementia?
  3. How accurate are the screening tests?
  4. What is the added efficacy of initiating the pharmacologic treatments below at screening detection compared with clinical detection in improving health outcomes?
    • Antihypertensives and aspirin for vascular dementia.
    • Cholinesterase inhibitors for Alzheimer's disease.
    • Other drugs (e.g., Ginkgo biloba, selegiline, vitamin E, estrogen) for Alzheimer's disease.
    • Neuroleptics.
    • Antidepressants.
  5. What is the efficacy of nonpharmacologic interventions for people with mild to moderate dementia and their caregivers?
  6. Does earlier knowledge of the diagnosis of dementia improve patient and family planning for future care and safety?
  7. What are the harms of screening?
  8. What are the harms of treatment?
Return to Table of Contents

Key QuestionInclusion CriteriaSystematic Reviews Found, nFull Articles Reviewed, nSystematic Reviews and Articles that Met Criteria, n
AllDementia syndrome.
Published 1 January 1994--1 September 2002
English language.
Human subjects, age ≥60y
Cochrane Library, MEDLINE, PsycINFO, EMBASE.



1. Direct evidence of screeningRCTs
Mass screening
000
2. Prevalence of undiagnosed dementiaSystematic reviews.
Cross-sectional prevalence.
Age ≥60 y in community or outpatient setting.
Blinded, independent evaluation for dementia syndrome diagnosed by DSM-III, DSM-III-R, or DSM-IV
Exclusion of subjects with prior diagnosis of dementia syndrome.
Valid assessment of lack of previous diagnosis.
0MEDLINE: 6
PsycINFO: 1
Total: 7
Systemic Reviews: 0
Additional Studies: 4
Total: 4
3. Accuracy of screening testsSystematic reviews.
Age ≥60 y in community or outpatient setting.
Blinded, independent evaluation for dementia syndrome diagnosed by DSM-III, DSM-III-R, or DSM-IV
Exclusion of subjects with prior diagnosis of dementia syndrome.
Data provided for true-positive, true-negative, false-positive, and false-negative results
Cochrane Library: 1
PsycINFO: 1
Other: 1
Total: 3
MEDLINE: 44
PsycINFO: 21
Other: 10
Total: 75
Systemic Reviews: 1
Additional Studies: 9
Total: 10
4. Efficacy of pharmacologic treatmentSystematic reviews.
RCTs.
Age ≥60 y in community or outpatient setting.
Reference standard for all patients.
Intervention one of defined key questions.
Any of the 6 outcomes from the analytic framework.
Cochrane Library: 12
MEDLINE®: 21
EMBASE: 3
Total: 36
Cochrane Library: 0
MEDLINE®: 232
PsycINFO®: 7
EMBASE: 27
Other: 33
Total: 299
Reviews: 12
RCTs: 19
Total: 31
5. Efficacy of nonpharmacologic treatmentSystematic reviews.
RCTs.
Age ≥60 y in community or outpatient setting.
Reference standard for all patients.
Intervention directed at patients or caregivers.
Health outcomes.
Cochrane Library: 0
MEDLINE: 1
EMBASE: 0
Total: 1
Cochrane Library: 0
MEDLINE: 6
PsycINFO: 0
EMBASE: 0
Total: 6
Reviews: 1
RCTs: 6
Total: 7
6. Efficacy of Interventions for PlanningSystematic reviews.
RCTs.
Age ≥60 y in community or outpatient setting.
Reference standard for all patients.
Improved planning for future care.
000
7. Harms of screeningSystematic reviews.
RCTs.
Age ≥60 yr in community or outpatient setting.
Reference standard for all patients.
Any treatment.
Any possible harms of screening.
000
8. Harms of treatmentSystematic reviews.
RCTs.
Age ≥60 y in community or outpatient setting.
Reference standard for all patients.
Any treatment.
Any possible harms of treatment.
Cochrane Library: 12
MEDLINE: 9
EMBASE: 2
Total: 23
Cochrane Library: 0
MEDLINE: 228
EMBASE: 20
PsycINFO: 7
Other: 2
Total: 257
Reviews: 9
RCTs: 10
Total: 19

Note: DSM-III = Diagnostic and Statistical Manual of Mental Disorders, third edition; DSM-III-R = Diagnostic and Statistical Manual of Mental Disorders, third edition, revised;
DSM-IV = Diagnostic and Statistical Manual of Mental Disorders, fourth edition; RCT = randomized, controlled trial.

Return to Table of Contents