CLINICAL REVIEW Guidelines for the Management of Cognitive and Behavioral Problems in Dementia Carl H. Sadowsky, MD, and James E. Galvin, MD, MPH Family physicians play a crucial role in the management and ongoing care of patients with Alzheimer disease (AD). This article reviews the effects of nonpharmacologic and pharmacologic interventions on the functional abilities and behavior of patients with dementia and how these can be implemented into clinical practice. Nonpharmacologic interventions are recommended as the initial strategy for managing problematic behaviors. Strategies for improving behavior include ensuring that the patient’s environ- ment is safe, calm, and predictable; removing environmental stressors; and identifying and avoiding situations that agitate or frighten the patient. Simple interventions include redirecting and refocusing the patient, increasing social interaction, establishing regular sleep habits, eliminating sources of con- flict and frustration, and establishing rewards for successes. The effectiveness of long-term behavioral management is largely dependent on the caregiver; as such, it is important to assess the role and needs of the caregiver. Because currently available therapies cannot reverse the pathologic processes of AD, the primary objective of pharmacotherapy is to preserve cognitive and functional ability, minimize behavioral dis- turbances, and slow disease progression. Cholinesterase inhibitors represent first-line therapy for pa- tients with mild to moderate AD, whereas a glutamate N-methyl D-aspartate antagonist is used in the treatment of moderate to severe AD. Looking forward, there are a number of therapies in development aimed at modifying the disease course; these include amyloid-lowering drugs, �-based and neuropro- tective approaches, acetylcholine agonists, and mitochondrial inhibitors. ( J Am Board Fam Med 2012; 25:350 –366.) Keywords: Dementia, Guidelines, Managed Care, Pharmacologic Therapy The Role of the Family Physician in Treating Dementia Primary care is the point of first medical contact for people with dementia and hence the cornerstone of ensuring early detection, timely intervention, and effective ongoing management.1 Inadequate detec- tion and poor management have been reported globally,2,3 leading to people with dementia and their families being denied optimal pharmacologic4 and psychosocial intervention.5 Alzheimer disease (AD), the most common cause of dementia world- wide, is a complex disorder that warrants a multi- dimensional approach with regular monitoring of the patient for increasing cognitive, functional, and behavioral challenges. Management consists of both pharmacologic and nonpharmacologic inter- ventions as well as referrals to social service agen- cies and support resources, such as the Alzheimer’s Association (www.alz.org). The family physician This article was externally peer reviewed. Submitted 5 August 2010; revised 14 October 2011; ac- cepted 14 November 2011. From the Division of Neurology, Nova Southeastern University, Fort Lauderdale, FL (CHS); Premiere Research Institute, Palm Beach Neurology, West Palm Beach, FL (CHS); and Center of Excellence for Brain Aging, New York University Langone School of Medicine, New York, NY (JEG). Funding: Administrative, editorial, and technical assis- tance was funded by Novartis Pharmaceuticals Corporation. Conflict of interest: none declared. Disclosure: CHS has served as a consultant to Novartis Pharmaceuticals Corporation; has served as a speaker for Novartis Pharmaceuticals Corporation, Forest Pharmaceu- ticals, and Accera; and has received honoraria from both companies. JEG is a paid consultant for Pfizer, Eisai, No- vartis Pharmaceuticals Corporation, Ortho-McNeil, and Forest. Neither author received financial support for this manuscript. Corresponding author: Carl H. Sadowsky, MD, Medical Director, Premiere Research Institute, Palm Beach Neurol- ogy, 4631 Congress Ave Ste 200, West Palm Beach, FL 33407 (E-mail: CHsadow@aol.com). 350 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ plays a key role in linking the family to community resources and other health care and social service providers who will help implement the overall care plan.6,7 Physicians also play a key role in coordi- nating the invaluable support network of nurse practitioners, physician assistants, social workers, and medical assistants. Moreover, the family phy- sician can assist in maintaining the physical health of patients with dementia, for example, assisting with the evaluation and treatment of visual and hearing defects, which are more common with ag- ing. Such assistance can help directly and indirectly in the management of dementia. For the purpose of this review, an electronic search of English-language articles (without time limits) was per- formed using PubMed and MEDLINE. The primary research parameters were Alzheimer’s disease, diagnosis, therapy, treatment, and therapeutic. Original research ar- ticles, reviews, and other articles of interest were re- viewed, and the most important information was iden- tified. This review provides a summary of these findings as well as practical advice for the busy clinician. Managing Cognitive, Memory, and Functioning Problems Goals of Therapy and Likely Outcomes The management of a patient with AD is a complex and evolving task because the natural history of AD is one of progressive decline; patients’ cognitive, physi- cal, and social functions gradually deteriorate.8 One of the key aspects of optimal management of demen- tia is realistic expectations for therapeutic outcomes, including treatment effects and potential outcomes; it is, therefore, imperative that the family physician is aware of these issues and discusses them with both the patient and caregiver.9 To be effective, interventions for patients with dementia ideally will improve func- tional status to a level that is detectable by caregivers or health care providers. In clinical trials, the Alzheimer’s Disease Assess- ment Scale, Cognitive Subscale (ADAS-Cog), a 20- minute, 11-item, 70-point scale that tests memory, language, orientation, and praxis, is often used to determine rate of cognitive decline. The total score ranges from 0 to 70, with a high score indicating greater impairment.10,11 Because of the progressive nature of AD, there may be brief plateaus during the illness; however, the decline is fairly consistent, tend- ing to increase or accelerate as patients enter the moderate stage.12 Therefore, any “improvement” from an intervention for dementia must take this into account. As such, “improvement” can be defined as a reduction in rate of decline.8 For example, patients with mild dementia experience an average rate of decline of �5 ADAS-Cog points,8 and slowing this decline by 2 to 3 ADAS-Cog points over a year could mean a delay of up to 7 months in disease progres- sion. In contrast, patients with moderate dementia (ADAS-Cog score �15 but �55) experience an aver- age decline in cognition of 7 to 11 ADAS-Cog points (2– 4 Mini Mental State Examination [MMSE] points) annually.13 Therefore, for people with mod- erate dementia, slowing decline by 2 to 3 ADAS-Cog points per year could mean a delay of 2 to 5 months in disease progression. In general, cholinesterase in- hibitors (ChEIs) do not delay ultimate progression of AD by more than 6 to 7 months. Nonpharmacologic Interventions An increasing number of nonpharmacologic therapies are now available for people with dementia, including behavioral therapy, reality orientation, art therapy, mu- sic therapy, complementary therapy, aromatherapy and bright-light therapy, as well as cognitive behavioral ther- apies. There are several areas of overlap between these therapies and each approach is rarely used in isolation14; it is therefore useful for clinicians to be familiar with several of these approaches to enable a combination of treatments to be tailored to individual requirements.15 Therapy is now directed toward person-centered forms of care and greater attempts are made to understand the individual’s experience of dementia and to employ strat- egies to improve the person’s quality of life (QoL).15 Individualized nonpharmacologic interventions include self-affirming exercises, such as reminiscence therapy, and structured socialization, such as pet therapy and viewing family videotapes.16 The efficacy of these inter- ventions has been demonstrated in both small and larger studies.17 Dietary Supplements Several nutrient deficiencies are known to be risk factors for AD. Evidence suggests that consumption of fish with high fat content and marine omega-3 polyunsaturated fatty acid decreases the risk of cog- nitive impairment and dementia.18 It is, therefore, not unusual in clinical practice to encounter patients and caregivers inquiring about dietary recommendations for lowering the risk of dementia. However, to date, there are no clinical trials to support a recommenda- tion of dietary and supplemental omega-3 polyunsat- doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 351 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ urated fatty acid for the sole purpose of preventing cognitive impairment or dementia.18 Nevertheless, it is not unwarranted to encourage adequate consump- tion of fatty fish as part of general dietary recommen- dations that may also confer benefits of reducing the risk of stroke and heart disease.19 There has been recent attention regarding the health benefits of curcumin (found in the commonly used Asian spice, turmeric) in AD. In animal studies, low-dose curcumin effectively disaggregates �-amy- loid and prevents fibril and oligomer formation, sup- porting the rationale for curcumin use in clinical trials preventing or treating AD.20 Indeed, a phase II clin- ical trial with patients with moderate to severe AD is ongoing, designed to determine whether curcumin can slow cognitive deterioration.21 Finally, recent studies from both the United States and Europe have suggested that vitamin D deficiency may be associated with increased odds of cognitive impairment in older persons.22,23 Indeed, results from a study in the United States in which cognitive impairment was assessed using measures of immediate and delayed verbal memory, orienta- tion, and attention reported a link between vitamin D deficiency and cognitive impairment in 3325 adults aged �65 years.22 The multivariate adjusted odds ratios (95% confidence interval) of cognitive impairment in patients who were vitamin D insuf- ficient (�50 � 75 nmol/L), deficient (�25 � 50 nmol/L), and severely deficient (�25 nmol/L) compared with those sufficient (�75 nmol/L) were 0.9 (0.6 –1.3), 1.4 (1.0 –2.1), and 3.9 (1.5–10.4), re- spectively (P for linear trend � .02), suggesting that vitamin D deficiency is associated with increased odds of cognitive impairment among the elderly population. Similar findings also have been re- ported in a European study.23 Although further exploration of a possible causal relationship be- tween vitamin D deficiency and cognitive impair- ment is warranted, these findings raise important new possibilities for treatment and prevention of cognitive decline in patients with AD. Medical Foods Medical foods as a class of intervention alternatives are not well known to most clinicians but are a growing area. Medical foods are a special category of US Food and Drug Administration (FDA)–regu- lated agents intended to provide specific nutritional requirements for patients with certain diseases; they can, therefore, provide an additional supple- ment in a comprehensive therapeutic regimen for patients with AD. Products being marketed cur- rently or developed in the United States for the management of dementia include Caprylic triglyc- eride (Axona, Accera, Inc., Broomfield, CO) and Souvenaid (Nutricia Advanced Medical Nutrition, Schiphol, The Netherlands). Axona has been developed for the clinical dietary management of the metabolic processes associated with mild to moderate AD. It is a formulation of caprylic triglyceride, a medium-chain triglyceride that is metabolized to ketone bodies, predomi- nantly �-hydroxybutyrate, a common metabolic sub- strate that is produced normally by the body for neurons in starvation states where glucose is less avail- able.24 A double-blind crossover study conducted in patients with AD or mild cognitive impairment dem- onstrated that Axona therapy was associated with sig- nificant improvements in ADAS-Cog; however, the effect was seen only in patients who were not carriers of apolipoprotein E ε4.24 Similar results were re- ported in a 90-day, randomized, placebo-controlled study in patients with mild to moderate AD.25 Sig- nificant gastrointestinal side effects have been associ- ated with Axona, and slow titration of the product is being recommended. Souvenaid (food) combines omega-3 fatty acids, choline, uridine monophosphate, and a mixture of antioxidants and B vitamins.26 In a randomized, controlled trial involving more than 200 patients with mild AD, Souvenaid was well tolerated and improved memory compared with placebo.27 Pharmacologic Interventions There are currently no means of reversing the pathologic processes of AD. Currently available medications do not halt the underlying degenera- tive process but can slow disease progression and therefore delay symptomatic decline.28 The specific goals of therapy are to preserve cognitive and func- tional ability, minimize behavioral disturbances, and slow disease progression with maintenance of patients’ and caregivers’ QoL.29 Nevertheless, re- alistic expectations of treatment outcomes are needed because the impact for most patients is likely to be modest and temporary, with not every patient responding to treatment. The main benefit of pharmacotherapy is an attenuation of decline over time rather than an improvement in cognitive or behavioral symptoms. It is important to discuss this point with patients and their families, who may 352 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ expect improvement rather than relative stability.30 Failure to do so often will result in patient and family dissatisfaction with prescribed therapies and the risk of discontinuation. Beneficial response to a ChEI (ie, delayed deterioration of cognitive or be- havioral problems) can be determined from the physician’s global assessment of the patient, the primary caregiver’s report, a neuropsychologic as- sessment or mental status questionnaire, or evi- dence of behavioral or functional changes.7 Four drugs are commonly used for treating AD: 3 ChEIs approved for mild to moderate disease, one of which also is approved for severe AD, and a glu- tamate N-methyl D-aspartate (NMDA) antagonist approved for moderate to severe disease (Table 1).30 Mild to Moderate Disease Since the introduction of the first ChEI in 1997, most clinicians would consider these agents to be first-line pharmacotherapy for mild to moderate AD.11 Four ChEIs are currently available: tacrine (Cognex, Shionogi, Inc., Atlanta, GA); donepezil (Aricept, Eisai Co, Ltd., Woodcliff Lake, NJ); riv- astigmine (Exelon, Novartis Pharmaceuticals Corp., East Hanover, NJ); and galantamine (Reminyl, Or- tho-McNeil Neurologics, Titusville, NJ). Tacrine is not commonly used because of a poor tolerability profile and low oral bioavailability, and it is, there- fore, excluded from this discussion.31 ChEIs raise acetylcholine levels in the brain by inhibiting ace- tylcholinesterase.6 Despite minor variations in their mode of action there is no evidence to suggest any difference in efficacy between the 3 commonly used ChEIs.11 Likewise, the tolerability profile is similar between the ChEIs for the oral formulations. How- ever, the 10-cm2 rivastigmine patch has shown ef- ficacy similar to oral rivastigmine formulations, but with approximately two-thirds fewer reports of nausea and vomiting, with adverse event (AE) rates similar to those of placebo32 (Table 1). AD often is accompanied and worsened by malnutrition, and weight loss is a frequent complication of AD, oc- curring in approximately 40% of patients at all stages.33 Donepezil, rivastigmine, and galantamine cause a broad spectrum of AEs, of which nausea, vomiting, diarrhea, and weight loss are the most common.34,35 There continues to be debate regarding the ex- tent of the benefits achieved with ChEIs. Although some assert that the most that can be achieved with ChEIs is symptom modification,11 others consider these agents to have disease-modifying effects.36 In one study, after discontinuation of therapy, riv- astigmine-treated patients showed less deteriora- tion in cognitive function compared with placebo- treated patients, suggesting an effect on disease progression.37 In another study, donepezil treat- ment slowed progression of hippocampal atrophy compared with untreated patients, suggesting a neuroprotective effect of donepezil in AD.38 How- ever, these early observations require confirmation, and, at present, the ChEIs generally are considered symptomatic medications. A systematic analysis of double-blind, placebo- controlled trials of ChEIs demonstrated treatment effects ranging from a 1.4- to 3.9-point improve- ment at 6 months and 1 year, in the midrange of the 70-point ADAS-Cog scale.11 In clinical trials, a change of 4 points is considered clinically signifi- cant for patients with mild to moderate demen- tia.39,40 As such, the symptomatic improvements observed are modest and of debatable clinical sig- nificance, despite being statistically significant.35 In a meta-analysis of 16 double-blind, placebo-con- trolled trials of ChEIs composed of almost 8000 patients, the numbers needed to treat for one ad- ditional patient to benefit were 7 for stabilization or better, 12 for minimal improvement or better, and 42 for marked improvement.41 Although the num- bers needed to treat seem favorable, uncertainty remains regarding the clinical relevance of these outcomes and the duration of the apparent benefit because the majority of trials reviewed were of less than 26 weeks’ duration. In addition to their effects on cognition, these agents also have demonstrated beneficial effects on measures of behavior, activities of daily living (ADLs), and global patient function. A recent meta-analysis that analyzed clinical results from 29 randomized, placebo-controlled trials of patients with mild to moderate AD found that ChEI ther- apy was associated with significant modest benefits in terms of neuropsychiatric and functional out- comes.42 Current guidelines acknowledge that pre- venting or delaying further loss of ADL function is an important goal of AD therapy43 and that the benefits of ChEIs may be diminished when treat- ment is delayed.28 Significant preservation of ADL function has been observed with donepezil, galan- tamine, and rivastigmine compared with placebo.29 ChEIs also have been shown to reduce AD care- giver burden: in patients with moderate to severe doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 353 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ Ta bl e 1. Ch ol in es te ra se In hi bi to rs an d M em an ti ne fo r th e Tr ea tm en t of Co gn it iv e D efi ci ts in Pa ti en ts w it h Al zh ei m er D is ea se (A D )6 ,5 9 D ru g A pp ro ve d In di ca ti on Su gg es te d D os ag e Si de E ff ec ts A dd it io na l N ot es /C au ti on C ho lin es te ra se in hi bi to rs D on ep ez il (A ri ce pt ) M ild to m od er at e A D O nc e da ily , be gi nn in g w it h 5 m g/ da y, w hi ch ca n be in cr ea se d to 10 m g/ da y (m ax im um do sa ge ) af te r 4 w ee ks A E s ar e m ild an d in cl ud e na us ea , vo m it in g, an d di ar rh ea G as tr oi nt es ti na l- re la te d A E s ca n be re du ce d if m ed ic at io n ta ke n w it h fo od So m e pa ti en ts ex hi bi t an in it ia l in cr ea se in ag it at io n, w hi ch su bs id es af te r fi rs t fe w w ee ks of th er ap y Se ve re A D R iv as ti gm in e (E xe lo n) M ild to m od er at e A D O ra l: T w ic e da ily , be gi nn in g w it h 1. 5 m g T ra ns de rm al pa tc h: O nc e da ily , 4. 6 or 9. 5 m g T he ta rg et do se is 9. 5 m g/ 24 hr pe r pa tc h (a 10 cm 2 pa tc h) an d re qu ir es a si m pl e on e- st ep do se ti tr at io n to th e th er ap eu ti c do se T he re is a hi gh er -d os e pa tc h (2 0 cm 2 ) av ai la bl e, de liv er in g 17 .4 m g/ 24 hr ; ho w ev er , it is cu rr en tl y an un ap pr ov ed tr ea tm en t in th e U ni te d St at es . L ac k of ap pr ov al w as ba se d on it ha vi ng si m ila r ef fi ca cy to th e 10 cm 2 pa tc h, bu t w it h a to le ra bi lit y pr ofi le co m pa ra bl e to th at of th e ca ps ul e fo rm ul at io n A E s in cl ud e na us ea , vo m it in g, di ar rh ea , w ei gh t lo ss , he ad ac he s, ab do m in al pa in , fa ti gu e, an xi et y, an d ag it at io n G as tr oi nt es ti na l- re la te d A E s ar e le ss pr om in en t w it h th e pa tc h: th e 9. 5 m g/ 24 hr pa tc h pr ov id es ef fi ca cy si m ila r to th at of th e hi gh es t do se of ca ps ul es , w it h 3 ti m es fe w er re po rt s of na us ea an d vo m it in g H ig he r do sa ge s ar e m or e ef fi ca ci ou s th an lo w er do sa ge s N o la bo ra to ry m on it or in g is re qu ir ed G al an ta m in e (R az ad yn e) M ild to m od er at e A D T w ic e da ily , be gi nn in g w it h 4 m g A ft er 4 w ee ks , do sa ge is in cr ea se d to 8 m g tw ic e da ily A n in cr ea se to 12 m g tw ic e da ily ca n be co ns id er ed on an in di vi du al ba si s af te r as se ss m en t of cl in ic al be ne fi t an d to le ra bi lit y A ls o av ai la bl e in an ex te nd ed -r el ea se fo rm ul at io n th at ca n be ta ke n on ce da ily M os t co m m on si de ef fe ct s ar e na us ea , vo m it in g, an d di ar rh ea G as tr oi nt es ti na l- re la te d A E s ca n be m in im iz ed by ti tr at in g th e do sa ge gr ad ua lly an d ta ki ng th e m ed ic at io n w it h m ea ls N M D A an ta go ni st M em an ti ne (N am en da ) M od er at e to se ve re A D T w ic e da ily , be gi nn in g w it h 5 m g, in cr ea si ng th e do se to 10 m g tw ic e da ily ov er 3 w ee ks A E s in cl ud e fa ti gu e, pa in , hy pe rt en si on , he ad ac he , co ns ti pa ti on , vo m it in g, ba ck pa in , so m no le nc e, di zz in es s M od er at e to se ve re A D m ay re sp on d be tt er w it h m em an ti ne /d on ep ez il co m bi na ti on ve rs us do ne pe zi l al on e A E , ad ve rs e ev en t; N M D A , N -m et hy l D -a sp ar ta te . 354 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ AD, donepezil treatment for 24 weeks significantly reduced caregiver time spent assisting patients with basic and instrumental ADLs (�52 minutes/day; P � .005).44 A small study has demonstrated that rivastigmine treatment reduces caregiver time spent assisting with ADLs (up to 690 hours over 2 years).45 Longer periods of treatment with ChEIs also decrease the risk for nursing home place- ment.46,47 A retrospective analysis of a large US medical claims database showed that over a 27- month follow-up period, more patients who were not treated with ChEIs were placed in nursing homes (11.0%) than were those who received ei- ther rivastigmine (3.7%) or donepezil (4.4%).47 These studies suggest that ChEIs enable patients to live longer in community settings with associated personal, social, and economic benefits.29 Memantine (Namenda, Forest Pharmaceuticals, St. Louis, MO) is sometimes used to treat patients with less severe disease, despite its use in early AD not being supported by the FDA. Although me- mantine has been reported to improve cognition, global status, and behavior in patients with mild to moderate AD,48 its mechanism of action would sug- gest that it does not have a place in early AD. Me- mantine is not a ChEI; it is a low- to moderate- affinity, noncompetitive (channel blocking), NMDA- receptor antagonist that seems to block pathologic neural toxicity associated with prolonged glutamate release.49 Blockade of NMDA receptors by meman- tine could confer disease-modifying activity in AD by inhibiting the “weak” NMDA receptor– dependent excitotoxicity that contributes to the neuronal loss underlying the progression of dementia.49 As such, memantine is not effective until weakened neurons become vulnerable to glutamate-induced excitotoxic- ity, and therefore it cannot substitute for ChEIs be- cause of its inability to enhance cholinergic neu- rotransmission required for memory and learning.49 Moderate to Severe Disease Memantine is approved for the treatment of mod- erate to severe AD on the basis of a study in which patients with moderate to severe AD who received 20 mg memantine monotherapy showed less decline in cognition and function while maintaining good tolerability after 6 months compared with those who received placebo.50 The ChEI donepezil also recently has been approved for use in severe AD. Recently, donepezil 23 mg/day has been ap- proved for the treatment of moderate to severe AD. Results from a 24-week, randomized, double-blind study reported that donepezil 23 mg/day was asso- ciated with greater benefits in cognition (as assessed by the Severe Impairment Battery) compared with donepezil 10 mg/day, although the between-treat- ment difference in the Clinician’s Interview-Based Impression of Change plus Caregiver Input Scale was not significant. The most commonly reported side effects with donepezil 23 mg/day were nausea vomiting, and diarrhea, which occurred at a higher incidence than with donepezil 10 mg/day.51 Combination therapy of a ChEI and memantine is rational from a pharmacologic perspective be- cause the agents have different mechanisms of ac- tion. In a randomized controlled trial, patients with moderate to severe AD who were already receiving donepezil derived significant benefit from the ad- dition of memantine in terms of cognition, ADLs, global outcome, and behavior.52 There are also economic benefits associated with the addition of memantine to donepezil treatment for patients with advanced AD. A recent study demonstrated improvement in clinical outcomes plus cost savings associated with the use of memantine.53 In a study by Tariot et al,52 the incidence of nausea was sub- stantially lower in patients receiving memantine add-on therapy compared with those receiving donepezil monotherapy. The safety and tolerability of combining rivastigmine capsule and memantine also has been studied in a 26-week, prospective, open-label study of patients with moderate AD.54 The combination was found to be both tolerable and safe, with a reduced incidence of gastrointesti- nal-related AEs compared with those documented in the US prescribing information for rivastigmine, suggesting that this beneficial effect of memantine may be applicable across ChEIs.54 Adjuncts to Pharmacotherapy for Improving Cognitive Function A recent study has demonstrated that ChEI-treated patients with early AD who received psychosocial support plus cognitive-motor intervention (CMI) had additional mood and cognitive benefits over those experienced by ChEI-treated patients who received psychosocial support alone. The CMI consisted of a 1-year structured program of 103 sessions, including reality orientation techniques, cognitive exercises, training of ADLs, and psy- chomotor exercises. Cognitive exercises were de- signed to stimulate memory, attention, language, doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 355 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ visuospatial abilities, calculation, and frontal/exec- utive functions. The ADL training was related to the particular cognitive function stimulated at each session (eg, money handling was trained after cal- culation exercises). The results showed that pa- tients in the CMI group maintained cognitive sta- tus at 6 months, whereas patients in the control group had significantly declined by that time. In addition, more patients who received CMI main- tained or improved their affective status after 1 year (CMI group, 75%; control group, 47%).55 Treatment Guidelines In 2006, a panel of leading experts published rec- ommendations for best practice in the treatment and management of AD. These recommendations were developed in an effort to address issues sur- rounding early diagnosis, treatment, and care man- agement of AD, as well as societal and managed- care implications.56 An algorithm was created to assist providers with the appropriate utilization of therapy and care management (Figure 1). This al- gorithm recommends initiating ChEI therapy in patients with mild AD and using combination ther- apy with a ChEI and memantine for patients who progress from mild to moderate AD. Alternatively, global guidelines recommend that patients who continue on the drug should be reviewed every 6 months by MMSE score and global, functional, and behavioral assessment.9 Treatment should be con- tinued only while the patient’s MMSE score re- mains �10 points and their global, functional, and behavioral condition indicates that the drug is hav- ing a worthwhile effect. In patients with moderate to severe AD (MMSE score �12), treatment with memantine can be considered, alone or in combi- nation with a ChEI.9 Managing Mood Disorders and Behavior Problems Traditionally, cognitive function has been the main focus of interest in treatment and research of peo- Figure 1. Treatment and management of Alzheimer disease. *Memory complaint may be raised by family or caregiver. All patients aged >75 years should be screened regardless of clinical presentation. †Cholinesterase inhibitors (ChEIs) are included for mild to moderate Alzheimer disease, excluding donepezil, which is indicated for mild, moderate, and severe Alzheimer disease. ‡Possible causes include medical comorbidities, the effects of other drugs, behavioral disturbances, or delirium. §Memantine is indicated for the treatment of moderate to severe Alzheimer disease. (This treatment algorithm is derived from recommendations published in Ref. 56. Reproduced with permission from RG Stefanacci. Reinforcing the value of combination therapy to treat moderate to severe alzheimer’s disease. Phys Week 2009;26(9). © 2009 Physician’s Weekly, LLC.) 356 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ ple with dementia. It is becoming increasingly rec- ognized, however, that noncognitive symptoms are those that are most disturbing to families and care- givers and may seriously impact not only the pa- tient’s well-being, but also the family’s, caregivers’, and providers’ approaches to managing the pa- tient.15 The most common symptoms are agitation, aggression, mood disorders/behavioral disturbance, apathy, depression, psychosis and hallucinations, with sexual disinhibition, elation/euphoria, appetite and eating disturbances, and abnormal vocaliza- tions occurring less frequently.15,57 These have been grouped together under the umbrella term behavioral and psychological symptoms of dementia by the International Psychogeriatric Association.58 As the disease progresses, these symptoms become predominant problems59 and impose an enormous toll, both emotionally and financially.17 They are also a common reason for institutionalization of people with dementia and they increase the burden and stress of caregivers.55,57,60 Nonpharmacologic Interventions Nonpharmacologic interventions are recom- mended as the most appropriate initial strategy for managing inappropriate behaviors in dementia be- cause (1) they address the psychosocial/environ- mental underlying reason for the behavior, and (2) they avoid the limitations of pharmacologic inter- ventions, namely, adverse side effects, drug– drug interactions, and limited efficacy.15,17,61 Increased involvement of caregivers often has a secondary benefit of providing overburdened caregivers with an opportunity to receive support, information, and skills. Furthermore, environmental factors (eg, con- fusing or noisy surroundings) or interpersonal factors (eg, arguing with the patient) are often the primary triggers of behavior problems. Attention to these fac- tors through nonpharmacologic approaches can be effective in alleviating or preventing behavioral prob- lems in individuals with dementia and should be con- sidered first.6,59,61 A recent consensus statement rec- ommended that all treatment approaches start with rigorous attempts to identify any reversible causes of these behaviors and alleviate these factors16 by modifying the physical and interpersonal environ- ments.62,63 Common triggers of agitation and ag- gression include pain, fecal impaction, medical ill- ness, boredom, loneliness, depression, and social and environmental stressors. Unfortunately, in practice, pharmacologic approaches involving neu- roleptic or other sedative medication are often used as the first-line treatment, despite the modest evi- dence of efficacy from clinical trials in which high placebo response rates frequently are seen.15,64 Patients with AD function best in an environ- ment that is safe, calm, and predictable, and their caregivers require ongoing support and education to develop realistic expectations throughout the course of the illness.30 Caregivers can be taught strategies to reduce agitation and anxiety in pa- tients with dementia.65 One such strategy utilizes the 3 Rs approach (repeat, reassure, and redirect), whereby the caregiver repeats an instruction or answer to a question as needed and redirects the patient to another activity to divert attention from a problematic situation. A predictable routine also is important and may avert certain behavioral problems. For example, scheduled toileting or prompted void- ing can reduce urinary incontinence.6 Training pro- grams for family caregivers of people with demen- tia, such as Savvy Caregiver, Staff Training in Assisted-Living Residences Caregivers, and Re- sources for Enhancing Alzheimer’s Caregiver Health, have resulted in decreased agitation among people with dementia who live at home and re- duced feelings of burden and depression among family caregivers.66 – 69 Nonpharmacologic interventions can be as simple as redirecting and refocusing the patient, increasing social interaction, initiating enjoyable activities, estab- lishing regular sleep habits, eliminating sources of conflict and frustration (eg, activities that the pa- tient can no longer undertake), and establishing rewards for successes, however small (Table 2).61 The principles of person-centered care, which aims to treat people as unique individuals with their own personality and preferences, are essential in the nonpharmacologic management of individuals with AD.17 For example, a person’s religious back- ground may influence his or her behavior. Patients of certain faiths may become agitated during inti- mate situations, such as bathing or dressing, when in the presence of caregivers who are of the oppo- site sex; a caregiver of the same sex may lead to improvement in behavior. The removal of any trig- gers of behavioral problems or the provision of comforting stimulation, such as the patient’s favor- ite music, also may be beneficial.70 The use of behavioral interventions in dementia is hindered by the fact that the patient’s cognitive functioning is declining progressively. As such, the doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 357 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ effects of interventions must be monitored contin- ually and adjustments made over time in response to new behaviors that may emerge.62 In patients with disruptive and hard-to-treat behavioral prob- lems, referral to a behavioral specialist such as a geriatric psychiatrist should be considered. Cognitive Behavioral Therapy Over the past 10 years there has been an increasing interest in applying therapeutic frameworks, such as cognitive behavioral therapy (CBT), cognitive stimulation therapy (CST), and interpersonal ther- apy to dementia. These therapies are designed to actively stimulate and engage people with demen- tia; group therapy, such as that used for CST, provides an optimal learning environment and the social benefits of a group and aims to create an environment in which people learn and strengthen their existing resources. The principles of person- centered care are essential when delivering CST for individuals with dementia; as such, group mem- bers often are assigned a role within the group according to their interests and abilities. During each themed session, there is a range of activities available, which allows the facilitator to adapt the level of difficulty of the activities depending on the group’s cognitive capabilities, interests, and gender mix; each individual can be provided with an activ- ity suitable for him or her personally. Sessions for CST include physical games, sound and word as- sociation, and faces/scenes. Individuals are asked to give their opinions rather than provide factual an- swers, and multisensory stimulation is used when possible. Teri and Gallagher-Thompson71 re- ported positive findings from a clinical trial of CBT with individuals with early AD, and individual and group CBT also has been used with some favorable results.15,72 A CBT perspective is appropriate for Table 2. Nonpharmacologic Interventions for Reducing Behavioral Disturbances in Alzheimer Disease (AD)6,95 Symptom Response Indecisiveness • Reduce choices Disorientation • Provide the patient with a predictable routine (eg, exercise, meals, and bedtime should be routine and punctual) • Avoid relocation; if necessary bring familiar items • Allow the patient to dress in his or her own clothing and keep possessions • Use calendars, clocks, labels, and newspapers for orientation to time • Use color-coded or graphic labels (eg, on closets, table service, drawers) as cues for orientation in the home environment Hallucinations • Do not be overly concerned if they are not distressing to the patient • Consider antipsychotic agents where necessary, but fully inform family and caregivers of the risks/benefits of these medications Delusions • Redirect and distract the patient • Consider using antipsychotic medications Repetitiveness • Answer decisively, then distract Lack of motivation • Ensure tasks are simple so that the patient can complete them; break up complex tasks into smaller steps • Before performing all procedures and activities, explain them to the patient in simple language Wandering (usually occurs later in the disease, ie, moderate to severe AD) • Register the patient in the Alzheimer’s Association Safe Return Program • Secure the environment with complex handles • Equip doors and gates with safety locks • Inform neighbors Agitation • Use distraction and redirection of activities to divert the patient from problematic situations • Reduce excess stimulation and outings to crowded places (overexposure to environmental stimuli can lead to agitation and disorientation) • Use lighting to reduce confusion and restlessness at night • Avoid glare from windows and mirrors, noise from a television, and household clutter Accident-prone • Provide a safe environment (eg, no sharp-edged furniture, no slippery floors or throw rugs, no obtrusive electrical cords) • Install grab bars by the toilet and in the shower Ensure that comorbid conditions are optimally treated Consider using a day care program for patients with AD 358 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ people with dementia because many of the behav- ioral difficulties encountered emerge through one or more of the following cognitive features: cogni- tive misinterpretations, biases, distortions, errone- ous problem-solving strategies, and communica- tion difficulties. Put simply, many of the challenges posed by people with dementia are caused by their thinking style—the very thing that is addressed in CBT. CBT, therefore, offers a framework within which to understand the individual’s distressing ex- periences, and this understanding allows the clini- cian to target interventions more appropriately.15 Pharmacologic Interventions Pharmacologic interventions are necessary when nonpharmacologic strategies fail to reduce behav- ioral symptoms sufficiently. Patients treated with ChEIs, memantine, or both may also experience behavioral benefits in terms of reduced severity of existing behavioral disturbances and fewer new be- havior symptoms29; usually agitation/aggression and irritability show responsiveness to ChEIs, me- mantine, or both, whereas depression, apathy, and anxiety do not. If behavioral disturbances persist despite the use of ChEIs, memantine, or both, a psychotropic agent may be necessary.6 In accordance with the principles of geriatric psychiatry “start low and go slow, but go,” the psychotropic agent should be initiated in a low dosage and then increased slowly until an adequate response occurs or side effects emerge. After behavioral disturbances have been controlled for 4 to 6 months, the dosage of the psychotropic agent can be reduced periodically to determine whether continued pharmacotherapy is required. The choice of psychopharmacologic agent is determined by specific target symptoms; some behaviors, such as wandering and pacing, are not amenable to drug therapy. Medications used to treat behavioral disturbances and mood disorders are summarized in Table 3.6 Atypical Antipsychotics Atypical antipsychotic drugs have been commonly used off-label in clinical practice for treatment of serious, dementia-associated agitation and aggres- sion, although they are not approved by the FDA for such use. In addition, these agents have a black- box warning of increased mortality among elderly patients with dementia-related psychosis. A meta- analysis assessed the evidence for increased mortal- ity from atypical antipsychotic drug treatment for people with dementia. Fifteen trials (9 unpub- lished), generally 10 to 12 weeks in duration and including 16 contrasts of atypical antipsychotic drugs with placebo, met criteria for inclusion (arip- iprazole [n � 3], olanzapine [n � 5], quetiapine [n � 3], risperidone [n � 5]; one trial was counted both as a risperidone trial and an olanzapine trial). A total of 3353 patients were randomized to study drug and 1757 were randomized to placebo. Results demonstrated that atypical antipsychotics may be associated with a 50% increased risk of death from all causes, which is similar to older antipsychotics, but there was no obvious difference in risk between the 4 agents.73 In general, drugs may be used only when nonpharmacologic approaches have failed to control serious behavioral disruption adequately within 5 to 7 days.16 Members of a recent consen- sus conference, who are experts in the field of geriatric mental health, reviewed the available evi- dence regarding the safety and efficacy of antipsy- chotic drugs. They concluded that problems in clinical trial design may have contributed to the negative results reported and suggested that future studies be required to address the benefit–risk bal- ance in this patient population.16 Nevertheless, the well-known incidence of side effects, such as seda- tion, falls, extrapyramidal signs, potential reduction in well-being and QoL,14 and even possible accel- eration of cognitive decline,15,74,75 mean that the risk– benefit ratio must be considered carefully when prescribing these drugs to a generally frail population. If antipsychotics are indicated, then it is recommended that they are used at the lowest effective dose, with dosage reduced or treatment discontinuation considered on a regular basis.59 Agitation Agitation and psychosis are distressing and are likely to overwhelm the caregiver’s ability to cope. If behavioral and nonpharmacologic interventions are inadequate, mild agitation can be managed with low doses of medications, such as trazodone, car- bamazepine, and valproate.59 Tricyclic antidepres- sants and benzodiazepines generally are avoided in this population.59 In patients with severe agitation and aggression, a recent consensus conference con- cluded that there is a need for an FDA-approved indication for treating dementia-related symptoms of severe and persistent or recurrent agitation and doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 359 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ Table 3. Pharmacologic Treatment of Behavior and Mood Disorders Antipsychotic drugs Atypical antipsychotic agents Recommended uses: control of problematic delusions, hallucinations, severe psychomotor agitation, and combativeness General cautions: diminished risk of developing extrapyramidal symptoms and tardive dyskinesia compared with typical antipsychotic agents Warning: atypical antipsychotic agents can cause an increased risk of cerebrovascular events (including stroke) in elderly patients with dementia-related psychosis Risperidone (Risperdal) Initial dosage: 0.25 mg/day at bedtime; maximum dosage: 2–3 mg/day, usually twice daily in divided doses Comments: current research supports use of low dosages; extrapyramidal symptoms may occur at 2 mg/day Olanzapine (Zyprexa) Initial dosage: 2.5 mg/day at bedtime; maximum dosage: 10 mg/day, usually twice daily in divided doses Comments: generally well tolerated Quetiapine (Seroquel) Initial dosage: 12.5 mg twice daily; maximum dosage: 200 mg twice daily Comments: more sedating; beware of transient orthostasis Typical antipsychotic agents Recommended uses: control of problematic delusions, hallucinations, severe psychomotor agitation, and combativeness; second-line therapy for patients who cannot tolerate or who do not respond to atypical antipsychotic agents General cautions: current research suggests that these drugs be avoided if possible because they are associated with significant, often severe side effects involving the cholinergic, cardiovascular, and extrapyramidal systems; there is also an inherent risk of irreversible tardive dyskinesia, which can develop in 50% of elderly patients after continuous use of typical antipsychotic agents for 2 years Warning: typical antipsychotic agents can cause an increased risk of cerebrovascular events (including stroke) in elderly patients with dementia-related psychosis Haloperidol (Haldol), fluphenazine (Prolixin), thiothixene (Navane) Dosage: varies by agent Comments: anticipated extrapyramidal symptoms; if these symptoms occur, decrease dosage or switch to another agent; avoid use of benztropine (Cogentin) or trihexyphenidyl (Artane) Trifluoperazine (Stelazine), molindone (Moban), perfenazine (Trilafon), loxapine (Loxitane) Dosage: varies by agent Comments: agents with “in-between” side- effect profile Mood-stabilizing (anti-agitation) drugs Recommended uses: control of problematic delusions, hallucinations, severe psychomotor agitation, and combativeness; useful alternatives to antipsychotic agents for control of severe agitated, repetitive, and combative behaviors General cautions: see comments about specific agents Trazodone (Desyrel) Initial dosage: 25 mg/day; maximum dosage: 200 to 400 mg/day in divided doses Comments: use with caution in patients with premature ventricular contractions Carbamazepine (Tegretol) Initial dosage: 100 mg twice daily; titrate to therapeutic blood level (4–8 �g/mL) Comments: monitor complete blood cell count and liver enzyme levels regularly; carbamazepine has problematic side effects Divalproex sodium (Depakote) Initial dosage: 125 mg twice daily; titrate to therapeutic blood level (40–90 �g/mL) Comments: generally better tolerated than other mood stabilizers; monitor liver enzyme levels; monitor platelets, prothrombin time, and partial thromboplastin time as indicated Anxiolytic drugs Benzodiazepines Recommended uses: management of insomnia, anxiety and agitation General cautions: regular use can lead to tolerance, addiction, depression and cognitive impairment; paradoxic agitations occurs in about 10% of patients treated with benzodiazepines; infrequent, low doses of agents with a short half-life are least problematic Lorazepam (Ativan), oxazepam (Serax), temazepam (Restoril), zolpidem (Ambien), triazolam (Halcion) Dosage: varies by agent See general cautions Continued 360 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ Table 3. Continued Nonbenzodiazepines Buspirone (BuSpar) Initial dosage: 5 mg twice daily; maximum dosage: 20 mg 3 times daily Comments: useful only in patients with mild to moderate agitation; may take 2 to 4 weeks to become effective Antidepressant drugs Recommended uses: see comments on specific agents General cautions: selection of an antidepressant is usually based on previous treatment response, tolerance and the advantage of potential side effects (eg, sedation vs activation); a full therapeutic trial requires 4–8 weeks; as a rule, dosage is increased using increments of initial dose every 5–7 days until therapeutic benefits or significant side effects become apparent; after 9 months, dosage reduction is used to reassess the need to medicate; discontinuing an antidepressant over 10–14 days limits withdrawal symptoms. Note: patients with depression and psychosis require concomitant antipsychotic medications. Tricyclic antidepressant agents Desipramine (Norpramin) Initial dosage: 10–25 mg in the morning; maximum dosage: 150 mg in the morning Comments: tends to be activating (eg, reduces apathy); lower risk for cardiotoxic, hypotensive and anticholinergic effects; may cause tachycardia; blood levels may be helpful Nortriptyline (Pamelor) Initial dosage: 10 mg at bedtime; anticipated dosage range: 10–40 mg/ day (given twice daily) Comments: tolerance profile is similar to that of desipramine, but nortriptyline tends to be more sedating; may be useful in patients with agitated depression and insomnia; therapeutic blood level “window” of 50–150 ng/mL (190–570 nmol/L) Heterocyclic and noncyclic antidepressant agents Nefazodone (Serzone) Initial dosage: 50 mg twice daily; maximum dosage: 150–300 mg twice daily Comments: effective, especially in patients with associated anxiety; reduced dose of coadministered alprazolam (Xanax) or triazolam by 50%; monitor for hepatotoxicity Buproprion (Wellbutrin) Initial dosage: 37.5 mg every morning, then increase by 37.5 mg every 3 days; maximum dosage: 150 mg twice daily Comments: activating; possible rapid improvement of energy level; should not be used in agitated patients and those with seizure disorders; to minimize risk of insomnia, give second dose before 3pm Mirtazapine (Remeron) Initial dosage: 7.5 mg at bedtime; maximum dosage: 30 mg at bedtime Comments: potent and well tolerated; promotes sleep, appetite, and weight gain SSRIs Recommended uses: may prolong half-life of other drugs by inhibiting various cytochrome P450 isoenzymes General cautions: typical side effects include sweating, tremors, nervousness, insomnia or somnolence, dizziness, and various gastrointestinal and sexual disturbances Fluoxetine (Prozac) Initial dosage: 10 mg every other morning; maximum dosage: 20 mg every morning Comments: activating, very long half-life; side effects may not manifest for a few weeks Paroxetine (Paxil) Initial dosage: 10 mg/day; maximum dosage: 40 mg/day (morning or evening) Comments: less activating but more anticholinergic than other SSRIs Sertraline (Zoloft) Initial dosage: 25–50 mg/day; maximum dosage: 200 mg/day (morning or evening) Comments: well tolerated; compared with other SSRIs, sertraline has less effect on metabolism of other medications Citalopram (Celexa) Initial dosage: 10 mg/day; maximum dosage: 40 mg/day Comments: well tolerated; some patients experience nausea and sleep disturbances Fluvoxamine (Luvox) Initial dosage: 50 mg twice daily; maximum dosage: 150 mg twice daily Comments: exercise caution when using fluvoxamine with alprazolam or triazolam Reproduced with permission from Cummings JL, et al. Am Fam Physician 2002; 65:2525 to 2534. © 2002 American Academy of Family Physicians.6 SSRI, selective serotonin reuptake inhibitor. doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 361 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ aggression, even in the absence of psychosis.16 Se- lective serotonin reuptake inhibitors seem to have efficacy for treatment of agitation in patients with AD. Studies have demonstrated benefits for agita- tion with citalopram compared with placebo76 and similar efficacy compared with risperidone.77 Apathy Apathy as a distinct psychiatric syndrome is an evolving concept but generally has been defined as poor initiation, impaired persistence, indifference, reduced emotional response, and low social en- gagement.78 Although once believed to be just a symptom of depression, apathy is characterized pri- marily as a loss of motivation and reduced emo- tional reactivity, as opposed to depression, which is a mood disturbance.79 Based on a limited but in- creasing body of evidence, methylphenidate seems to have some efficacy for the treatment of apathy in older adults with AD.80 Depression Depression is common in older adults, including those with AD, and often is undiagnosed and un- treated. The efficacy of antidepressants in patients with AD who also suffer depression has been dem- onstrated in clinical trial; the most useful medica- tions are those with minimal anticholinergic side effects. Selective serotonin reuptake inhibitors, such as citalopram (Celexa, Forest Laboratories, Inc.) and sertraline (Zoloft, Pfizer, New York, NY), seem to be effective and have fewer side effects compared with other antidepressants; as such, they are considered the agents of choice for the treat- ment of depression in patients with dementia, al- though direct head-to-head studies have yet to be undertaken.7,81 The Needs of the Caregiver Caregivers can become exhausted and frustrated; suffer depression, anxiety, and health problems; and be at increased risk of death.2,9 Ideally, care- givers would receive assistance in caregiving, peri- odic assessment of their own health and welfare, support from family and friends, and respite care. One study has reported that the most consistently effective method of caregiver treatment interven- tions is to teach caregivers how to change or modify their interaction with the patient.30 Mittelman and colleagues82 have demonstrated the effectiveness of long-term behavioral interven- tions for caregivers. Caregivers of patients with AD often suffer from depression, and optimizing long- term social support (individual and family counsel- ing, the continuous availability of ad hoc counsel- ing, and support group participation) can have a significant impact on depression in caregivers.82 The same authors subsequently demonstrated that a program of counseling and support substantially increased the time spousal caregivers were able to care for AD patients at home. Patients whose spouses received the intervention experienced a 28% reduction in the rate of nursing home place- ment compared with usual care controls, with a difference in time to placement of 557 days. Im- provements in caregivers’ satisfaction with social support, response to patient behavior problems, and symptoms of depression collectively accounted for 61% of the intervention’s beneficial impact on placement.83,84 Furthermore, these benefits were greatest in patients who had only mild dementia, when nursing home placement is generally least appropriate.83,84 In the event that insufficient resources are avail- able to provide for and protect both patient and caregiver, nursing home placement needs to be considered.2 The progressive nature of dementia also must be emphasized, such that in the event of nursing home placement the caregiver does not consider it to be a failure on their part.2 Discussing the benefits and disadvantages of institutional care with caregivers is often challenging. Although con- sideration of the patient’s previously expressed wishes is essential, caregivers often feel constrained by comments made years earlier and believe that the patient would not accept long-term care. It can be helpful to remind caregivers that earlier com- ments were made without a full appreciation of the current circumstances and that expectations almost always change with chronic illnesses.70 Future Therapies ChEIs and memantine are symptomatic therapies that help maintain neuronal function but do not have a significant impact on the underlying disease process. Their benefits are mild, and treatments that modify the disease course are urgently needed.30,39 AD is the destruction of brain that cannot be regenerated, and any effective treatment 362 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ needs to start before much brain is destroyed. There recently has been intense research interest in characterizing the earliest stages of AD that pre- cede the crossing of the dementia threshold, de- fined by functional disability.85 Such preclinical disease detection may allow earlier therapeutic in- tervention before critical numbers of neurons are lost. AD currently is thought to be a complex, mul- tifactorial syndrome, unlikely to arise from a single causal factor; instead, a number of related biologic alterations are thought to contribute to its patho- genesis. In light of this, drug combinations that can act at different levels of the neurotoxic cascade offer new avenues toward curing AD and other neuro- degenerative diseases.86 Effective treatment will re- quire attacking multiple targets.87 At present, key therapeutic approaches include reduction of brain amyloid levels,30,88,89 prevention of � hyperphospho- rylation into intraneuronal neurofibrillary tan- gles,30,89 and stimulation of muscarinic acetylcholine receptors,90,91 although novel therapies increasingly are targeted to preserving energy metabolism in the mitochondria.92–94 Conclusion Family physicians play a crucial role in the care of patients with AD in terms of early detection, timely intervention, and effective ongoing management. Optimal management involves a multidimensional approach to treatment that includes the physician, geriatric care managers, social services, and the patient’s family. The treatment of AD consists of both pharmacologic and nonpharmacologic inter- ventions. Nonpharmacologic interventions are recom- mended as the most appropriate initial strategy for managing problematic behaviors. Patients with AD function best in an environment that is safe, calm, and predictable. Interventions for improving be- havior include reduction of environmental stressors and strategies to reduce the agitation and anxiety of the patient. These interventions can be as simple as redirecting and refocusing the patient, increasing social interaction, establishing regular sleep habits, eliminating sources of conflict and frustration, and establishing rewards for successes, however small. The role and needs of the caregiver are important, and the effectiveness of long-term behavioral inter- ventions for caregivers has been demonstrated. In the absence of means to reverse the patho- logic processes of AD, the primary objectives of pharmacologic interventions are to preserve cogni- tive and functional ability, minimize behavioral dis- turbances, and slow disease progression. At present, four drugs are widely used to treat AD: 3 ChEIs, which are first-line treatment for patients with mild to moderate AD, and an NMDA antagonist ap- proved for treatment of moderate to severe AD. The authors would like to thank Frances Gambling for her editorial assistance with the manuscript. Administrative, edito- rial, and technical assistance was funded by Novartis Pharma- ceuticals Corporation. References 1. Downs M, Turner S, Bryans M, et al. Effectiveness of educational interventions in improving detection and management of dementia in primary care: clus- ter randomised controlled study. BMJ 2006;332: 692– 6. 2. van Hout H, Vernooij-Dassen M, Bakker K, Blom M, Grol R. General practitioners on dementia: tasks, practices and obstacles. Patient Educ Couns 2000; 39:219 –25. 3. Knopman D, Donohue JA, Gutterman EM. Patterns of care in the early stages of Alzheimer’s disease: impediments to timely diagnosis. J Am Geriatr Soc 2000;48:300 – 4. 4. Bullock R. New drugs for Alzheimer’s disease and other dementias. Br J Psychiatry 2002;180:135–9. 5. Woods RT, Moniz-Cook E, Iliffe S, et al. Dementia: issues in early recognition and intervention in pri- mary care. J R Soc Med 2003;96:320 – 4. 6. Cummings JL, Frank JC, Cherry D, et al. Guidelines for managing Alzheimer’s disease: part II. Treat- ment. Am Fam Physician 2002;65:2525–34. 7. Cummings JL, Frank JC, Cherry D, et al. Guidelines for managing Alzheimer’s disease: part I. Assess- ment. Am Fam Physician 2002;65:2263–72. 8. Boustani M, Peterson B, Hanson L, Harris R, Lohr KN. Screening for dementia in primary care: a sum- mary of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med 2003;138:927–37. 9. Waldemar G, Dubois B, Emre M, et al. Recommenda- tions for the diagnosis and management of Alzheimer’s disease and other disorders associated with dementia: EFNS guideline. Eur J Neurol 2007;14:e1–26. 10. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984;141: 1356 – 64. 11. Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst Rev 2006;(1): CD005593. 12. National Institute of Clinical Excellence. Dementia: supporting people with dementia and their carers in doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 363 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ health and social care. 2006. November 2006, amended March 2011. Available from: http://www.nice.org.uk/ nicemedia/live/10998/30318/30318.pdf. Accessed 29 March 2012. 13. Salmon DP, Thal LJ, Butters N, Heindel WC. Lon- gitudinal evaluation of dementia of the Alzheimer type: a comparison of 3 standardized mental status examinations. Neurology 1990;40:1225–30. 14. Ballard CG, O’Brien J, James I. Dementia: manage- ment of behavioral and psychological symptoms. Ox- ford: Oxford University Press; 2001. 15. Douglas S, James I, Ballard C. Non-pharmacological interventions in dementia. Adv Psychiatric Treat- ment 2004;10:171–9. 16. Salzman C, Jeste DV, Meyer RE, et al. Elderly patients with dementia-related symptoms of severe agitation and aggression: consensus statement on treatment options, clinical trials methodology, and policy. J Clin Psychiatry 2008;69:889 –98. 17. Cohen-Mansfield J. Nonpharmacologic interven- tions for inappropriate behaviors in dementia: a re- view, summary, and critique. Am J Geriatr Psychia- try 2001;9:361– 81. 18. Lim WS, Gammack JK, Van Niekerk J, Dangour AD. Omega 3 fatty acid for the prevention of de- mentia. Cochrane Database Syst Rev 2006;(1): CD005379. 19. Friedland RP. Fish consumption and the risk of Alzheimer disease: is it time to make dietary recom- mendations? Arch Neurol 2003;60:923– 4. 20. Yang F, Lim GP, Begum AN, et al. Curcumin in- hibits formation of amyloid beta oligomers and fi- brils, binds plaques, and reduces amyloid in vivo. J Biol Chem 2005;280:5892–901. 21. Poncha F. Efficacy and safety of curcumin formulation in Alzheimer’s disease. ClinicalTrials.gov identifier: NCT01001637. 15 October 2009, updated 23 October 2009. Available from: http://clinicaltrials.gov/ct2/show/ NCT01001637?term�NCT01001637&rank�1. Ac- cessed 15 March 2012. 22. Llewellyn DJ, Lang IA, Langa KM, Melzer D. Vi- tamin D and cognitive impairment in the elderly U.S. population. J Gerontol A Biol Sci Med Sci 66:59 – 65. 23. Llewellyn DJ, Lang IA, Langa KM, et al. Vitamin D and risk of cognitive decline in elderly persons. Arch Intern Med 170:1135– 41. 24. Reger MA, Henderson ST, Hale C, et al. Effects of beta-hydroxybutyrate on cognition in memory-im- paired adults. Neurobiol Aging 2004;25:311– 4. 25. Accera. Axona description. Available from: http:// www.about-axona.com/wordpress/wp-content/ uploads/prescribinginformation.pdf. Accessed 29 March 2012. 26. Nutricia. The Science Behind Souvenaid®. Available from: http://souvenaid.nutricia.com/the-science-behind- souvenaid.html. Accessed 29 March 2012. 27. Scheltens P, Verhey FRJ, Olde Rikkert MGM, Kam- phuis PJGH, Wilkinson D, Kurz A. The efficacy of a medical food (Souvenaid) in Alzheimer’s disease: results from the first trial and design of future trials. Alzheimers Dement 2009;5(Suppl):258 –9. 28. Farlow MR, Cummings JL. Effective pharmacologic management of Alzheimer’s disease. Am J Med 2007; 120:388 –97. 29. Geldmacher DS. Treatment guidelines for Alzhei- mer’s disease: redefining perceptions in primary care. Prim Care Companion J Clin Psychiatry 2007; 9:113–21. 30. Salloway S, Correia S. Alzheimer disease: time to improve its diagnosis and treatment. Cleve Clin J Med 2009;76:49 –58. 31. Birks J, Grimley Evans J, Iakovidou V, Tsolaki M, Holt FE. Rivastigmine for Alzheimer’s disease. Co- chrane Database Syst Rev 2009;(2):CD001191. 32. Winblad B, Grossberg G, Frolich L, et al. IDEAL: a 6-month, double-blind, placebo-controlled study of the first skin patch for Alzheimer disease. Neurology 2007;69:S14 –22. 33. Guerin O, Andrieu S, Schneider SM, et al. Different modes of weight loss in Alzheimer disease: a pro- spective study of 395 patients. Am J Clin Nutr 2005; 82:435– 41. 34. Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt HP, van den Bussche H. Cholinesterase inhibitors for patients with Alzheimer’s disease: systematic re- view of randomised clinical trials. BMJ 2005;331: 321–7. 35. Qaseem A, Snow V, Cross JT Jr, et al. Current pharmacologic treatment of dementia: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med 2008;148:370 – 8. 36. Cummings JL. Searching for methods to detect, pre- vent, and treat Alzheimer’s disease. Am J Psychiatry 2005;162:645–7. 37. Farlow M, Potkin S, Koumaras B, Veach J, Mirski D. Analysis of outcome in retrieved dropout patients in a rivastigmine vs placebo, 26-week. Alzheimer dis- ease trial. Arch Neurol 2003;60:843– 8. 38. Hashimoto M, Kazui H, Matsumoto K, Nakano Y, Yasuda M, Mori E. Does donepezil treatment slow the progression of hippocampal atrophy in patients with Alzheimer’s disease? Am J Psychiatry 2005;162: 676 – 82. 39. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treat- ing dementia: evidence review for a clinical practice guideline. Ann Intern Med 2008;148:379 –97. 40. Doraiswamy PM, Kaiser L, Bieber F, Garman RL. The Alzheimer’s Disease Assessment Scale: evalua- tion of psychometric properties and patterns of cog- nitive decline in multicenter clinical trials of mild to moderate Alzheimer’s disease. Alzheimer Dis Assoc Disord 2001;15:174 – 83. 364 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ 41. Lanctot KL, Herrmann N, Yau KK, et al. Efficacy and safety of cholinesterase inhibitors in Alzheimer’s disease: a meta-analysis. CMAJ 2003;169:557– 64. 42. Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase inhibitors in the treatment of neu- ropsychiatric symptoms and functional impairment in Alzheimer disease: a meta-analysis. JAMA 2003; 289:210 – 6. 43. Doody RS, Stevens JC, Beck C, et al. Practice pa- rameter: management of dementia (an evidence- based review). Report of the Quality Standards Sub- committee of the American Academy of Neurology. Neurology 2001;56:1154 – 66. 44. Feldman H, Gauthier S, Hecker J, et al. Efficacy of donepezil on maintenance of activities of daily living in patients with moderate to severe Alzheimer’s dis- ease and the effect on caregiver burden. J Am Geriatr Soc 2003;51:737– 44. 45. Marin D, Amaya K, Casciano R, et al. Impact of rivastigmine on costs and on time spent in caregiving for families of patients with Alzheimer’s disease. Int Psychogeriatr 2003;15:385–98. 46. Lopez OL, Becker JT, Saxton J, Sweet RA, Klunk W, DeKosky ST. Alteration of a clinically meaning- ful outcome in the natural history of Alzheimer’s disease by cholinesterase inhibition. J Am Geriatr Soc 2005;53:83–7. 47. Beusterien KM, Thomas SK, Gause D, Kimel M, Arcona S, Mirski D. Impact of rivastigmine use on the risk of nursing home placement in a US sample. CNS Drugs 2004;18:1143– 8. 48. Peskind ER, Potkin SG, Pomara N, et al. Meman- tine treatment in mild to moderate Alzheimer dis- ease: a 24-week randomized, controlled trial. Am J Geriatr Psychiatry 2006;14:704 –15. 49. Rogawski MA, Wenk GL. The neuropharmacolog- ical basis for the use of memantine in the treatment of Alzheimer’s disease. CNS Drug Rev 2003;9:275– 308. 50. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. Memantine in moderate-to-severe Alz- heimer’s disease. N Engl J Med 2003;348:1333– 41. 51. Farlow MR, Salloway S, Tariot PN, et al. Effective- ness and tolerability of high-dose (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther;32:1234 –51. 52. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA 2004;291:317–24. 53. Weycker D, Taneja C, Edelsberg J, et al. Cost- effectiveness of memantine in moderate-to-severe Alzheimer’s disease patients receiving donepezil. Curr Med Res Opin 2007;23:1187–97. 54. Olin JT, Bhatnagar V, Reyes P, Koumaras B, Meng X, Brannan S. Safety and tolerability of rivastigmine capsule with memantine in patients with probable Alzheimer’s disease: a 26-week, open-label, prospec- tive trial (study ENA713B US32). Int J Geriatr Psy- chiatry 25:419 –26. 55. Olazaran J, Muniz R, Reisberg B, et al. Benefits of cognitive-motor intervention in MCI and mild to moderate Alzheimer disease. Neurology 2004;63: 2348 –53. 56. Fillit HM, Doody RS, Binaso K, et al. Recommen- dations for best practices in the treatment of Alzhei- mer’s disease in managed care. Am J Geriatr Phar- macother 2006;4(Suppl 1):S9 –S24; quiz S5–S8. 57. deSouza L, Sarazin M, Goetz C, Dubois B. Clinical investigations in primary care. Front Neurol Neuro- sci 2009;24:1–11. 58. Finkel SI, Costa e Silva J, Cohen G, Miller S, Sar- torius N. Behavioral and psychological signs and symptoms of dementia: a consensus statement on current knowledge and implications for research and treatment. Int Psychogeriatr 1996;3(8 Suppl):497– 500. 59. Christensen DD, Lin P. Practical treatment strate- gies for patients with Alzheimer’s disease. J Fam Pract 2007;56:S17–23. 60. Schultz R, Williamson GH. A 2-year longitudinal study of depression among Alzheimer’s caregivers. Psychol Aging 1991;6:569 –78. 61. Teri L, Logsdon RG, McCurry SM. Nonpharmaco- logic treatment of behavioral disturbance in demen- tia. Med Clin North Am 2002;86:641–56, viii. 62. Logsdon RG, McCurry SM, Teri L. Evidence-based psychological treatments for disruptive behaviors in individuals with dementia. Psychol Aging 2007;22: 28 –36. 63. Emerson E. Working with people with challenging behavior. Chichester: John Wiley and Sons; 1998. 64. Ballard C, O’Brien J. Treating behavioural and psy- chological signs in Alzheimer’s disease. BMJ 1999; 319:138 –9. 65. Haupt M, Karger A, Janner M. Improvement of agitation and anxiety in demented patients after psy- choeducative group intervention with their caregiv- ers. Int J Geriatr Psychiatry 2000;15:1125–9. 66. Hepburn KW, Lewis M, Sherman CW, Tornatore J. The savvy caregiver program: developing and test- ing a transportable dementia family caregiver train- ing program. Gerontologist 2003;43:908 –15. 67. Kovach CR, Taneli Y, Dohearty P, Schlidt AM, Cashin S, Silva-Smith AL. Effect of the BACE in- tervention on agitation of people with dementia. Gerontologist 2004;44:797– 806. 68. Teri L, McCurry SM, Logsdon R, Gibbons LE. Training community consultants to help family members improve dementia care: a randomized con- trolled trial. Gerontologist 2005;45:802–11. 69. Ostwald SK, Hepburn KW, Caron W, Burns T, Mantell R. Reducing caregiver burden: a random- ized psychoeducational intervention for caregivers of doi: 10.3122/jabfm.2012.03.100183 Guidelines for the Management of Dementia 365 o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/ persons with dementia. Gerontologist 1999;39:299 – 309. 70. Herrmann N, Gauthier S. Diagnosis and treatment of dementia: 6. Management of severe Alzheimer disease. CMAJ 2008;179:1279 – 87. 71. Teri L, Gallagher-Thompson D. Cognitive-behav- ioral interventions for treatment of depression in Alzheimer’s patients. Gerontologist 1991;31:413– 6. 72. Kipling T, Bailey M, Charlesworth G. The feasibil- ity of a cognitive behavioural therapy group for men with a mild/moderate cognitive impairment. Behav Cogn Psychother 1999;27:189 –93. 73. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for de- mentia: meta-analysis of randomized placebo-con- trolled trials. JAMA 2005;294:1934 – 43. 74. McShane R, Keene J, Gedling K, Fairburn C, Jacoby R, Hope T. Do neuroleptic drugs hasten cognitive decline in dementia? Prospective study with nec- ropsy follow up. BMJ 1997;314:266 –70. 75. Schneider LS, Tariot PN, Dagerman KS, et al. Ef- fectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med 2006;355: 1525–38. 76. Pollock BG, Mulsant BH, Rosen J, et al. Compari- son of citalopram, perphenazine, and placebo for the acute treatment of psychosis and behavioral distur- bances in hospitalized, demented patients. Am J Psy- chiatry 2002;159:460 –5. 77. Pollock BG, Mulsant BH, Rosen J, et al. A double- blind comparison of citalopram and risperidone for the treatment of behavioral and psychotic symptoms associated with dementia. Am J Geriatr Psychiatry 2007;15:942–52. 78. Landes AM, Sperry SD, Strauss ME, Geldmacher DS. Apathy in Alzheimer’s disease. J Am Geriatr Soc 2001;49:1700 –7. 79. Boyle PA, Malloy PF. Treating apathy in Alzhei- mer’s disease. Dement Geriatr Cogn Disord 2004; 17:91–9. 80. Dolder CR, Davis LN, McKinsey J. Use of psycho- stimulants in patients with dementia. Ann Pharma- cother 2010;44:1624 –32. 81. Lyketsos CG, Sheppard JM, Steele CD, et al. Ran- domized, placebo-controlled, double-blind clinical trial of sertraline in the treatment of depression com- plicating Alzheimer’s disease: initial results from the Depression in Alzheimer’s Disease study. Am J Psy- chiatry 2000;157:1686 – 0. 82. Mittelman MS, Ferris SH, Shulman E, et al. A com- prehensive support program: effect on depression in spouse-caregivers of AD patients. Gerontologist 1995;35:792– 802. 83. 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. 84. Mittelman MS, Haley WE, Clay OJ, Roth DL. Im- proving caregiver well-being delays nursing home placement of patients with Alzheimer disease. Neu- rology 2006;67:1592–9. 85. Dubois B, Picard G, Sarazin M. Early detection of Alzheimer’s disease: new diagnostic criteria. Dia- logues Clin Neurosci 2009;11:135–9. 86. Bolognesi ML, Rosini M, Andrisano V, et al. MTDL design strategy in the context of Alzheimer’s disease: from lipocrine to memoquin and beyond. Curr Pharm Des 2009;15:601–13. 87. Duara R, Barker W, Loewenstein D, Bain L. The basis for disease-modifying treatments for Alzhei- mer’s disease: the Sixth Annual Mild Cognitive Im- pairment Symposium. Alzheimers Dement 2009;5: 66 –74. 88. Bates KA, Verdile G, Li QX, et al. Clearance mech- anisms of Alzheimer’s amyloid-beta peptide: impli- cations for therapeutic design and diagnostic tests. Mol Psychiatry 2009;14:469 – 86. 89. Tarawneh R, Holtzman DM. Critical issues for suc- cessful immunotherapy in Alzheimer’s disease: de- velopment of biomarkers and methods for early de- tection and intervention. CNS Neurol Disord Drug Targets 2009;8:144 –59. 90. Caccamo A, Fisher A, LaFerla FM. M1 agonists as a potential disease-modifying therapy for Alzheimer’s disease. Curr Alzheimer Res 2009;6:112–7. 91. Conn PJ, Jones CK, Lindsley CW. Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends Pharmacol Sci 2009;30:148 –55. 92. Lermontova NN, Redkozubov AE, Shevtsova EF, Serkova TP, Kireeva EG, Bachurin SO. Dimebon and tacrine inhibit neurotoxic action of beta-amyloid in culture and block L-type Ca(2�) channels. Bull Exp Biol Med 2001;132:1079 – 83. 93. Bachurin SO, Shevtsova EP, Kireeva EG, Oxenkrug GF, Sablin SO. Mitochondria as a target for neuro- toxins and neuroprotective agents. Ann N Y Acad Sci. 2003;993:334 – 44. 94. Doody RS, Gavrilova SI, Sano M, et al. Effect of dimebon on cognition, activities of daily living, be- haviour, and global function in patients with mild- to-moderate Alzheimer’s disease: a randomised, dou- ble-blind, placebo-controlled study. Lancet 2008; 372:207–15. 95. Leifer BP. Alzheimer’s disease: seeing the signs early. J Am Acad Nurse Pract 2009;21:588 –95. 366 JABFM May–June 2012 Vol. 25 No. 3 http://www.jabfm.org o n 5 A p ril 2 0 2 1 b y g u e st. P ro te cte d b y co p yrig h t. h ttp ://w w w .ja b fm .o rg / J A m B o a rd F a m M e d : first p u b lish e d a s 1 0 .3 1 2 2 /ja b fm .2 0 1 2 .0 3 .1 0 0 1 8 3 o n 8 M a y 2 0 1 2 . D o w n lo a d e d fro m http://www.jabfm.org/