22
Symptomatic and Nonamyloid/Tau Based Pharmacologic Treatment for Alzheimer Disease Paul S. Aisen 1 , Jeffrey Cummings 2 , and Lon S. Schneider 3 1 University of California, San Diego, California 92093 2 Cleveland Clinic, Cleveland, Ohio 44195 3 University of Southern California, Los Angeles, California 90007 Correspondence: [email protected] In this work we consider marketed drugs for Alzheimer disease (AD) including acetylcholin- esterase inhibitors (AChE-Is) and antiglutamatergic treatment involving the N-methyl- D-aspartate (NMDA) receptor. We discuss medications and substances available for use as cognitive enhancers that are not approved for AD or cognitive impairment, and other neuro- transmitter-related therapies in development or currently being researched. We also review putative therapies that aim to slow disease progression by mechanisms not directly related to amyloid or tau. REGULATORY LANDSCAPE AND CLINICAL TRIALS N orth American and European Union regu- latory criteria for marketing approval of putative symptomatic and disease-modifying therapeutic agents for Alzheimer disease (AD) are based on a demonstration of efficacy supported by improvements compared to pla- cebo treatment on cognitive function, activities of daily living (ADL), and often evidence of overall clinical improvement or less overall decline, accompanied by adequate evidence of safety (Schneider 2008b). In practice this has led to rather standardized protocols by sponsors of experimental drugs. For example, mild to moderate AD is indexed by a Mini- Mental State Examination (MMSE) score of 10 – 26 and standardized outcomes including the Alzheimer’s Disease Assessment Scale— Cognitive Portion (ADAS-cog), the Alzheimer’s Disease Cooperative Study Activities of Daily Living (ADCS-ADL) scale, or the Disability Assessment for Dementia (DAD). A clinician’s global assessment (known as a Clinician Inter- view Based Impression of Change with caregiver input [CIBICþ] or ADCS Clinical Global Impression of Change [CGIC]) or the Clinical Dementia Rating (CDR), an interview-based overall dementia severity assessment, are the conventional outcomes measures. The trials are commonly 6–18 months in duration: 6 months for symptomatic and 12–18 months for disease-modifying trials. There is a current trend to include more mild patients with AD, operationalized as MMSE greater than 20, amnestic mild cognitive impairment (MCI), or MCI due to AD, prodromal or early AD Editors: Dennis J. Selkoe, Eckhard Mandelkow, and David M. Holtzman Additional Perspectives on The Biologyof Alzheimer Disease available at www.perspectivesinmedicine.org Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a006395 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 1 www.perspectivesinmedicine.org on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/ Downloaded from

Symptomatic and Nonamyloid Tau Based Pharmacologic

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Symptomatic and Nonamyloid Tau Based Pharmacologic

Symptomatic and Nonamyloid/Tau BasedPharmacologic Treatment for Alzheimer Disease

Paul S. Aisen1, Jeffrey Cummings2, and Lon S. Schneider3

1University of California, San Diego, California 920932Cleveland Clinic, Cleveland, Ohio 441953University of Southern California, Los Angeles, California 90007

Correspondence: [email protected]

In this work we consider marketed drugs for Alzheimer disease (AD) including acetylcholin-esterase inhibitors (AChE-Is) and antiglutamatergic treatment involving the N-methyl-D-aspartate (NMDA) receptor. We discuss medications and substances available for use ascognitive enhancers that are not approved for AD or cognitive impairment, and other neuro-transmitter-related therapies in development or currently being researched. We also reviewputative therapies that aim to slow disease progression by mechanisms not directly related toamyloid or tau.

REGULATORY LANDSCAPE AND CLINICALTRIALS

North American and European Union regu-latory criteria for marketing approval of

putative symptomatic and disease-modifyingtherapeutic agents for Alzheimer disease (AD)are based on a demonstration of efficacysupported by improvements compared to pla-cebo treatment on cognitive function, activitiesof daily living (ADL), and often evidence ofoverall clinical improvement or less overalldecline, accompanied by adequate evidenceof safety (Schneider 2008b). In practice thishas led to rather standardized protocols bysponsors of experimental drugs. For example,mild to moderate AD is indexed by a Mini-Mental State Examination (MMSE) score of10–26 and standardized outcomes including

the Alzheimer’s Disease Assessment Scale—Cognitive Portion (ADAS-cog), the Alzheimer’sDisease Cooperative Study Activities of DailyLiving (ADCS-ADL) scale, or the DisabilityAssessment for Dementia (DAD). A clinician’sglobal assessment (known as a Clinician Inter-view Based Impression of Change with caregiverinput [CIBICþ] or ADCS Clinical GlobalImpression of Change [CGIC]) or the ClinicalDementia Rating (CDR), an interview-basedoverall dementia severity assessment, are theconventional outcomes measures. The trialsare commonly 6–18 months in duration: 6months for symptomatic and 12–18 monthsfor disease-modifying trials. There is a currenttrend to include more mild patients with AD,operationalized as MMSE greater than 20,amnestic mild cognitive impairment (MCI),or MCI due to AD, prodromal or early AD

Editors: Dennis J. Selkoe, Eckhard Mandelkow, and David M. Holtzman

Additional Perspectives on The Biology of Alzheimer Disease available at www.perspectivesinmedicine.org

Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a006395

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

1

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 2: Symptomatic and Nonamyloid Tau Based Pharmacologic

(supported by a positive cerebrospinal fluid[CSF] or imaging biomarker) (Schneider2008b; Albert et al. 2011).

Five cholinesterase inhibitors, memantine,the Ginkgo biloba extract EGb 761, and cerebro-lysin have some level of marketing approvalfor the treatment of AD in the Western hemi-sphere, Europe, Australia/New Zealand, Japan,and many Asian countries. One antipsychotic,risperidone, is specifically approved in severalcountries, e.g., UK, Spain, and Canada for thetreatment of agitation, psychosis, or the behav-ioral and psychological symptoms of dementia(BPSD) occurring in AD.

Considering Mechanisms of Action

Demonstration of a mechanism, pharmacody-namic effect, or target engagement either in pre-clinical in vitro or in animal models, or inhumans does not establish the relevance of themechanism to the effect the drug might haveon clinical manifestations. Many agents, includ-ing various antioxidant and so-called anti-aging cocktails, are marketed based on thechemical properties of ingredients rather thanrandomized controlled trials of safety and effi-cacy. Such products may not have establishedsafety, and certainly have no evidence of efficacy.Moreover, most drug products have multipleactions. Thus, with the few exceptions of drugsand antibodies that clearly engage only one tar-get, characterizing a particular drug as having acertain mechanism of action may be mislead-ing; yet this is often done as a matter of conve-nience or for categorization.

ACETYLCHOLINESTERASE INHIBITORS

Acetylcholinesterase inhibitors (AChE-Is) arethe first class of agents specifically approved bythe US Food and Drug Administration (FDA)for the treatment of AD (Fig. 1). Tacrine (Cog-nex) was approved in 1993 followed by approvalof donepezil (Aricept) in 1996, rivastigmine (Exe-lon) in 2000, and galantamine (Reminyl, Raza-dyne) in 2001. Memantine (Namenda)—anNMDA receptor antagonist—approval followedin 2004 (Fig. 1). Tacrine had a short half-life

requiring administration every 4 hours andhad substantial associated hepatotoxicity, re-quiring frequent monitoring of liver enzymes.These limitations were not present with laterAChE-Is. Tacrine is rarely used, is no longeravailable in many countries, and will not be dis-cussed here.

All AChE-Is share the characteristic of in-hibiting acetylcholinesterase and each of theseagents has additional distinctive pharmaco-logic aspects. AChE-Is are thought to bind ace-tylcholinesterase in the synaptic cleft so thatacetylcholine released from the presynapticcholinergic terminal has an increased residencetime within the synapse and is more likely tointeract with the postsynaptic cholinergicreceptor. The enhanced postsynaptic activityrenders more normal the function of the cho-linergic system.

Donepezil is a selective AChE-I, rivastig-mine is a mixed acetylcholinesterase andbutyrylcholinesterase inhibitor, and galant-amine is described as having an allosteric nico-tinic modulating effect as well as being anAChE-I. The clinical consequences of the differ-ential pharmacology of the AChE-Is, if any, areunknown.

For purposes of drug development, patientswere identified for AChE-I clinical trials as hav-ing mild to moderate AD by requiring a MMSE(Folstein et al. 1975) score between ten and 26in most trials. All AChE-Is are approved fortreatment of mild to moderate AD. In theclinical development of donepezil, trials ofpatients with moderate to severe AD wereconducted and established efficacy of this agentin patients with MMSE scores of 0–15.Donepezil is approved for mild, moderate,and severe AD.

To meet the criteria for approval by the FDAas a treatment for AD, an agent must be shownin two well-conducted trials to be statisticallysignificantly superior to placebo on a test ofcognition (regarded as the central featureof AD) and a global scale or an assessment ofADL (Schneider 2008) The usual measure ofcognition in mild to moderate AD is the ADAS-Cog (Rosen et al. 1984). In patients with mod-erate to severe AD the cognitive measure most

P.S. Aisen et al.

2 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 3: Symptomatic and Nonamyloid Tau Based Pharmacologic

commonly used is the Severe ImpairmentBattery (SIB) (Schmitt et al. 1997). The globaloutcome used in most AChE-I trials is theCIBICþ (Schneider et al. 1997). ADL scalesused in AChE-I trials include the ADCS ADLscale (Galasko et al. 1997) or the DAD (Gelinaset al. 1999). Changes in behavior are commonlymeasured as secondary outcomes in AChE-Iclinical trials, and the most commonly usedinstrument is the Neuropsychiatric Inventory(NPI) (Cummings et al. 1994). Pharmacoeco-nomic data are collected in some trials usingthe Resource Utilization in Dementia (RUD)scale (Wimo and Winblad 2003). Most clinicaltrials of AChE-Is have been 6 months in dura-tion; some have been as short as 3 months andsome as long as 2 years (AD2000 CollaborativeGroup 2004).

The responses to treatment with differentAChE-Is have overlapping confidence intervals(CI), and no individual cholinesterase inhibitorhas been shown to be superior to others interms of efficacy. The mean response on theADAS-cog is approximately 2.0 points with CIof 1.5–2.5 points (Birks 2006). The responseon the CIBICþ is usually in the range of 1.9(CI 1.3–3.0) on a scale in which 4 representsno change. 3, 2 and 1 represent mild, moderateand marked improvement, and 5, 6, and 7 rep-resent mild, moderate, and marked worsening(Whitehead et al. 2004). A two-point (CI 0.5–

2.5) drug–placebo difference is common at trialconclusion on the ADCS ADL and a two-point(CI 0.5–4.0) drug–placebo difference on theNPI total score is common at trial conclusion.Approximately 25% of patients have a measur-able improvement on the ADAS-Cog comparedto 15% of patients on placebo. In addition tothe drug–placebo difference in improvement,AChE-Is produce a delay in decline that affectsas many as 80% of patients participating in aclinical trial (Geldmacher et al. 2006). Improve-ment on the ADAS-Cog is taken as an indicationof improvement in the core clinical featuresof AD, whereas a drug–placebo difference inCIBICþ or an ADL scale is accepted as a meas-ure of clinically meaningful improvement. Asnoted, most clinical trials of AChE-Is are 6months in duration and establish benefit for 6months of therapy; some 1-year trials (Winbladet al. 2001) or 2-year trials (AD2000 Collabora-tive Group 2004) have been conducted and con-tinued to show a drug–placebo difference atstudy conclusion. These observations supportthe long-term use of AChE-Is in the treatmentof AD.

AD dementia is preceded by a period ofcognitive impairment in which patients showdecline in episodic memory and other cogni-tive abilities, but the changes are not sufficient-ly severe to reach the criteria for dementia.This clinical syndrome has been called mild

TacrineCognexTM

(1993)

DonepezilAriceptTM

(1996)

GalantamineRazadyneTM

(2001)

RivastigmineExelonTM

(2000)

MemantineNamendaTM

(2004)

Figure 1. Timeline of approved treatments for Alzheimer disease.

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 3

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 4: Symptomatic and Nonamyloid Tau Based Pharmacologic

cognitive impairment (MCI) (Petersen et al.2001). Clinical trials of AChE-Is for the treat-ment of cognitive deficits of MCI have been uni-formly negative (Jelic et al. 2006). It is nowrecognized that MCI is an etiologically hetero-geneous state with only approximately 60–70% of patients having underlying AD (Jichaet al. 2006). The lack of response to AChE-Isin MCI may reflect the absence of an AD-typepathophysiology in a substantial number ofpatients included in the trial. In the ADCSMCI study, there was a drug–placebo benefitin favor of donepezil in MCI patients whowere apolipoprotein E4 carriers, a group likelyto have a high underlying rate of AD patho-physiology (Petersen et al. 2001). This suggeststhat prodromal AD may respond to treatmentby AChE-Is. However, many of the patientsenrolled in MCI trials could also have fulfilledcriteria for AD.

Gastrointestinal side effects are common inpatients placed on AChE-Is. Anorexia, nausea,vomiting, diarrhea, and weight loss may occurand should be monitored in patients treatedwith these agents. In addition, cholinergic influ-ences may slow heart rate and bradycardia is acontraindication to use of AChE-Is. Occasionalpatients have experienced changes in urinaryfunction. Muscle cramps have been reportedwith donepezil and abnormalities in dreamingalso have been reported with donepezil.

In addition to the use of AChE-Is in AD,rivastigmine has been approved by the FDAfor treatment of mild to moderate Parkinson’sdisease dementia (Emre et al. 2004). AChE-Ishave also been assessed in clinical trials of vascu-lar dementia (Black et al. 2003; Kavirajan andSchneider 2007), mixed AD and cerebrovascu-lar disease (Erkinjuntti et al. 2002), and demen-tia with Lewy bodies (McKeith et al. 2000).None of these indications has been approvedby the FDA and use of AChE-Is in these settingsis off label.

Donepezil has a half-life of 70 hours and isadministered once daily (Table 1). It is 96% pro-tein bound with 100% bioavailability; it ismetabolized by 2D6 and 3A4 cytochromeP450 enzymes. Treatment is begun with 5 mg/day and, if the patient shows no intolerance,

the dose is advanced to 10 mg/day usually after1 month. A 23 mg once-daily dose has recentlybeen approved for use in patients with moderateto severe AD (Okamura et al. 2008). Acetyl-cholinesterase positron emission tomography(PET) studies suggest that the 10 mg dose pro-duces an approximately 60% inhibition of cen-tral acetylcholinesterase (Okamura et al. 2008).Rivastigmine has a peripheral half-life of 1.5hours and a central half-life of 8 hours. The cap-sules are given twice daily and, when adminis-tered as the patch formulation, the patch isreplaced once daily. Rivastigmine is not metab-olized through the cytochrome P450 system; itis 40% protein bound and has 40% bioavailabil-ity. Rivastigmine is initiated in an oral dose of1.5 mg orally twice daily and, as tolerance isdetermined, advanced to 3 mg, 4.5 mg, and6 mg twice daily for a total target dose of12 mg/day. Titration is typically at 1-monthintervals and is determined by patient tolerancefor the agent. The transdermal patch formula-tion of rivastigmine is initiated at a patchstrength of 4.6 mg and advanced to 9.5 mg after1 month if no intolerance is observed. Galant-amine has a half-life of 7 hours and is giventwice daily, unless the extended release formula-tion is used and administered once daily. Gal-antamine is initiated at a dose of 4 mg twicedaily (8 mg/day in a single dose for theextended release formulation) and advancedto 16 and 24 mg/day at 1-month intervals. Gal-antamine is metabolized by the cytochromeP450 enzymes 2D6 and 3A4. It is 18% proteinand has 90% bioavailability.

There are conflicting data as to whetherAChE-Is have any disease-modifying propertieswith respect to AD. They affect disease course byimproving symptoms and delaying decline asdescribed above. Basic science observationssuggest that enhancement of cholinergic func-tion may reduce the generation of b-amyloidprotein (Kimura et al. 2005). Some imagingstudies have suggested less brain atrophy overtime in patients treated with AChE-Is (Krishnanet al. 2003). In addition, some long-termobservations suggest less decline in patientstreated with AChE-Is or combination therapyof AChE-Is with memantine compared to

P.S. Aisen et al.

4 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 5: Symptomatic and Nonamyloid Tau Based Pharmacologic

patients not receiving therapy with these agents(Lopez et al. 2009; Rountree et al. 2009). Otherclinical and imaging studies, however, suggestno disease-modifying benefit from treatmentwith AChE-Is (Jack et al. 2008; Schneider andSano 2009). Disease-modifying effects, if pre-sent, must be small in magnitude.

Patients intolerant to one AChE-I may beable to tolerate an alternate cholinesteraseinhibitor and the effort to keep patients on ther-apy should be made when intolerance occurs.Patients started on an oral medication mightbe switched to patch therapy or vice versa.Patients showing no efficacy in response totreatment with one cholinesterase inhibitor(uninterrupted continuing decline of cogni-tion) may also be switched to another. Anybenefit from switching in this circumstancehowever is not established.

Continuing benefit from use of AChE-Is hasbeen shown in trials lasting up to 2 years, amongpatients followed over that length of time(AD2000 Collaborative Group 2004). In addi-tion, patients with severe AD—not previouslytreated with AChE-Is—respond to treatmentwith AChE-Is, and donepezil is approved fortreatment in this advanced phase of the disease.These observations suggest that long-termtreatment with AChE-Is may continue to pro-vide benefit. When AChE-Is are discontinuedbecause the patient or the clinician believethat no further benefit is possible, patientsshould be observed for cognitive decline, lossof ADL, or emergent behavioral disturbances(Holmes et al. 2004). If adverse cognitive, func-tional, or behavioral changes occur soon afterdiscontinuation then physicians and patientsshould consider whether the patient had been

Table 1. Characteristics of cholinesterase inhibitors

Cholinesterase inhibitors

Characteristic Donepezil Rivastigmine Galantamine

Trade name Aricept (Aricet insome countries)

Exelon Razadyne and Razadyne-ER

Indications Mild to moderateand severe AD

Mild to moderate AD;Parkinson’s disease dementia

Mild to moderate AD

Half-life 70 hours 1.5 hours (brain half-life is8 hours)

7 hours

Administrationschedule

q.d. b.i.d. for capsules; q.d. for thepatch

b.i.d. for the non-ER form: q.d.for the ER form

Metabolism byhepatic CYPenzymes

2D6, 3A4 No 2D6, 3A4

Protein binding 96% 40% 18%Bioavailability 100% 40% 90%Time to peak

serum level3–4 hours 1 hour 1 hour (2.5 hours with food); 4.5

hours for ER formAbsorption delayed

by foodNo No Yes (1 hour to 2.5 hours)

Titration Begin with 5 mgand advance to10 mg after 1month

Oral form: 1.5 mg b.i.d. for 4weeks; 3 mg b.i.d. for 4 weeks;4.5 mg b.i.d. for 4 weeks;advance to 6 mg b.i.d. iftolerated

Patch form: begin the 5 cm2

patch for 1 month thenadvance to 10 cm2 patch

Non-ER form: begin 4 mg b.i.d.;advance to 8 mg b.i.d. after 1month and to 12 mg b.i.d.after 1 month

ER form: begin at 8 mg q.d.;advance after 1 month to16 mg DQ and after 1 monthto 24 mg q.d.

Tacrine (Cognex) is now rarely used because of associated liver enzyme elevations and is not included in this chart.

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 5

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 6: Symptomatic and Nonamyloid Tau Based Pharmacologic

benefiting from treatment and whether to reini-tiate medications at the starting doses.

MEMANTINE

Memantine was used in Germany for the treat-ment of Parkinson’s disease prior to its approvalin the USA and globally for treatment of AD. Akey 12-week clinical trial of memantine forpatients with dementia, mainly AD, residingin nursing homes was published in 1999 (Win-blad and Poritis 1999) followed by publicationof a monotherapy trial in 2000 (Reisberg et al.2003a) and of a donepezil add-on trial in 2004(Tariot et al. 2004). Memantine was approvedby the FDA in the USA in 2004 for treatmentof moderate to severe AD, meaning patientswith MMSE scores less than 15. Memantine isan NMDA receptor antagonist that replacespotassium in the NMDA receptor channel toreduce entry of calcium into neurons and avoidcalcium-stimulated apoptotic cell death cas-cades. It is uncertain if the neuroprotectiveactivities observed consistently in in vitro stud-ies account for the symptomatic benefitobserved in clinical trials. In vitro experimentalstudies suggest reduced Ab plaque depositionand reduced tau hyperphosphorylation follow-ing treatment with memantine (Martinez-Coria et al. 2010). Physiological studies estab-lish a beneficial effect for long-term potentia-tion (LTP), the physiological correlate ofmemory (Frankiewicz and Parsons 1999). Thislatter observation provides a potential explana-tion for the symptomatic benefit observed withtreatment with memantine.

Clinical trials of memantine have been verysimilar in design to those conducted withAChE-Is, but have involved patients with mod-erate to severe AD, rather than mild to moderateAD. Trials have typically been 6 months in dura-tion with the SIB, CIBICþ, or ADCS-ADL asprimary outcomes and ADL or behavior as sec-ondary outcomes. The magnitude of benefitfrom treatment with memantine is similar tothe magnitude observed for AChE-Is. Two ofthree moderate to severe AD trials with meman-tine showed statistical significance on their pri-mary outcomes (Reisberg et al. 2003b; van Dyck

et al. 2007), including an add-on trial, in whichpatients on long-term treatment with donepe-zil, randomized to receive either placebo oradd-on memantine, showed that add-on ther-apy with memantine produced a statisticallysignificant benefit compared to add-on therapywith placebo (Tariot et al. 2004).

Several clinical trials have been conductedwith memantine patients with mild to moder-ate AD. One of these trials showed benefit oncognition, global assessment, and behavior(Peskind et al. 2006); two trials did not showsignificant drug–placebo differences (Bakchineand Loft 2008; Porsteinsson et al. 2008). Mem-antine is not approved by the FDA for patientswith mild AD. European regulatory authoritiesextended the range of approval for use of mem-antine to patients with MMSE scores of 19 andbelow.

Memantine has a half-life of 60–80 hours, is50% protein bound, is not metabolized by thecytochrome P450 hepatic enzymes, and itsabsorption is not delayed by food (Table 2).Memantine is subject to renal excretion andthe dose should be decreased by 50% in ADpatients with renal failure. Memantine is initi-ated at a dose of 5 mg per day for 1 week, it isthen advanced to 5 mg twice daily for 1 week;the next dose is 10 mg in the morning and5 mg in the evening for 1 week; and the finaldose is 10 mg twice daily. This is the continuingpermanent dose for patients who do not haveevidence of intolerance. Adverse events reportedwith memantine therapy include headache, diz-ziness, and somnolence. There have been re-ports of confusion during the titration periodand occasional reports of hallucinations.

Memantine is approved for moderate tosevere AD and may be given either alone or inconjunction with an AChE-I. No adverse druginteractions have been reported with patientsreceiving AChE-Is plus memantine, and thedrugs are safe when used together. Althoughsome long-term open-label observations sug-gest that combination therapy with an ACHE-Iand memantine may ameliorate the course ofAD (Lopez et al. 2009; Rountree et al. 2009),other studies suggest otherwise (Schneideret al. 2011). Observations in the course of

P.S. Aisen et al.

6 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 7: Symptomatic and Nonamyloid Tau Based Pharmacologic

double-blind placebo-controlled trials sug-gested that patients on therapy decline at equalrates when compared to patients not receivingtherapy (Schneider and Sano 2009).

Patients are usually treated with memantineor a combination of an ACHE-I plus meman-tine until late in the course of AD. Pharmaco-therapy may be continued until the patientssuccumbs or until their physicians and familyconclude that the quality of life of the patientis sufficiently compromised that pharmacother-apy is futile.

Use of Psychotropic Agents to TreatBehavioral Disturbances in AD

Behavioral disturbances are common in AD,including depression, agitation, irritability,aberrant motor behaviors, and psychosis(Cummings 2003). There are no agentsapproved by the FDA specifically for treatmentof behavioral disturbances in AD. Antipsy-chotics—both conventional and atypical—areassociated with increased mortality and someantipsychotics are associated with increasedrisk for cerebrovascular accidents or strokewhen administered to elderly patients withAD. The risk of death is increased from�2.6% to �4.5% during an average of 10 weeksof therapy. Long-term observations suggest thatcontinuing therapy is associated with a continu-ing increased risk for mortality (Kales et al.2007; Ballard et al. 2009). Several 1–12 weeklong clinical trials suggest that risperidone and

possibly other atypical antipsychotic agents inlow doses are efficacious in reducing psychosisand agitation in mainly nursing home patientswith AD (Ballard and Waite 2006; Schneideret al. 2006). Given the evidence of benefitand harm for the use of antipsychotics in AD,the clinician must exercise caution whenprescribing these agents. Strategies for use ofantipsychotics in patients with AD involveavoiding their use in patients with cardiovascu-lar or pulmonary disease (the two most com-mon causes of death in mortality studies),using these agents only in patients for whomnonpharmacologic interventions have failedand the behaviors are extreme, employing treat-ment for only the period required and attempt-ing to eliminate the agents as soon as possible,and informing the patient and caregiver of therisks involved.

Clinical trials have been largely negative inshowing benefit for treatment of depression inAD with antidepressant medications (Lyketsoset al. 2000; Olin et al. 2002; Rosenberg et al.2010; Weintraub et al. 2010). There is no consis-tent evidence base for the use of antidepressantsin AD. Individual practitioners may usepractice-based evidence to guide their thera-peutic decisions. Several recent trials suggestthat use of valproate to treat agitation has nosuperiority over placebo and is associated withsubstantial toxicity (Tariot et al. 2005; Herr-mann et al. 2007). Early trials suggested thebenefit of carbamazepine as a treatment for agi-tation (Tariot et al. 1998). Anxiolytics and

Table 2. Characteristics of memantine

Trade name Namenda (Ebixa or Axura in some countries)Indications Moderate to severe AD (USA) up to MMSE scores of 20 in Europe and AsiaHalf-life 60–80 hoursProtein binding 50%Metabolism by hepatic CYP

enzymesNo

Absorption delayed by food NoTime to peak serum level 3–7 hoursAdjustment in hepatic or renal

diseaseDecrease dose by 50% in patients with renal failure

Titration 5 q.d. for 1 week; 5 b.i.d. for 1 week; 10 mg in a.m. and 5 mg in p.m. for 1 week;10 mg b.i.d. thereafter

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 7

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 8: Symptomatic and Nonamyloid Tau Based Pharmacologic

hypnotics are generally to be avoided in patientswith AD as they may increase confusion. Short-term use of benzodiazepines such as lorazepamor clonazepam may be useful in patients withepisodes of agitation.

There is an urgent need for effective, safe,psychotropic agents for treatment of behavioraldisturbances in AD and other dementingdisorders.

OTHER TREATMENTS MARKETED FOR ADIN SOME COUNTRIES

The following three substances are approved foruse or widely used in some individual countriesfor cognitive impairment syndromes but are notFDA or EMA approved.

Ginkgo Biloba

Ginkgo biloba leaves and extracts are widely usedin over-the-counter preparations marketed inthe USA as food supplements or nutraceuticalsand, as such, explicit health claims are not listedin their labeling (Schneider 2008a). In severalcountries G. biloba is advocated for the treat-ment of a broad range of medical conditionsincluding, as examples, tinnitus and dizziness.The extract that is approved for use in somecountries is Ginkgo biloba extract EGb 761(Ipsen Pharma, and Schwabe Pharmaceuticals),standardized to contain two major constituents:22–27% flavonoids and 5–7% terpene lactones(ginkgolides and bilobalide).

The flavonoids are active as antioxidantsand appear neuroprotective. Ginkgolide B is apotent antagonist of the platelet-activating fac-tor receptor. Ginkgolides A and J variouslyinhibit hippocampal neuron dysfunction andneuronal cell death caused by amyloid-b pro-tein-42 (Ab42). Ginkgolides A and J decreaseAb42-induced pathological behaviors, enhanceneurogenesis in animal models of AD, andinhibit Ab aggregation, providing considerablerationale for G. biloba extracts as potential treat-ments for AD.

Trials in older and younger adults who donot have cognitive impairment show mixedresults at best (Schneider 2008a). One meta-

analysis of eight trials did not find evidencefor cognitive benefits with G. biloba in noncog-nitively impaired participants younger than 60years treated for up to 13 weeks. Two placebo-controlled trials reported contradictory effectsin noncognitively impaired older adults, andthe magnitude of the cognitive effects weresmall in the positive trial.

A systematic review that included 35 clinicaltrials and 4247 participants reported inconsis-tent evidence that G. biloba had clinically sig-nificant benefits for dementia or cognitiveimpairment (Birks and Grimley Evans 2009).One 6-month trial in mild to moderate ADsponsored by Schwabe Pharmaceuticals, con-ducted with the hope of gaining US FDA mar-keting approval, failed to demonstrate efficacy(Schneider et al. 2005), as did another 6-monthtrial performed at British primary care siteswith 120 mg/day doses of EGb 761 (McCarneyet al. 2008).

Perhaps because of its popularity and per-ceived safety there have been three preventiontrials undertaken using EGb 761 at 240 mgdaily doses. A trial involving 118 participantswithout MCI or dementia, all older than 85years, randomized to receive G. biloba extractor placebo and followed up for 42 months,showed a nonsignificant effect for G. biloba todelay progression to MCI (Dodge et al. 2008).Of potential concern, however, was thatmore ischemic strokes and transient ischemicepisodes occurred in the G. biloba group.The GEM trial randomized 3069 persons toG. biloba extract or placebo who had no cogni-tive impairment or MCI for a median durationof more than 6 years and found no clinicaleffects for the extract on cognition or time todementia (DeKosky et al. 2008). A second pre-vention trial, GuidAge, conducted in Franceand involving 2854 participants with memorycomplaints or MCI, randomized to G. bilobaor placebo and followed for more than 5 years,also failed to find effects for ginkgo on the pri-mary outcome of time to onset of AD or otherdementia (Ipsen press release, 22 June 2010).Thus, there is very little evidence for the efficacyof G. biloba either for improving symptoms orpreventing AD.

P.S. Aisen et al.

8 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 9: Symptomatic and Nonamyloid Tau Based Pharmacologic

Cerebrolysin

Cerebrolysin is a somewhat controversial ap-proach to neurotrophic therapy for AD. Thisproduct is a parenterally administered digestedpeptide preparation derived from pig brain. Invitro studies suggest that this peptide mixturehas neuroprotective effects. There is some evi-dence of brain penetration with peripheraladministration. There have been a number ofclinical studies of Cerebrolysin infusion therapyin AD, with some results suggesting sympto-matic benefit (Okamura et al. 2008). As a result,it has being widely used in many countries.Skepticism arises from the poorly defined com-position and mechanisms and inconsistenciesin clinical findings, but some have called formore definitive trials.

Huperzine A

Huperzine A is a plant extract with potent,selective AchE1 inhibition used in China forthe treatment of dementia. Preclinical studiessuggest possible neuroprotective mechanisms.A recent Phase II trial in the USA failed to dem-onstrate efficacy of the usual dose used in China,200 mcg twice daily, but did provide some evi-dence of cognitive enhancement at twice thisdose (Rafii et al. 2011).

DIETARY SUPPLEMENTS, VITAMINS,NUTRICEUTICALS, MEDICAL FOODS

A dietary supplement is defined by US law as aproduct (other than tobacco) that is intendedto supplement the diet; contains one or moredietary ingredients (including vitamins, miner-als, herbs or other botanicals, amino acids, andother substances) or their constituents; isintended to be taken by mouth; and is labeledon the front panel as being a dietary supplement(United States Dietary Supplement Health andEducation Act of 1994 (http://www.fda.gov/opacom/laws/dshea.html#sec3).

The word nutraceutical has no official mean-ing, but was coined to imply nontraditionalproducts with pharmaceutical effects. A dietarysupplement promoter cannot make a health-disease treatment claim for the substance.

Thus, for example, a supplement could be ad-vertised as enhancing “brain power,” brain cellsor concentration, but not as a treatment for ADor attention deficit disorder, as the latter arehealth claims.

Several dietary supplements and vitaminshave been used in clinical trials for AD, cogni-tive impairment, or age associated memory im-pairment (AAMI; older individuals who havecognitive test scores below the norms for youngadults) and have been indirectly promoted byvarious interest groups as treatments for AD.Formulations of these substances vary, with noregulatory standard except that products mustcontain the substance advertised.

Docosahexaenoic Acid

Docosahexaenoic acid (DHA) is the primaryconstituent of membranes in the central nerv-ous system (CNS). DHA levels in brain declinewith age, but can be restored with dietary sup-plementation. Adding DHA to the diet of trans-genic mice with amyloid deposition in brainreduces amyloid accumulation and improvescognitive performance. There is thus a reason-able rationale for studying DHA supplementa-tion as a therapeutic intervention for cognitiveaging and AD.

One trial conducted by a manufacturer ofDHA, Martek, in individuals with AAMI didnot yield significant results. Post-hoc analysessuggest possible benefits in measures of episodicmemory (Okamura et al. 2008).

An NIA-funded trial of DHA for mildto moderate AD was, however, negative(Quinn et al. 2010). Treatment with DHA for18 months increased CSF levels (in the subsetwho underwent lumbar punctures), but didnot alter the decline in cognitive and functionalassessments.

B Vitamins

Elevated homocysteine in blood is considered arisk factor for cardiovascular disease, vasculardementia, and AD. Plasma concentrationsincrease when homocysteine’s metabolism tomethionine or cysteine is impaired, which may

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 9

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 10: Symptomatic and Nonamyloid Tau Based Pharmacologic

occur under a number of circumstances, includ-ing natural aging. Increased blood levels ofhomocysteine are associated with damage tovascular endothelial cells through lipid peroxi-dation and release of endothelium-derivedrelaxing factor, and may increase the risk of vas-cular thrombosis. More relevant to AD, homo-cysteine potentiates the neurotoxic effects ofb-amyloid peptides in vitro. Homocysteineplasma levels can be reduced by as much as30% using regimens of B vitamins (e.g., B121 mg/day, B6 25 mg/day, and folic acid 5 mg/day). A placebo-controlled clinical trial of thesevitamins over 18 months confirmed that sub-stantial reduction in homocysteine could beachieved but did not slow the rate of cognitivedecline in AD compared to placebo (Aisenet al. 2008; Okamura et al. 2008). A study of asimilar regimen in individuals with MCIshowed reduction of whole brain atrophy withvitamin use for 2 years (Smith et al. 2010). Afixed combination of B6, B12, and folate knownas Cerefolin is marketed as a medical food (seebelow) for treatment of metabolic abnormal-ities associated with AD.

Vitamin E

The antioxidant effects of vitamin E (DL-a-tocopherol) might have an impact on reducingclinical progression. In the prototypical antiox-idant trial, vitamin E (1000 IU twice a day) andselegiline (5 mg twice a day), each given alone tomoderately impaired patients with AD, delayedthe time until patients required nursing homeplacement, died, or lost ADL (Sano et al. 1997).

Overall, the delayed time to these end pointswas approximately 7 months compared to pla-cebo. There were no effects on cognition in thisgroup of patients, who were more severelyimpaired than in many other studies. Adverseeffects included falls and syncope in selegilinepatients (9 and 10%, respectively),a-tocopherolpatients (14 and 7%, respectively), and the com-bination (22 and 16%, respectively).

A multicenter trial in MCI patients reportedno significant delay in conversion to AD ordementia in cognitive effects with a-tocopherol(or donepezil) over 3 years of treatment

(Petersen et al. 2005). Taken together, consider-ing the absence of cognitive efficacy and thepotential risks for treatment, vitamin E cannotbe recommended as treatment.

Homotaurine

Originally synthesized and manufactured as adrug in development for AD under the nametramiprosate (3-amino-1-propanesulfonic acid)and the trade name Alzhemed, the amino acidhomotaurine is now sold in Canada as a nutra-ceutical under the brand name Vivimind. Therationale for this is that, although the productis synthesized, homotaurine is also a naturalcomponent of certain seaweeds.

The rationale for its development as a pre-scription drug for AD was as an Ab aggregationinhibitor. After a Phase II safety study in whichCSF Ab was decreased (Aisen et al. 2006), two18-month Phase III trials (one of which wasstopped early) did not show efficacy. The com-pany currently advances its use without a spe-cific health claim as a food supplement.

MEDICAL FOODS

A medical food is “a food which is formulatedto be consumed or administered enterally (ororally) under the supervision of a physician,and which is intended for the specific dietarymanagement of a disease or condition forwhich distinctive nutritional requirements,based on recognized scientific principles, areestablished by medical evaluation,” under theOrphan Drug Amendments of 1988 and theNutrition Labeling and Education Act of 1990(see 21 U.S.C. sec. 360ee(b)(3), 21 C.F.R.sec. 101.9( j)(8), and “Guidance for Industry:Frequently Asked Questions About MedicalFoods” (May 2007), FDA website).

In addition, the contents of a medical foodmust fulfill the Generally Recognized as Safe(GRAS) designation for foods.

Axona (Ketasyn; AC-1202)

A mixture of medium-chain triglycerides,brand named Axona, was marketed in March2009 as a medical food for AD. The rationale

P.S. Aisen et al.

10 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 11: Symptomatic and Nonamyloid Tau Based Pharmacologic

is that AD or other cognitive impairment maybe partly a consequence of impaired glucosemetabolism (see below), and that this can betreated by using medium-chain triglyceridesthat are converted to ketones, raising ketone lev-els, which enhance mitochondrial electrontransport that otherwise was impaired by theimpaired glucose metabolism (Hendersonet al. 2009). Separately, some studies show thata ketogenic diet reduces Ab40 and 42 in trans-genic mice.

In two 12-week placebo-controlled ran-domized trials for AD and AAMI there was nostatistical evidence for cognitive or behavioraleffects (Henderson et al. 2009). In the trialwith 152 AD patients significant improvementson the ADAScog were noted at 6 weeks but notat 12 weeks, and post hoc the very few APOE4-negative patients in the trial seemed to im-prove at 12 weeks (Henderson et al. 2009).The 159-patient trial with AAMI patientswas also not significant, showing two of thememory subscales to be significant in post hocanalyses.

Souvenaid

Another company, Danone, is developing andtesting in clinical trials a brand name medicalfood, Souvenaid, that is comprised of a combi-nation of food supplements, including uridinemonophosphate, choline, omega-3 fatty acids(EPA, DHA), phospholipids, B vitamins, andantioxidants (Scheltens et al. 2010). The ration-ale is that this specific combination might syn-ergistically enhance dendritic spine growth,synapse formation, neurotransmitter precur-sors, and neurotransmitter release, ultimatelyimproving cognitive function. The companyalso claims the combination reduced amyloidproduction and toxicity in the preclinical mod-els. Results of a 12-week, placebo-controlledtrial in 225 patients with AD were nonsignifi-cant on most of the outcomes (Scheltens et al.2010). There are ongoing, longer, and larger tri-als in the USA and Europe.

Thus, there is no evidence for efficacy forthese two medical foods and more studies areneeded.

OTHER NEUROTRANSMITTER-BASEDTREATMENTS

Cholinergic Agonists

A range of M1, muscarinic agonists with variablespecificity for the M1 receptor subtype have beentested in 6-month symptomatic trials. Althoughsome have shown distinct cognitive signals onstandard clinical trials outcomes, they also pro-duce considerable and troublesome cholinergicadverse effects such as gastrointestinal disturban-ces, diaphoresis, syncope, and hypersalivation.The M1 agonists also show salutary effects onAb in preclinical models. There have been no tri-als of lower doses over longer time periods toassess the potential for disease modification.

Neuronal Nicotinic Receptor Agonists

Neuronal nicotinic acetylcholine receptors(NNRs) are widespread throughout the centraland peripheral nervous system. They mediateaspects of memory, attention, arousal, mood,anxiety, and sensory perception. NNR agonistscan affect acetylcholine receptors through fulland partial agonism or positive allosteric mod-ulation. Receptor stimulation may increaserelease of several neurotransmitters implicatedin CNS disorders including dopamine, seroto-nin, glycine, glutamate, and GABA. There maybe a physiological inverted dose-response wherelow doses of NNR agonists enhance cognitionand higher doses do not.

Two classes of NNRs may be involved inCNS disorders: (1) a7, and (2) a4/b2, andserve as targets. The a7 agonists predominatein areas more directly linked to memory andmay also serve a neuroprotective function byreducing oxygen free radicals and nitric oxide.An a7 agonist in particular could be expectedto be both symptomatic and have diseasemodifying properties as it enhances cognitiveperformance in behavioral models that capturedomains of working memory, recognitionmemory, memory consolidation, and sensorygating deficit, increases the release of bothpresynaptic and postsynaptic calcium, andincreases the release of Ach, glutamate, seroto-nin, and dopamine.

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 11

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 12: Symptomatic and Nonamyloid Tau Based Pharmacologic

PDE Inhibition

A rational behind phosphdiesterase 4 (PDE4)inhibition is that it leads to CREB up-regula-tion, which should promote neurotrophinexpression, and thus may have both cognitionenhancing and neuroprotective effects. Themolecules in development so far have not suc-ceeded, having manifested significant cardiacand gastrointestinal adverse effects.

EHT0202 (etazolate HCl) shows bothpotential symptomatic and disease-modifyingeffects in AD, acting as a PDE-4 inhibitorand GABA-A receptor modulator, increasinga-secretase activity and sAPP a secretion, thuslowering Ab. PDE inhibitors involving a varietyof PDE enzymes are being tested in early-phaseAD trials (Okamura et al. 2008).

AMPA Receptor Modulation

More challenging, in terms of safety, is thework being done on neurotrophically high-impact AMPA modulators. Lilly’s high-impactLY451395 failed in an AD Phase II trial,but this trial used tiny doses (0.2 mg for28 days, then 1.0 mg thereafter) which speaksto the low therapeutic index, probably relatedto seizure risk. Cortex believes they haveachieved the necessary balance between trophiceffect and seizure risk with CX-1837, but havenot completed that molecule’s preclinicaltesting.

H3 Antagonists

The H3 receptor has the highest affinity for his-tamine among the four histamine receptors andis predominantly expressed in the cerebralcortex, hippocampus, and hypothalamus inwhich it functions as a presynaptic autoreceptorto regulate histamine release and cholinergicand monoaminergic neurotransmitter release.H3 blockage results in the release of neurotrans-mitters and is associated with enhanced cogni-tive function in preclinical models. Several H3receptor agonists are in early stage clinical trialsfor treatment of the cognitive or motivationalaspects of AD.

OTHER APPROACHES TO AD TREATMENT

There are many drug development programspursuing strategies not directly related to amy-loid, tau, or neurotransmission. As discussedabove, the putative mechanisms may be impos-sible to specify and may be multiple; there isgenerally no way to connect drug activity inexperimental symptoms specific to clinicalresults in the management of AD. Many suchagents are listed in Table 3, and examples arediscussed below.

DRUGS WITH METABOLIC ACTIONS

There is evidence that diabetes may increase therisk of AD, and a variety of animal and humanstudies support the concept that insulin resist-ance in brain may play a role in AD (Craft2007). Increasing attention is being paid tothe possible interrelationship between AD anddiabetes as epidemiologic studies suggest thatpeople with Type 2 diabetes mellitus are twiceas likely to develop AD as nondiabetics. Insulinmay prevent the soluble toxic form of Ab fromdamaging neurons. Insulin protection wasenhanced by the presence of rosiglitazone.Intranasal or IV insulin has been shown toimprove cognitive performance in small studiesof individuals with AD. Additional studies ofinsulin therapy are planned.

PPARg-Agonists

The peroxisome proliferator activated receptorg (PPARg) is activated by a variety of fatty acidsand fatty acid derivatives, and regulates adipo-cyte differentiation and function. PPARg-agonists such as rosiglitazone (Avandia) andpioglitazone (Actos) further mediate insulinactions and are marketed drugs for diabetes.They also may have actions relevant to AD,including anti-inflammatory effects. However,a study of a pioglitazone found mixed results,with no difference on most cognitive measures(Sato et al. 2009; Geldmacher et al. 2011).Six- and 12-month Phase III trials of rosiglita-zone, a PPARg agonist also targeting insu-lin resistance and related pathways failed to

P.S. Aisen et al.

12 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 13: Symptomatic and Nonamyloid Tau Based Pharmacologic

Table 3. Selected drug development programs

Name Sponsor

Proposed mechanism or

therapeutic rationale

Target

population Supportive studies

Nonsupportive

studies/issues Current status

Insulin NIA Correction of insulinabnormalities that maycontribute to pathology andsynaptic dysfunction

MCI, AD Small Phase II RCTs withsome positive effects(Craft et al. 2011)

Additional Phase IItrials planned

Rosiglitazone GSK Anti-inflammatory effect;improve brain insulin resistance

AD Exploratory Phase IIanalyses (Risneret al. 2006)

Phase III Not actively pursued

Dimebon Medivation/Pfizer

Mitochondrial neuroprotectant AD, HD Phase II (Doodyet al. 2008)

Phase IIImonotherapy

Phase III add-onstudy in progress

DHA NIA, Martek Restore membrane function AD, AAMI Epidemiology pluspost-hoc analyses incognitive aging trial

ADCS trial in AD Additional studies ingenetic subgroupsand/or MCI areunder discussion

Cerebrolysin EBEWE Neurotrophic activity AD Effect of some doses oncognition or globalstatus in randomizedtrials

Inconsistentresults

Marketed in manycountries

Statins NIA, Pfizer Alter cholesterol/amyloidpathways

Normal, AD Epidemiology, smallrandomized study

ADCS, LEADe Not actively pursuedfor AD

Estrogen NIA Engage brain estrogen receptors Normal, AD Epidemiology, smallrandomized study

ADCS, WHI, Not actively pursuedfor AD

NSAIDs NIA, Merck Reduce harmful braininflammation

AD, MCI Epidemiology ADCS, Merck Not actively pursuedfor AD

B vitamins NIA Reduce homocysteine AD Epidemiology ADCS ?MCIHGH, IGF-1 Merck, others Normalize HGH-IGF-1 pathways AD, aging Epidemiology MK-677 trial Not actively pursued

for ADLithium Inhibit tau phosphorylation plus

other neuroprotectivemechanisms

AD, tauopathies Preclinical data Negative RCT Not actively pursuedfor AD

Valproic acid NIA Inhibit tau phosphorylation plusother neuroprotectivemechanisms

AD Preclinical data Negative RCT Not actively pursuedfor AD

Continued

Symp

tom

atican

dO

ther

Treatmen

tsfo

rA

lzheim

erD

isease

Cite

this

articleas

Cold

Sprin

gH

arbPersp

ectM

ed2012;2

:a006395

13

www.perspectivesinmedicine.org

on May 19, 2022 - P

ublished by Cold S

pring Harbor Laboratory P

ress http://perspectivesinm

edicine.cshlp.org/D

ownloaded from

Page 14: Symptomatic and Nonamyloid Tau Based Pharmacologic

Table 3. Continued

Name Sponsor

Proposed mechanism or

therapeutic rationale

Target

population Supportive studies

Nonsupportive

studies/issues Current status

Resveratrol NIA SIRT-1 AD, normalaging

In vitro, animal studies Limited brainpenetration

RCT in AD beginning

Axona Accera Correct ketone bodyaccumulation

AD Post-hoc results Negative RCT Marketed as medicalfood

Souvenaid Combination of vitamins toimprove brain cell function

AD Post-hoc results Negative RCT Marketed as medicalfood

Xaliproden Sanofi-Aventis Neuroprotection AD Preclinical data Negative RCT Not actively pursuedfor AD

Vitamin E NIA Antioxidant Normal, MCI,AD

Positive AD trial Negative MCI trial Use limited bytoxicity concerns

Ginkgo biloba NIA, Schwabe,Ipsen

Antioxidant Normal, AD Some randomized trialsin AD

Negative AD andprevention trials

Over-the-counterpreparationswidely used

Idebenone Takeda Antioxidant AD Preclinical data Randomized trial Not actively pursuedfor AD

Acetyl-L-carnitine Sigma Tau Antioxidant AD Preclinical data Randomized trials Two negative RCTsLeuprolide Curaxis Gonadotropin pathways AD Preclinical studies,

epidemiologyRandomized trial Negative RCT

NGF gene delivery NIA Ceregene Neurotrophic AD Phase I open label study Requiresneurosurgicalprocedure

Phase II in progress

P.S.Aisen

etal.

14C

iteth

isarticle

asC

old

Sprin

gH

arbPersp

ectM

ed2012;2

:a006395

www.perspectivesinmedicine.org

on May 19, 2022 - P

ublished by Cold S

pring Harbor Laboratory P

ress http://perspectivesinm

edicine.cshlp.org/D

ownloaded from

Page 15: Symptomatic and Nonamyloid Tau Based Pharmacologic

demonstrate significant benefit (Harringtonet al. 2009; Gold et al. 2010), although post-hocanalysis in an early study suggested benefit inthe APOE 14 negative subgroup (Risner et al.2006).

Dimebon (Latrepirdine)

The course of the dimebon development pro-gram includes a number of interesting lessonson the process of bringing new AD treatmentsforward (Doody 2009). Dimebon was initiallyidentified by Russian scientists who screenedcompounds available for clinical use in thatcountry for activity similar to that of the estab-lished AD treatments. They sought a singlecompound that would combine cholinesteraseinhibition with NMDA antagonism. The leadcompound to arise from this process wasdimebon, a drug used as an antihistamine inRussia until the late 1980s. Following an8-week open trial in 14 patients (Bachurinet al. 2001), dimebon was licensed by a US com-pany, Medivation, and underwent a 6-month,placebo-controlled Phase II trial, conducted inRussia, in 183 individuals with mild to moder-ate AD, similar in design to standard trials usedwith cholinesterase inhibitors and memantine.The results were strikingly positive, with consis-tent favorable effects on both primary and sec-ondary measures of cognition, function, andbehavior (Doody et al. 2008). Further, a blindedextension of the trial to 12 months indicatedthat benefits of the drug in comparison toplacebo appeared to increase with time. Thistrial suggested that dimebon might havebenefits beyond the expectations for a com-bined cholinesterase inhibitor/NMDA antago-nist, and indeed the very limited potencyagainst those targets argued that dimebonmust work by a novel mechanism. Activity invitro against models of mitochondrial toxicitysupported that dimebon may be a mitochondrialcytoprotectant. Thus, a drug identified usingstandard screens appeared to be highly effectivebased on an unexpected and novel mechanism.

An international Phase III trial waslaunched to replicate the Russian study findingsbut including nearly 600 patients, and several

other studies were started to explore the efficacyof dimebon added onto donepezil treatment,and to evaluate efficacy in moderate to severeAD. However, the confirmatory study resultsshowed no efficacy, with no apparent explana-tion for this inconsistency with the RussianPhase II trial. Another Phase III trial, assessingthe addition of dimebon to standard therapyfor 1 year, continues. Lessons from the dimebonprogram include the role of serendipity, and theunpredictability and inconsistency in Phase IIand smaller sample size AD trials learned fromother programs (Schneider 2008b), as well asreasons specific to this drug (Jones 2010).

Statins

Epidemiological evidence suggests that cho-lesterol lowering and the use of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) re-ductase inhibitors (“statins”) may modulatethe enzymatic processing of amyloid precursorprotein and consequently the production ofb-amyloid, and impact the development orprogression of AD. Furthermore, cholesterol-fed rabbits show increased Ab in brain, andhypercholesterolemia induced by a high-cho-lesterol diet results in increased levels of Ab inthe CNS. Statin administration in vitro reducesintra- and extracellular Ab levels in neuronalcultures. Observational studies of patientrecords showed a 60–73% lower prevalence ofand markedly reduced risk for AD in lovastatin(Mevacor)- or pravastatin (Pravachol)-treatedpatients.

Clinical trials, however, have not supportedthe use of statins for AD. A randomized,placebo-controlled trial of pravastatin in 5804subjects with risk factors for vascular diseasereported favorable cardiovascular outcomesbut no significant effect on cognitive functionor disability during an approximately 3-yearfollow-up period (Trompet et al. 2010). In arandomized, placebo-controlled trial of theeffect of simvastatin on cardiovascular out-comes in a subgroup of 5806 patients aged 70or more years, there was no significant differ-ence in the proportion of patients with cog-nitive impairment or in the incidence of

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 15

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 16: Symptomatic and Nonamyloid Tau Based Pharmacologic

dementia over the average 5-year follow-upperiod. Two large placebo-controlled trials,one with simvastatin and another with atorvas-tatin, over 18 months did not show positiveeffects for the drugs in slowing the clinicaldecline (McGuinness et al. 2009).

Estrogens

Preclinical evidence amply demonstrates thatestrogens improve cognitive function throughcholinergic neuroprotective and neurotrophiceffects. Clinical evidence of a possible effectfor estrogen replacement therapy (ERT) hasincluded observations of an inverse relationshipbetween ERT and death-certificate diagnoses ofdementia, several case-control and cohort stud-ies in which hormonal therapy reducing the riskof AD were reported, and small clinical reportssuggesting that estradiol, estrone, or conjugatedequine estrogens enhance cognitive function inAD (Henderson 2006). However, larger studieshave not confirmed benefits (Mulnard et al.2000). Indeed, the large Women’s Health Initia-tive trial revealed an increased risk of dementiaand cognitive impairment in women treatedwith hormone replacement therapy (Shumakeret al. 2003). It is possible but not proven thatestrogen treatment in midlife around meno-pause could be neurotrophic and preventativeof the onset of AD, but beyond a few epidemio-logical studies, evidence is lacking. Particularlyin view of growing concerns about adversehealth effects, the use of estrogen replacementlate in life to prevent or treat MCI or AD cannotbe recommended.

ANTI-INFLAMMATORY DRUGS

There has been enormous interest for more thantwo decades in the suppression of brain inflam-matory activity as an approach to slowingdisease progression in AD. Unfortunately,randomized trials of multiple anti-inflamma-tory regimens, including glucocorticoids (Aisenet al. 2000), nonsteroidal anti-inflammatorydrugs (both nonselective and COX-2 selective)(Aisen et al. 2003; Lyketsos et al. 2007),hydroxychloroquine (Van Gool et al. 2001),

and dapsone have all failed to demonstrateefficacy.

Curcumin is the active ingredient of curryspicy and is widely used as a food additive inAsian cooking. It has antioxidant propertiesand has been shown in experimental modelsto have anti-inflammatory, antioxidant, andanti-amyloid effects (Ringman et al. 2005).There is concern about the brain bioavailabilityof curcumin, however, and improved formula-tions are being sought.

INHIBITOR OF THE RECEPTOR FORADVANCED GLYCATION ENDPRODUCTS

The receptor for advanced glycation endprod-ucts (RAGE) is present on neurons, glia, andendothelial cells and is mainly of interest in dia-betes and vascular disease in which there is anincreased level of various ligands includingadvanced glycation endproducts that bind toit, effecting pro-inflammatory activity. It is ofinterest in AD because of a relationship of ADto diabetes and because the amyloid peptide isalso a RAGE ligand, and this interaction maycontribute to brain inflammation and accumu-lation of amyloid. The latter effect might resultin increased CNS Ab as well as inflammatoryeffects in AD patients. Thus RAGE blockerscould be therapeutically useful in diabetes andAD. After a small Phase II trial indicated the tol-erability of an oral inhibitor of RAGE in AD(Sabbagh et al. 2010), an 18-month trial waslaunched, with results expected soon.

NGF GENE DELIVERY

The most important component of the brain’scapacity to protect the function of cholinergicneurons is nerve growth factor (NGF). Earlyattempts to utilize NGF to protect these neu-rons in AD were unsuccessful because of toxic-ity; NGF delivered into the CSF stimulatesependymal cell proliferation with resulting tox-icity such as pain. The advance of gene deliverymethods has led to a resurgence of interest inNGF; primate studies showed that NGF genedelivery to the cholinergic nucleus basalis

P.S. Aisen et al.

16 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 17: Symptomatic and Nonamyloid Tau Based Pharmacologic

resulted in long-term gene expression with neu-rotrophic effects.

A Phase I study of stereotactic NGF genedelivery using transformed fibroblasts derivedfrom skin biopsies showed that NGF expressioncould be induced in nucleus basalis cells inhumans with AD, with some evidence ofincreased brain function by FDG-PET scanning(Tuszynski et al. 2005). An NIA-funded, ran-domized, sham-surgery controlled Phase II trialof NGF gene delivery using a viral vector is cur-rently in progress.

SIRTUINS AND RESVERATROL

Modifications of DNA and post-translationalnuclear proteins may produce lasting altera-tions in chromatin, alter patterns of geneexpression, and may affect neuroplasticity. Dys-regulation of such epigenetic mechanisms mayaffect aging brain and cognitive impairment.Histone deacetylation (HDAC) may contributeto pathologic transcriptional aberrations inbrain disease and HDAC activation may havetherapeutic potential. Sirtuins, a family of his-tone deacetylation compounds, may be impor-tant to epigenetic mechanisms, and may beinvolved in the cellular protection afforded bycalorie restriction (Gan 2007). Targeting sir-tuins represents a new approach to neurodege-nerative disorders.

There is great interest in the potential bene-fits of polyphenolic compounds derived fromvarious fruits, vegetables, and plants. Polyphe-nols are potent antioxidants in vitro, and havebeen studied as potential therapeutics in cardi-ovascular disease and cancer, in part based onepidemiological evidence. Similarly, there isinterest in polyphenols to reduce the adverseeffects of aging on the function of organsincluding brain; neuroprotective effects havebeen shown in vitro.

Resveratrol is a polyphenol particularlyabundant in the skin of red grapes; it has beensuggested that this may in part explain epide-miological links between red wine consumptionand reduced risk of AD (Vingtdeux et al. 2008).In vivo data have clearly shown the neuropro-tective properties of the naturally occurring

polyphenol resveratrol in rodent models forstress and diseases. Furthermore, recent workin cell cultures and animal models has shed lighton the molecular mechanisms potentiallyinvolved in the beneficial effects of resveratrolintake against the neurodegenerative processin AD. Laboratory studies indicate that resvera-trol is an activator of sirtuins. Resveratrol andother sirtuin activators increase longevity andslow brain atrophy in a number of speciesincluding nonhuman primates.

Although brain penetration of resveratrol islimited, some studies suggest that it may have aperipheral action that reduces brain amyloidaccumulation. A NIA-funded study of the im-pact of resveratrol on biomarkers of AD willbe initiated in early 2011.

SUMMARY

AChE-Is and the NMDA receptor antagonistmemantine are FDA approved for treatment ofAD. Transmitter based therapies represent theonly validated treatments of AD. Deficits intransmitters not addressed by current therapiessuggest that manipulation of these transmittersystems are worthy targets for cognitive en-hancement. A plethora of metabolic, intracellu-lar, and neuroprotective approaches populatethis potential therapeutic space and representnew and emerging treatments for AD. Particu-larly for drug development programs aimingto slow progression of underlying pathobiolog-ical mechanisms, clinical impact is likely to begreatest if treatment is initiated early, prior tothe onset of dementia or even in the presympto-matic stage of the disease process.

ACKNOWLEDGMENTS

Dr. Aisen serves on a scientific advisory boardfor NeuroPhage and serves as a consultant toElan Corporation, Wyeth, Eisai Inc., Schering-Plough Corp., Bristol-Myers Squibb, Eli Lillyand Company, NeuroPhage, Merck & Co.,Roche, Amgen, Genentech, Inc., Abbott, PfizerInc, Novartis, Bayer, and Medivation, Inc. Hereceives research support from Pfizer Inc,Baxter International Inc., and the NIH [NIA

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 17

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 18: Symptomatic and Nonamyloid Tau Based Pharmacologic

U01-AG10483 (PI), NIA U01-AG024904(Coordinating Center Director), NIA R01-AG030048 (PI), and R01-AG16381 (Co-I)];and he has received stock options from Mediva-tion, Inc. and NeuroPhage.

Dr. Cummings has provided consultation toAbbott, Acadia, ADAMAS, Astellas, Baxter,Bayer, Bristol-Meyers Squibb, Eisai, EnVivo,Forest, Genentech, GlaxoSmithKline, Janssen,Lilly, Lundbeck, Medivation, Merck, Neurokos,Novartis, Pfizer, Prana, QR Pharma, reMYND,Signum, Sonexa, Takeda, and Toyama pharma-ceutical companies. He has stock options inADAMAS, Prana, Sonexa, and Neurokos.Dr. Cummings owns the copyright of the Neu-ropsychiatric Inventory.

Dr. Schneider reports being an editor onthe Cochrane Collaboration Dementia andCognitive Improvement Group, which overseessystematic reviews of drugs for cognitiveimpairment and dementia. He received a grantfrom the Alzheimer’s Association for a registryfor dementia and cognitive impairment trials;he is also in receipt of grant or research supportfrom Baxter, Elan Pharmaceuticals, Johnson &Johnson, Eli Lilly, Myriad, Novartis, and Pfizer.He has served as a consultant for or receivedconsulting fees from Abbott Laboratories, ACImmune, Allergan, Allon, Alzheimer Drug Dis-covery Foundation, AstraZeneca, Bristol-MyersSquibb, Elan, Eli Lilly, Exonhit, Forest, Glaxo-SmithKline, Ipsen Pharmaceuticals, Johnson &Johnson, Lundbeck, Myriad, Medavante, Med-ivation, Merck, Merz, Novartis, Pfizer, Roche,Sanofi-Aventis, Schering-Plough, Schwabe,Toyama, and Transition Therapeutics.

REFERENCES

AD2000 Collaborative Group. 2004. Long-term donepeziltreatment in 565 patients with Alzheimer’s disease(AD2000): Randomised double-blind trial. Lancet 363:2105–2115.

Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Tho-mas RG, Weiner MF, Farlow MR, Sano M, GrundmanM, et al. 2000. A randomized controlled trial of predni-sone in Alzheimer’s disease. Alzheimer’s Disease Cooper-ative Study. Neurology 54: 588–593.

Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M,Davis KL, Farlow MR, Jin S, Thomas RG, Thal LJ.2003. Effects of rofecoxib or naproxen vs placebo on

Alzheimer disease progression: A randomized controlledtrial. J Am Med Assoc 289: 2819–2826.

Aisen PS, Saumier D, Briand R, Laurin J, Gervais F, TremblayP, Garceau D. 2006. A Phase II study targeting amyloid-beta with 3APS in mild-to-moderate Alzheimer disease.Neurology 67: 1757–1763.

Aisen PS, Schneider LS, Sano M, Diaz-Arrastia R, van DyckCH, Weiner MF, Bottiglieri T, Jin S, Stokes KT, ThomasRG, et al. 2008. High-dose B vitamin supplementationand cognitive decline in Alzheimer disease: A random-ized controlled trial [see comment]. J Am Med Assoc300: 1774–1783.

Albert MS, Dekosky ST, Dickson D, Dubois B, Feldman HH,Fox NC, Gamst A, Holtzman DM, Jagust WJ, PetersenRC, et al. 2011. The diagnosis of mild cognitive impair-ment due to Alzheimer’s disease: Recommendationsfrom the National Institute on Aging–Alzheimer’s Asso-ciation workgroups on diagnostic guidelines for Alz-heimer’s disease. Alzheimers Dement 7: 270–279.

Bachurin S, Bukatina E, Lermontova N, Tkachenko S, Afa-nasiev A, Grigoriev V, Grigorieva I, Ivanov YU, Sablin S,Zefirov N. 2001. Antihistamine agent dimebon as a novelneuroprotector and a cognition enhancer. Ann NY AcadSci 939: 425–435.

Bakchine S, Loft H. 2008. Memantine treatment in patientswith mild to moderate Alzheimer’s disease: Results of arandomised, double-blind, placebo-controlled 6-monthstudy. J Alzheimers Dis 13: 97–107.

Ballard C, Waite J. 2006. The effectiveness of atypical anti-psychotics for the treatment of aggression and psychosisin Alzheimer’s disease. Cochrane Database Syst RevCD003476.

Ballard C, Hanney ML, Theodoulou M, Douglas S,McShane R, Kossakowski K, Gill R, Juszczak E, Yu LM,Jacoby R. 2009. The dementia antipsychotic withdrawaltrial (DART-AD): Long-term follow-up of a randomisedplacebo-controlled trial. Lancet Neurol 8: 151–157.

Birks J. 2006. Cholinesterase inhibitors for Alzheimer’s dis-ease. Cochrane Database Syst Rev CD005593.

Birks J, Grimley Evans J. 2009. Ginkgo biloba for cognitiveimpairment and dementia. John Wiley, Chichester, UK.

Black S, Roman GC, Geldmacher DS, Salloway S, Hecker J,Burns A, Perdomo C, Kumar D, Pratt R. 2003. Efficacyand tolerability of donepezil in vascular dementia: Posi-tive results of a 24-week, multicenter, international,randomized, placebo-controlled clinical trial. Stroke 34:2323–2330.

Craft S. 2007. Insulin resistance and Alzheimer’s diseasepathogenesis: Potential mechanisms and implicationsfor treatment. Curr Alzheimer Res 4: 147–152.

Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS,Claxton A, Arbuckle M, Callaghan M, Tsai E, PlymateSR, et al. 2011. Intranasal insulin therapy for Alz-heimer disease and amnestic mild cognitive impairment.Arch Neurol doi: 10.1001/archneurol.2011.233.

Cummings J. 2003. Neuropsychiatry of Alzheimer’s diseaseand related dementias. Martin Dunitz, London.

Cummings JL, Mega M, Gray K, Rosenberg-Thompson S,Carusi DA, Gornbein J. 1994. The NeuropsychiatricInventory: Comprehensive assessment of psychopathol-ogy in dementia. Neurology 44: 2308–2314.

P.S. Aisen et al.

18 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 19: Symptomatic and Nonamyloid Tau Based Pharmacologic

DeKosky ST, Williamson JD, Fitzpatrick AL, Kronmal RA,Ives DG, Saxton JA, Lopez OL, Burke G, Carlson MC,Fried LP, et al. 2008. Ginkgo biloba for prevention ofdementia: A randomized controlled trial. J Am Med Assoc300: 2253–2262.

Dodge HH, Zitzelberger T, Oken BS, Howieson D, Kaye J.2008. A randomized placebo-controlled trial of Ginkgobiloba for the prevention of cognitive decline. Neurology70: 1809–1817.

Doody RS. 2009. Dimebon as a potential therapy for Alz-heimer’s disease. CNS Spectr 14: 16–18.

Doody RS, Gavrilova SI, Sano M, Thomas RG, Aisen PS,Bachurin SO, Seely L, Hung D. 2008. Effect of dimebonon cognition, activities of daily living, behaviour, andglobal function in patients with mild-to-moderateAlzheimer’s disease: A randomised, double-blind, pla-cebo-controlled study. Lancet 372: 207–215.

Emre M, Aarsland D, Albanese A, Byrne EJ, Deuschl G, DeDeyn PP, Durif F, Kulisevsky J, van Laar T, Lees A, et al.2004. Rivastigmine for dementia associated with Parkin-son’s disease. N Engl J Med 351: 2509–2518.

Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S,Damaraju CV. 2002. Efficacy of galantamine in probablevascular dementia and Alzheimer’s disease combinedwith cerebrovascular disease: A randomised trial. Lancet359: 1283–1290.

Folstein MF, Folstein SE, McHugh PR. 1975. “Mini-mentalstate.” A practical method for grading the cognitive stateof patients for the clinician. J Psychiatr Res 12: 189–198.

Frankiewicz T, Parsons CG. 1999. Memantine restoreslong term potentiation impaired by tonic N-methyl-D-aspartate (NMDA) receptor activation followingreduction of Mg2þ in hippocampal slices. Neuropharma-cology 38: 1253–1259.

Galasko D, Bennett D, Sano M, Ernesto C, Thomas R,Grundman M, Ferris S. 1997. An inventory to assessactivities of daily living for clinical trials in Alzheimer’sdisease. The Alzheimer’s Disease Cooperative Study. Alz-heimer Dis Assoc Disord 11 (Suppl 2): S33–S39.

Gan L. 2007. Therapeutic potential of sirtuin-activatingcompounds in Alzheimer’s disease. Drug News Perspect20: 233–239.

Geldmacher DS, Frolich L, Doody RS, Erkinjuntti T, VellasB, Jones RW, Banerjee S, Lin P, Sano M. 2006. Realisticexpectations for treatment success in Alzheimer’s disease.J Nutr Health Aging 10: 417–429.

Geldmacher DS, Fritsch T, McClendon MJ, Landreth G.2011. A randomized pilot clinical trial of the safety of pio-glitazone in treatment of patients with Alzheimer disease.Arch Neurol 68: 45–50.

Gelinas I, Gauthier L, McIntyre M, Gauthier S. 1999.Development of a functional measure for persons withAlzheimer’s disease: The disability assessment fordementia. Am J Occup Ther 53: 471–481.

Gold M, Alderton C, Zvartau-Hind M, Egginton S, Saun-ders AM, Irizarry M, Craft S, Landreth G, LinnamagiU, Sawchak S. 2010. Rosiglitazone monotherapy inmild-to-moderate Alzheimer’s disease: Results from arandomized, double-blind, placebo-controlled Phase IIIstudy. Dement Geriatr Cogn Disord 30: 131–146.

Harrington C, Sawchak S, Chiang C, Davies J, Saunders A,Irizarry M, Zvartau-Hind M, van Dyck C, Gold M.

2009. Effects of rosiglitazone-extended release as adjunc-tive therapy to acetylcholinesterase inhibitors over 48weeks on cognition in apoe4-stratified subjects withmild-to-moderate Alzheimer’s disease. AlzheimersDement 5: e17–e18.

Henderson VW. 2006. Estrogen-containing hormone ther-apy and Alzheimer’s disease risk: Understanding discrep-ant inferences from observational and experimentalresearch. Neuroscience 138: 1031–1039.

Henderson S, Vogel J, Barr L, Garvin F, Jones J, Costantini L.2009. Study of the ketogenic agent AC-1202 in mild tomoderate Alzheimer’s disease: A randomized, double-blind, placebo-controlled, multicenter trial. Nutr Metab6: 31.

Herrmann N, Lanctot KL, Rothenburg LS, Eryavec G. 2007.A placebo-controlled trial of valproate for agitation andaggression in Alzheimer’s disease. Dement Geriatr CognDisord 23: 116–119.

Holmes C, Wilkinson D, Dean C, Vethanayagam S, OlivieriS, Langley A, Pandita-Gunawardena ND, Hogg F, Clare C,Damms J. 2004. The efficacy of donepezil in the treatmentof neuropsychiatric symptoms in Alzheimer disease.Neurology 63: 214–219.

Jack CR Jr, Petersen RC, Grundman M, Jin S, Gamst A,Ward CP, Sencakova D, Doody RS, Thal LJ. 2008. Lon-gitudinal MRI findings from the vitamin E and done-pezil treatment study for MCI. Neurobiol Aging 29:1285–1295.

Jelic V, Kivipelto M, Winblad B. 2006. Clinical trials in mildcognitive impairment: Lessons for the future. J NeurolNeurosurg Psychiatry 77: 429–438.

Jicha GA, Parisi JE, Dickson DW, Johnson K, Cha R, IvnikRJ, Tangalos EG, Boeve BF, Knopman DS, Braak H, etal. 2006. Neuropathologic outcome of mild cognitiveimpairment following progression to clinical dementia.Arch Neurol 63: 674–681.

Jones R. 2010. Dimebon disappointment. Alzheimers ResTher 2: 25.

Kales HC, Valenstein M, Kim HM, McCarthy JF, Ganoczy D,Cunningham F, Blow FC. 2007. Mortality risk in patientswith dementia treated with antipsychotics versusother psychiatric medications. Am J Psychiatry 164:1568–1576.

Kavirajan H, Schneider LS. 2007. Efficacy and adverse effectsof cholinesterase inhibitors and memantine in vasculardementia: A meta-analysis of randomised controlled tri-als [see comment]. Lancet Neurol 6: 782–792.

Kimura M, Akasofu S, Ogura H, Sawada K. 2005. Protectiveeffect of donepezil against Ab (1–40) neurotoxicity in ratseptal neurons. Brain Res 1047: 72–84.

Krishnan KR, Charles HC, Doraiswamy PM, Mintzer J,Weisler R, Yu X, Perdomo C, Ieni JR, Rogers S. 2003.Randomized, placebo-controlled trial of the effects ofdonepezil on neuronal markers and hippocampal vol-umes in Alzheimer’s disease. Am J Psychiatry 160:2003–2011.

Lopez OL, Becker JT, Wahed AS, Saxton J, Sweet RA, WolkDA, Klunk W, Dekosky ST. 2009. Long-term effects ofthe concomitant use of memantine with cholinesteraseinhibition in Alzheimer disease. J Neurol Neurosurg Psy-chiatry 80: 600–607.

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 19

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 20: Symptomatic and Nonamyloid Tau Based Pharmacologic

Lyketsos CG, Sheppard JM, Steele CD, Kopunek S, SteinbergM, Baker AS, Brandt J, Rabins PV. 2000. Randomized,placebo-controlled, double-blind clinical trial of sertra-line in the treatment of depression complicating Alz-heimer’s disease: Initial results from the Depression inAlzheimer’s Disease study. Am J Psychiatry 157:1686–1689.

Lyketsos CG, Breitner JC, Green RC, Martin BK, Meinert C,Piantadosi S, Sabbagh M. 2007. Naproxen and celecoxibdo not prevent AD in early results from a randomizedcontrolled trial. Neurology 68: 1800–1808.

Martinez-Coria H, Green KN, Billings LM, Kitazawa M,Albrecht M, Rammes G, Parsons CG, Gupta S, BanerjeeP, LaFerla FM. 2010. Memantine improves cognitionand reduces Alzheimer’s-like neuropathology in trans-genic mice. Am J Pathol 176: 870–880.

McCarney R, Fisher P, Iliffe S, van Haselen R, Griffin M, vander Meulen J, Warner J. 2008. Ginkgo biloba for mild tomoderate dementia in a community setting: A pragmatic,randomised, parallel-group, double-blind, placebo-controlled trial. Int J Geriatr Psychiatry 23: 1222–1230.

McGuinness B, Craig D, Bullock R, Passmore P. 2009. Statinsfor the prevention of dementia. Cochrane Database SystRev CD003160.

McKeith I, Del Ser T, Spano P, Emre M, Wesnes K, Anand R,Cicin-Sain A, Ferrara R, Spiegel R. 2000. Efficacy of riva-stigmine in dementia with Lewy bodies: A randomised,double-blind, placebo-controlled international study.Lancet 356: 2031–2036.

Mulnard RA, Cotman CW, Kawas C, van Dyck CH, Sano M,Doody R, Koss E, Pfeiffer E, Jin S, Gamst A, et al. 2000.Estrogen replacement therapy for treatment of mild tomoderate Alzheimer disease: A randomized controlledtrial. Alzheimer’s Disease Cooperative Study. J Am MedAssoc 283: 1007–1015.

Okamura N, Funaki Y, Tashiro M, Kato M, Ishikawa Y, Mar-uyama M, Ishikawa H, Meguro K, Iwata R, Yanai K. 2008.In vivo visualization of donepezil binding in the brain ofpatients with Alzheimer’s disease. Br J Clin Pharmacol 65:472–479.

Olin JT, Katz IR, Meyers BS, Schneider LS, Lebowitz BD.2002. Provisional diagnostic criteria for depression ofAlzheimer disease: Rationale and background. Am J Ger-iatr Psychiatry 10: 129–141.

Peskind ER, Potkin SG, Pomara N, Ott BR, Graham SM,Olin JT, McDonald S. 2006. Memantine treatment inmild to moderate Alzheimer disease: A 24-week random-ized, controlled trial. Am J Geriatr Psychiatry 14:704–715.

Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, RabinsPV, Ritchie K, Rossor M, Thal L, Winblad B. 2001. Cur-rent concepts in mild cognitive impairment. Arch Neurol58: 1985–1992.

Petersen RC, Thomas RG, Grundman M, Bennett D, DoodyR, Ferris S, Galasko D, Jin S, Kaye J, Levey A, et al. 2005.Vitamin E and donepezil for the treatment of mild cog-nitive impairment [see comment]. New Engl J Med 352:2379–2388.

Porsteinsson AP, Grossberg GT, Mintzer J, Olin JT. 2008.Memantine treatment in patients with mild to moderateAlzheimer’s disease already receiving a cholinesterase

inhibitor: A randomized, double-blind, placebo-controlled trial. Curr Alzheimer Res 5: 83–89.

Quinn JF, Raman R, Thomas RG, Yurko-Mauro K, NelsonEB, Van Dyck C, Galvin JE, Emond J, Jack CR Jr, WeinerM, et al. 2010. Docosahexaenoic acid supplementationand cognitive decline in Alzheimer disease: A random-ized trial. J Am Med Assoc 304: 1903–1911.

Rafii MS, Walsh S, Little JT, Behan KE, Reynolds B, Ward CJ,Thomas R, Aisen PS. 2011. A phase II trial of huperzine Ain mild to moderate Alzheimer disease. Neurology 76:1389–1394.

Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, MobiusHJ. 2003a. Memantine in moderate-to-severe Alzhei-mer’s disease. N Engl J Med 348: 1333–1341.

Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, MobiusHJ, Memantine Study Group. 2003b. Memantine inmoderate-to-severe Alzheimer’s disease [see comment].New Engl J Med 348: 1333–1341.

Ringman JM, Frautschy SA, Cole GM, Masterman DL,Cummings JL. 2005. A potential role of the curry spicecurcumin in Alzheimer’s disease. Curr Alzheimer Res 2:131–136.

Risner ME, Saunders AM, Altman JF, Ormandy GC, Craft S,Foley IM, Zvartau-Hind ME, Hosford DA, Roses AD.2006. Efficacy of rosiglitazone in a genetically definedpopulation with mild-to-moderate Alzheimer’s disease.Pharmacogenomics J 6: 246–254.

Rosen WG, Mohs RC, Davis KL. 1984. A new rating scale forAlzheimer’s disease. Am J Psychiatry 141: 1356–1364.

Rosenberg PB, Drye LT, Martin BK, Frangakis C, Mintzer JE,Weintraub D, Porsteinsson AP, Schneider LS, Rabins PV,Munro CA, et al. 2010. Sertraline for the treatment ofdepression in Alzheimer disease. Am J Geriatr Psychiatry18: 136–145.

Rountree SD, Chan W, Pavlik VN, Darby EJ, Siddiqui S,Doody RS. 2009. Persistent treatment with cholinesteraseinhibitors and/or memantine slows clinical progressionof Alzheimer disease. Alzheimers Res Ther 1: 7.

Sabbagh MN, Agro A, Bell J, Aisen PS, Schweizer E, GalaskoD. 2010. PF-04494700, an oral inhibitor of receptor foradvanced glycation end products (RAGE), in Alzheimerdisease. Alzheimer Dis Assoc Disord 25: 206–212.

Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K,Grundman M, Woodbury P, Growdon J, Cotman CW,Pfeiffer E, et al. 1997. A controlled trial of selegiline,alpha-tocopherol, or both as treatment for Alzheimer’sdisease. The Alzheimer’s Disease Cooperative Study[see comment]. New Engl J Med 336: 1216–1222.

Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, IwamotoT. 2011. Efficacy of PPAR-gamma agonist pioglitazone inmild Alzheimer disease. Neurobiol Aging 32: 1626–1633.

Scheltens P, Kamphuis PJGH, Verhey FRJ, Olde RikkertMGM, Wurtman RJ, Wilkinson D, Twisk JWR, Kurz A.2010. Efficacy of a medical food in mild Alzheimer’s dis-ease: A randomized, controlled trial. Alzheimers Dement6: 1–10; e11.

Schmitt FA, Ashford W, Ernesto C, Saxton J, Schneider LS,Clark CM, Ferris SH, Mackell JA, Schafer K, Thal LJ.1997. The severe impairment battery: Concurrent valid-ity and the assessment of longitudinal change in Alz-heimer’s disease. The Alzheimer’s Disease CooperativeStudy. Alzheimer Dis Assoc Disord 11 (Suppl 2): S51–S56.

P.S. Aisen et al.

20 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 21: Symptomatic and Nonamyloid Tau Based Pharmacologic

Schneider LS. 2008a. Ginkgo biloba extract and preventingAlzheimer disease [comment]. J Am Med Assoc 300:2306–2308.

Schneider LS. 2008b. Issues in design and conduct of clinicaltrials for cognitive-enhancing drugs. In Animal andtranslational models for CNS drug discovery (ed. RobertAM, Franco B), pp. 21–76. Academic, San Diego.

Schneider LS, Sano M. 2009. Current Alzheimer’s diseaseclinical trials: Methods and placebo outcomes. Alz-heimers Dement 5: 388–397.

Schneider LS, Olin JT, Doody RS, Clark CM, Morris JC,Reisberg B, Schmitt FA, Grundman M, Thomas RG, Fer-ris SH. 1997. Validity and reliability of the Alzheimer’sDisease Cooperative Study—clinical global impressionof change. The Alzheimer’s Disease Cooperative Study.Alzheimer Dis Assoc Disord 11 (Suppl 2): S22–S32.

Schneider LS, DeKosky ST, Farlow MR, Tariot PN, Hoerr R,Kieser M. 2005. A randomized, double-blind, placebo-controlled trial of two doses of Ginkgo biloba extract indementia of the Alzheimer’s type [see comment]. CurrAlzheimer Res 2: 541–551.

Schneider LS, Dagerman K, Insel PS. 2006. Efficacy andadverse effects of atypical antipsychotics for dementia:Meta-analysis of randomized, placebo-controlled trials.Am J Geriatr Psychiatry 14: 191–210.

Schneider LS, Insel PS, Weiner MW, for the Alzheimer’s Dis-ease Neuroimaging Initiative. 2011. Treatment with cho-linesterase inhibitors and memantine of patients in theAlzheimer’s Disease Neuroimaging Initiative. Arch Neu-rol 68: 58–66.

Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ock-ene JK, Hendrix SL, Jones BN 3rd, Assaf AR, Jackson RD,et al. 2003. Estrogen plus progestin and the incidence ofdementia and mild cognitive impairment in postmeno-pausal women: The Women’s Health Initiative MemoryStudy: A randomized controlled trial. J Am Med Assoc289: 2651–2662.

Smith AD, Smith SM, de Jager CA, Whitbread P, Johnston C,Agacinski G, Oulhaj A, Bradley KM, Jacoby R, Refsum H.2010. Homocysteine-lowering by B vitamins slows therate of accelerated brain atrophy in mild cognitiveimpairment: A randomized controlled trial. PLoS One5: e12244.

Tariot PN, Erb R, Podgorski CA, Cox C, Patel S, JakimovichL, Irvine C. 1998. Efficacy and tolerability of carbamaze-pine for agitation and aggression in dementia. Am J Psy-chiatry 155: 54–61.

Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDo-nald S, Gergel I. 2004. Memantine treatment in patientswith moderate to severe Alzheimer disease already receiv-ing donepezil: A randomized controlled trial. J Am MedAssoc 291: 317–324.

Tariot PN, Raman R, Jakimovich L, Schneider L, Porsteins-son A, Thomas R, Mintzer J, Brenner R, Schafer K, Thal L.2005. Divalproex sodium in nursing home residents withpossible or probable Alzheimer Disease complicated byagitation: A randomized, controlled trial. Am J GeriatrPsychiatry 13: 942–949.

Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM,Murphy MB, Ford I, Macfarlane PW, Sattar N, PackardCJ, et al. 2010. Pravastatin and cognitive function in theelderly. Results of the PROSPER study. J Neurol 257:85–90.

Tuszynski MH, Thal L, Pay M, Salmon DP, HS UBakay R,Patel P, Blesch A, Vahlsing HL, Ho G, et al. 2005. A phase1 clinical trial of nerve growth factor gene therapy for Alz-heimer disease. Nat Med 11: 551–555.

van Dyck CH, Tariot PN, Meyers B, Resnick ME. 2007. A24-week randomized, controlled trial of memantine inpatients with moderate-to-severe Alzheimer disease. Alz-heimer Dis Assoc Disord 21: 136–143.

Van Gool WA, Weinstein HC, Scheltens P, Walstra GJ. 2001.Effect of hydroxychloroquine on progression of dementiain early Alzheimer’s disease: An 18-month randomised,double-blind, placebo-controlled study. Lancet 358:455–460.

Vingtdeux V, Dreses-Werringloer U, Zhao H, Davies P, Mar-ambaud P. 2008. Therapeutic potential of resveratrol inAlzheimer’s disease. BMC Neurosci 9 (Suppl 2): S6.

Weintraub D, Rosenberg PB, Drye LT, Martin BK, FrangakisC, Mintzer JE, Porsteinsson AP, Schneider LS, Rabins PV,Munro CA, et al. 2010. Sertraline for the treatment ofdepression in Alzheimer disease: Week-24 outcomes.Am J Geriatr Psychiatry 18: 332–340.

Whitehead A, Perdomo C, Pratt RD, Birks J, Wilcock GK,Evans JG. 2004. Donepezil for the symptomatic treat-ment of patients with mild to moderate Alzheimer’s dis-ease: A meta-analysis of individual patient data fromrandomised controlled trials. Int J Geriatr Psychiatry 19:624–633.

Wimo A, Winblad B. 2003. Resource utilisation in dementia:RUD Lite. Brain Aging 3: 48–59.

Winblad B, Poritis N. 1999. Memantine in severe dementia:results of the 9M-Best Study (benefit and efficacy inseverely demented patients during treatment with mem-antine). Int J Geriatr Psychiatry 14: 135–146.

Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G,Wimo A, Wetterholm AL, Zhang R, Haglund A, SubbiahP. 2001. A 1-year, randomized, placebo-controlled studyof donepezil in patients with mild to moderate AD. Neu-rology 57: 489–495.

Symptomatic and Other Treatments for Alzheimer Disease

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006395 21

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 22: Symptomatic and Nonamyloid Tau Based Pharmacologic

November 30, 20112012; doi: 10.1101/cshperspect.a006395 originally published onlineCold Spring Harb Perspect Med 

 Paul S. Aisen, Jeffrey Cummings and Lon S. Schneider Alzheimer DiseaseSymptomatic and Nonamyloid/Tau Based Pharmacologic Treatment for

Subject Collection The Biology of Alzheimer Disease

Animal Models of Alzheimer DiseaseFrank M. LaFerla and Kim N. Green

Alzheimer Disease in 2020

Dennis J. SelkoeDavid M. Holtzman, Eckhard Mandelkow and

-Peptide Clearance in Alzheimer DiseaseβNeurovascular Dysfunction and Faulty Amyloid

ZlokovicAbhay P. Sagare, Robert D. Bell and Berislav V.

The Genetics of Alzheimer DiseaseRudolph E. Tanzi

-ProteinβTreatment Strategies Targeting Amyloid

PangalosDale Schenk, Guriqbal S. Basi and Menelas N.

Fluid Biomarkers in Alzheimer DiseaseKaj Blennow, Henrik Zetterberg and Anne M. Fagan

DiseaseLysosomal System in Alzheimer−Autophagic

Proteasome System and the−The Ubiquitin

Ralph NixonYasuo Ihara, Maho Morishima-Kawashima and

Epidemiology of Alzheimer DiseaseRichard Mayeux and Yaakov Stern

Network Dysfunction-Protein: Synaptic andβNeurotoxicity of Amyloid

Lennart Mucke and Dennis J. SelkoeNeurofibrillary DegenerationBiochemistry and Cell Biology of Tau Protein in

Eva-Maria Mandelkow and Eckhard Mandelkow-ProteinβProteolytic Degradation of Amyloid

Takaomi Saido and Malcolm A. Leissring Deposits in Alzheimer Disease-Protein and AmyloidβBiochemistry of Amyloid

Colin L. Masters and Dennis J. SelkoeBrain Imaging in Alzheimer Disease

al.Keith A. Johnson, Nick C. Fox, Reisa A. Sperling, et Disease

The Neuropsychological Profile of Alzheimer

SalmonSandra Weintraub, Alissa H. Wicklund and David P.

Pharmacologic Treatment for Alzheimer DiseaseSymptomatic and Nonamyloid/Tau Based

SchneiderPaul S. Aisen, Jeffrey Cummings and Lon S.

Normal Biology and Roles in Alzheimer DiseaseApolipoprotein E and Apolipoprotein E Receptors:

David M. Holtzman, Joachim Herz and Guojun Bu

http://perspectivesinmedicine.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2012 Cold Spring Harbor Laboratory Press; all rights reserved

on May 19, 2022 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from