Alzheimer's disease: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
→‎Epidemiology: better no quotes in refs
Line 932: Line 932:
[[Image:Donepezil3d.png‎|right|thumb|3d molecular spacefill of [[donepezil]], an [[acetylcholinesterase inhibitor]] used in the treatment of AD.]]
[[Image:Donepezil3d.png‎|right|thumb|3d molecular spacefill of [[donepezil]], an [[acetylcholinesterase inhibitor]] used in the treatment of AD.]]


AS of 2008 only four medications are approved for AD by regulatory agencies as the U.S. [[Food and Drug Administration]] (FDA), the [[European Medicines Agency]] (EMEA) and the Japanese [[Ministry of Health, Labour and Welfare (Japan)|PMDA]]. Three are [[acetylcholinesterase inhibitor]]s and the other is [[memantine]], an [[NMDA receptor]] [[receptor antagonist|antagonist]].
There is an observed reduction in activity of the [[cholinergic]] neurons in the disease. [[Acetylcholinesterase inhibitor]]s reduce the rate at which [[acetylcholine]] (ACh) is broken down and hence increase the concentration of ACh in the brain (combatting the loss of ACh caused by the death of the cholinergin neurons). AChE-inhibitors seem to modestly moderate symptoms but do not alter the course of the underlying dementing process.<ref name="pmid17979717">{{cite journal

|author=Musiał A, Bajda M, Malawska B
A reduction in the activity of the [[cholinergic]] neurons in the disease is well known.<ref name="pmid8534419">{{cite journal
|title=Recent developments in cholinesterases inhibitors for Alzheimer's disease treatment
|author=Geula C, Mesulam MM
|journal=Current Medicinal Chemistry
|title=Cholinesterases and the pathology of Alzheimer disease
|volume=14
|journal=Alzheimer Dis Assoc Disord
|issue=25
|volume=9 Suppl 2
|pages=2654–2679
|issue=
|year=2007
|pages=23-8
|pmid=17979717
|year=1995
}}</ref>They have also been critized for their reduced clinical utility, cost and side effects.<ref>Effectiveness of cholinesterase inhibitors questioned:
|pmid=8534419
* {{cite journal
|doi=
|author=Courtney C, Farrell D, Gray R, Hills R, Lynch L, Sellwood E, Edwards S, Hardyman W, Raftery J, Crome P, Lendon C, Shaw H, Bentham P
}}</ref> [[Acetylcholinesterase inhibitor]]s reduce the rate at which [[acetylcholine]] (ACh) is broken down and hence increase the concentration of ACh in the brain, combatting the loss of ACh caused by the death of the cholinergin neurons.<ref name="pmid11105732">{{cite journal
|title=Long-term donepezil treatment in 565 patients with Alzheimer's disease (AD2000): randomised double-blind trial
|author=Stahl SM
|journal=Lancet
|title=The new cholinesterase inhibitors for Alzheimer's disease, Part 2: illustrating their mechanisms of action
|volume=363
|journal=J Clin Psychiatry
|issue=9427
|volume=61
|pages=2105–15
|issue=11
|year=2004
|pages=813-4
|pmid=15220031
|year=2000
|doi=10.1016/S0140-6736(04)16499-4
|pmid=11105732
}}
|doi=
* {{cite journal
}}</ref> Cholinesterase inhibitors currently approved include [[donepezil]] (Brand name ''Aricept''), [[galantamine]] (''Razadyne''), and [[rivastigmine]] (Marketed as ''Exelon'' and ''Exelon Patch''). <ref>{{cite web
|url=http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a697032.html
|Donepezil
|accessdate=2008-03-20
|date=2007-01-08
|publisher= US National Library of Medicine (Medline)
}}</ref><ref>{{cite web
|url=http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a699058.html
|Galantamine
|accessdate=2008-03-20
|date=2007-01-08
|publisher= US National Library of Medicine (Medline)
}}</ref><ref>{{cite web
|url=http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a602009.html
|Rivastigmine
|accessdate=2008-03-20
|date=2007-01-08
|publisher= US National Library of Medicine (Medline)
}}</ref><ref>{{cite web
|url=http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a607078.html
|Rivastigmine Transdermal
|accessdate=2008-03-20
|date=2007-01-08
|publisher= US National Library of Medicine (Medline)
}}</ref> There is substantial evidence for the efficacy of these medications in mild to moderate Alzheimer’s disease,<ref name="pmid16437532">{{cite journal
|author=Birks J
|title=Cholinesterase inhibitors for Alzheimer's disease
|journal=Cochrane Database Syst Rev
|volume=
|issue=1
|pages=CD005593
|year=2006
|pmid=16437532
|doi=10.1002/14651858.CD005593
}}</ref> and some evidence for their use in the advanced stage, being only donepezil approved for this stage.<ref name="pmid16437430">{{cite journal
|author=Birks J, Harvey RJ
|title=Donepezil for dementia due to Alzheimer's disease
|journal=Cochrane Database Syst Rev
|volume=
|issue=1
|pages=CD001190
|year=2006
|pmid=16437430
|doi=10.1002/14651858.CD001190.pub2
}}</ref> Their use in [[mild cognitive impairment]] has not been associated with any delay in the onset of AD.<ref name="pmid18044984">{{cite journal
|author=Raschetti R, Albanese E, Vanacore N, Maggini M
|author=Raschetti R, Albanese E, Vanacore N, Maggini M
|title=Cholinesterase inhibitors in mild cognitive impairment: a systematic review of randomised trials
|title=Cholinesterase inhibitors in mild cognitive impairment: a systematic review of randomised trials
|journal=PLoS Medicine
|journal=PLoS Med.
|volume=4
|volume=4
|issue=11
|issue=11
Line 963: Line 1,008:
|pmid=18044984
|pmid=18044984
|doi=10.1371/journal.pmed.0040338
|doi=10.1371/journal.pmed.0040338
}}</ref> Donepezil and galantamine are taken orally while rivastigmine has oral forms and a once-daily [[transdermal patch]].<ref>Rivastigmine transdermal patch:
}}
* {{cite journal
| author = Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt H, van den Bussche H
| title = Cholinesterase inhibitors for patients with Alzheimer's disease: systematic review of randomised clinical trials
| journal = BMJ
| volume = 331
| issue = 7512
| pages = 321–327
| year = 2005
| pmid = 16081444
}}</ref>
Examples currently marketed include [[donepezil]] (Brand name ''Aricept''), [[galantamine]] (''Razadyne'') and [[rivastigmine]] (Marketed as ''Exelon'' and ''Exelon Patch''). Donepezil and galantamine are taken orally. Rivastigmine has oral forms and a once-daily [[transdermal patch]].<ref>Rivastigmine transdermal patch:
* {{cite web
* {{cite web
|url=http://www.aricept.com/content/pi.pdf
|url=http://www.aricept.com/content/pi.pdf

Revision as of 11:32, 21 March 2008

Alzheimer's disease
SpecialtyNeurology Edit this on Wikidata
Frequency5.05% (Europe)

Alzheimer's disease (AD), also called Alzheimer disease or simply Alzheimer's, is the most common cause of dementia, afflicting 24 million people worldwide. Alzheimer's is a degenerative and terminal disease for which there is currently no known cure. In its most common form, it occurs in people over 65 years old although a less-prevalent early-onset form also exists.[1] The disease can begin many years before it is eventually diagnosed. In its early stages, short-term memory loss is the most common symptom, often initially thought to be caused by aging or stress by the sufferer.[2] Later symptoms include confusion, anger, mood swings, language breakdown, long-term memory loss, and the general withdrawal of the sufferer as his or her senses decline.[2][3] Gradually the sufferer loses minor, and then major bodily functions, until death occurs.[4] Although the symptoms are common, each individual experiences the symptoms in unique ways.[5] The duration of the disease is estimated as being between 5 and 20 years.[6][7]

The symptoms of Alzheimer's disease are generally reported to a physician when memory-loss causes concern, and on suspecting Alzheimer’s disease, the physician or healthcare specialists will confirm the diagnosis with a behavioral assessment and cognitive tests, often followed by a brain scan.[8]

The cause and progression of Alzheimer's disease is not well understood, but is associated with plaques and tangles in the brain.[9] Possible causes and potential cures of the disease have been conjectured, with varying evidence supporting each claim. No treatment has been found to stop or reverse the disease, and it is not known whether current treatments slow the progression, or simply manage the symptoms. Many preventative measures have been suggested for Alzheimer's disease, but their value is often uncertain: mental stimulation, exercise and a balanced diet are usually recommended, both as a possible prevention and as a sensible way of managing the disease.[10]

Due to the incurable and degenerative nature of the disease care-management of Alzheimer's is essential. The role of the main caregiver is often taken by the spouse or a close relative.[11] Caregivers may themselves suffer from stress, over-work, depression, and being physically hit or struck.[12]

Characteristics

The disease course is typically divided into four stages, with a different pattern of cognitive and functional impairment occurring at each stage.

Predementia

Detailed neuropsychological testing can reveal mild cognitive difficulties up to eight years before a person fullfils clinical criteria of diagnosis.[13] It is not yet clear if these early symptoms affect daily living activities. Recent studies show impairments in the most complex activities.[14] The most noticeable deficit is short-term memory loss and the resultant inability to acquire new information. In addition, subtle executive problems or semantic memory impairments can also occur.[15][16] Apathy can be observed at this stage, and is the most common and persistent neuropsychiatric symptom throughout the course of the disease.[17][18][19] This stage of the disease has also been termed mild cognitive impairment,[20] but there is still a debate on whether this term corresponds to a different diagnostic entity by itself or just a first step of the disease.[21]

Early dementia

File:Portion of Reagan's Alzheimer's letter.png
In 1994 United States ex-president R. Reagan informed via a hand-written letter of his diagnosis of AD. In this letter, partly shown in picture, clear signs of the disease can be recognised.

In most people with the disease the increasing impairments in learning and memory will lead to diagnosis, while in a small proportion of them language, executive or visuoconstructional difficulties will be more salient.[22] Nevertheless memory problems do not affect all memory subcapacities equally. Older memories of the patient's life (episodic memory) and facts he learned (declarative memory); or implicit memory (the memory of the body on how to do things, such as using a knife to eat) are affected to a much lesser degree than the capacities needed to learn new facts or make new memories.[23][24] On the other hand, language problems are mainly characterized by a shrinking vocabulary and a decreased word fluency which leads to a general impoverishment of oral and written language but the person with the disease is usually capable of communicating ideas adequately.[25][26][27] While performing fine motor tasks such as writing, drawing or dressing, certain visoconstructional difficulties, or apraxia, may be present, which may appear as clumsiness.[28] As the disease progresses to the middle stage, patients might still be able to live and perform tasks independently for most of the time, but may need assistance or supervision with the most complicated activities.[22]

Moderate dementia

In the early stage, people with Alzheimer's can usually care for themselves. At the moderate stage, progressive deterioration seriously hinders the possibility of independence.[22] Language difficulties become clearly noticeable: the person makes frequent paraphasias due to difficulties in finding words, and content is poor. Reading and writing are also progressively forgotten.[25][29] As time passes, complex motor sequences become less coordinated, costing the patient most of their daily-living abilities.[30] Memory problems worsen, and the person may not recognize close relatives.[31] Long-term memory, which was previously left intact, is now also impaired.[32] Patients are usually almost completely unaware of their own deficits, and behavior changes are the norm. Common neuropsychiatric manifestations in this stage are irritability and labile affect, leading to crying or outbursts of unpremeditated aggression and physical violence, even in patients whose life-long behavior has been peaceful. Approximately 30% of the patients also develop illusionary misidentifications and other delusional symptoms.[17][33] Often urinary incontinence develops.[34] Because of the communication deficit along with delusions, patients often resist when caregivers attempt to provide care.[35] It is important to prevent escalation of resistiveness to care into combativeness when patient might strike out. All these symptoms create stress for relatives and caretakers, increasing the likelihood of moving the patient from home care to other long-term care facilities.[22][36]

Advanced

Pressure ulcers are a common complication in advanced AD.

In the last stage of Alzheimer's disease all human behavior is likely to become entirely automatic. Language is reduced to simple phrases or even single words before being lost altogether.[25] Nevertheless many patients can receive and return emotional signals long after the loss of verbal language.[37] Although aggressiveness can still present, extreme apathy and exhaustion are much more common.[22] Patients will ultimately not be able to perform even the most simple tasks independently. Finally, deterioration of muscle and mobility will develop, leading the patient to become bedridden[38] and to lose the ability to feed oneself[39] if death from some external cause, such as infection due to pressure ulcers or pneumonia, does not occur first.[40][41]

Causes

Most cases of Alzheimer's disease do not exhibit familial inheritance. At least 80% of sporadic AD cases involve genetic risk factors. Inheritance of the ε4 allele of the apolipoprotein E (ApoE) gene is regarded as a risk factor for development of up to 50% of late-onset sporadic Alzheimer's. The presence of this gene allele along with infection by Herpes simplex virus type 1 (HSV-1) further increases the risk of Alzheimer's disease. Several viral-host interactions are postulated, most relating to HSV-1’s targeting of Alzheimer’s susceptibility genes.[42][43] Genetic experts agree that there are other risk and protective factor genes that influence the development of late onset Alzheimer's disease. Over 400 genes have been tested for association with late-onset sporadic AD.[44] [45]

Five to ten percent of AD cases involve a clear familial pattern of inheritance in which the patient has at least two first-degree relatives with a history of AD. These cases often have an early age of onset (usually younger than sixty years). Nearly 200 different mutations in the presenilin-1 or presenilin-2 genes have been documented in over 500 families. Mutations of presenilin 1 (PS1) lead to the most aggressive form of familial Alzheimer's disease. Over twenty different mutations in the amyloid precursor protein (APP) gene on chromosome 21 can also cause early onset of the disease. The presenilins have been identified as essential components of the proteolytic processing machinery that produces beta amyloid peptides through cleavage of APP. Most mutations in the APP and presenilin genes increase the production of a small protein (peptide) called Abeta42, the main component of senile plaques in brains of AD patients.[citation needed]

Pathophysiology

Neuropathology

File:Alzheimer's disease - MRI.jpg
MRI images of a normal aged brain (right) and an Alzheimer's patient's brain (left). In the Alzheimer brain, atrophy is clearly seen.

At a macroscopic level, AD is characterized by loss of neurons and synapses in the cerebral cortex and certain subcortical regions. This results in gross atrophy of the affected regions, including degeneration in the temporal lobe and parietal lobe, and parts of the frontal cortex and cingulate gyrus.[46]

Both amyloid plaques and neurofibrillary tangles are clearly visible by microscopy in AD brains.[9] Plaques are dense, mostly insoluble deposits of amyloid-beta protein and cellular material outside and around neurons. Tangles are insoluble twisted fibers that build up inside the nerve cell. Though many older people develop some plaques and tangles, the brains of AD patients have them to a much greater extent and in different brain locations.[47]

Biochemical characteristics

Alzheimer's disease has been identified as a protein misfolding disease, or proteopathy, due to the accumulation of abnormally folded A-beta and tau proteins in the brains of AD patients.[48] Plaques are made of a peptide called beta-amyloid (also A-beta or Aβ), a protein fragment snipped from a larger protein called amyloid precursor protein (APP). APP is a transmembrane protein; which means that it sticks through the neuron's membrane; and is believed to help neurons grow, survive and repair themselves after injury.[49][50] In AD, something causes APP to be divided by enzymes through a mechanism called proteolysis.[51] One of these fragments is beta-amyloid. Beta-amyloid fragments (amyloid fibrils) outside the cell come together into clumps that deposit outside neurons in dense formations known as senile plaques.[52][9]

AD is also considered a tauopathy due to abnormal aggregation of the tau protein. Healthy neurons have an internal support structure, or cytoskeleton, partly made up of structures called microtubules. These microtubules act like tracks, guiding nutrients and molecules from the body of the cell down to the ends of the axon and back. A special kind of protein, tau, makes the microtubules stable through a process named phosphorylation and is therefore called a microtubule-associated protein.[53] In AD, tau is changed chemically, becoming hyperphosphorylated. Hyperphosphorylated tau begins to pair with other threads of tau and they become tangled up together inside nerve cell bodies in masses known as neurofibrillary tangles.[54] When this happens, the microtubules disintegrate, collapsing the neuron's transport system. This may result first in malfunctions in communication between neurons and later in the death of the cells.[55]

Disease mechanism

Three major competing hypotheses exist to explain the cause of the disease. The oldest, on which most currently available drug therapies are based, is known as the cholinergic hypothesis and suggests that AD is due to reduced biosynthesis of the neurotransmitter acetylcholine. However, the medications that treat acetylcholine deficiency only affect symptoms of the disease and neither halt nor reverse it.[56] The cholinergic hypothesis has not maintained widespread support in the face of this evidence, although cholinergic effects have been proposed to initiate large-scale aggregation,[57] leading to generalized neuroinflammation.[46]

In 1991 the amyloid hypothesis was proposed, [58] while research after 2000 is also centered on tau proteins. The two positions differ with one stating that the tau protein abnormalities initiate the disease cascade, while the other states that amyloid beta (Aβ) deposits are the causative factor in the disease.[59] The tau hypothesis is supported by the long-standing observation that deposition of amyloid plaques does not correlate well with neuron loss,[60] but a majority of researchers support the alternative hypothesis that Aβ is the primary causative agent.[59] The amyloid hypothesis is compelling because the gene for the amyloid beta precursor (APP) is located on chromosome 21, and patients with trisomy 21 (Down Syndrome) who thus have an extra gene copy almost universally exhibit AD-like disorders by 40 years of age.[61][62] The traditional formulation of the amyloid hypothesis points to the cytotoxicity of mature aggregated amyloid fibrils, which are believed to be the toxic form of the protein responsible for disrupting the cell's calcium ion homeostasis and thus inducing apoptosis.[63] It should be noted further that ApoE4, the major genetic risk factor for AD, leads to excess amyloid build-up in the brain before AD symptoms arise. Thus, Aβ deposition precedes clinical AD.[64] Another strong support for the amyloid hypothesis, which looks at Aβ as the common initiating factor for Alzheimer's disease, is that transgenic mice solely expressing a mutant human APP gene develop fibrillar amyloid plaques.[65]

Diagnosis

Dementia is by definition a clinical condition but not an exact diagnosis. Alzheimer's disease is usually diagnosed clinically from the patient history, collateral history from relatives, and clinical observations, based on the presence of characteristic neurological and neuropsychological features and the absence of alternative conditions.[66][67] Advanced medical imaging with CT or MRI are generally used to help to diagnose the subtype of dementia and exclude other cerebral pathology.[68] Neuropsychological evaluation including memory testing and assessment of intellectual functioning can further characterize the dementia.[69] Medical organizations have created diagnostic criteria to ease and standardize the process for practicing physicians. Sometimes the diagnoses can be confirmed or made at postmortem when brain material is available and can be examined histologically and histochemically.[70]

Diagnostic criteria

The diagnostic criteria for Alzheimer of the NINCDS-ADRDA (NINCDS and the ADRDA) are among the most used.[71] These criteria require that the presence of cognitive impairment and a suspected dementia syndrome be confirmed by neuropsychological testing for a clinical diagnosis of possible or probable AD while they need histopathologic confirmation (microscopic examination of brain tissue) for the definitive diagnosis. They have shown good reliability and validity.[72] They specify as well eight cognitive domains that may be impaired in AD (i.e., memory, language, perceptual skills, attention, constructive abilities, orientation, problem solving and functional abilities). Similar to the NINCDS-ADRDA Alzheimer's Criteria are the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV-TR) criteria published by the American Psychiatric Association.[73][74]

Diagnostic tools

File:InterlockingPentagons.png
Neuropsychological screening tests can help in the diagnosis of AD. In them patients have to copy drawings similar to the one shown in the picture, remember words, read or sum.

Neuropsychological screening tests as the Mini mental state examination (MMSE) are widely used to evaluate the cognitive impairments needed for diagnosis, but more comprehensive batteries are necessary for high reliability by this method; especially in the earliest stages of the disease.[75][76] On the other hand neurological examination in early AD will usually be normal, independently of cognitive impairment; but for many of the other dementing disorders is key for diagnosis. Therefore, neurological examination is crucial in the differential diagnosis of Alzheimer and other diseases.[69] In addition, interviews with family members are also utilized in the assessment of the disease. Caregivers can supply important information on the daily living abilities, as well as on the decrease over time of the patient's mental function.[77] This is especially important since a patient with AD is commonly unaware of his or her own deficits (anosognosia).[78] Many times families also have difficulties in the detection of initial dementia symptoms and in adequately communicating them to a physician.[79] Finally supplemental testing provide extra information on some features of the disease or are utilized to rule out other diagnoses. Examples are blood tests, which can identify other causes for dementia different than AD;[69] which rarely may even be reversible;[80] or psychological tests for depression, as depression can both co-occur with AD or on the contrary be at the origin of the patient's cognitive impairment.[81][82]

Increasingly, the functional neuroimaging modalities of single photon emission computed tomography (SPECT) and positron emission tomography (PET) are being used to diagnose Alzheimer's, as they have shown similar ability to diagnose Alzheimer's disease as methods involving mental status examination.[83] Furthermore, the ability of SPECT to differentiate Alzheimer's disease from other possible causes, in a given patient already known to be suffering from dementia, appears to be superior to attempts to differentiate the cause of dementia cause by mental testing and history.[84] Another recent objective marker of the disease is the analysis of cerebrospinal fluid for amyloid beta or tau proteins.[85] Both advances (neuroimaging and cerebrospinal fluid analysis) have led to the proposal of new diagnostic criteria.[71][69]

Prevention

Intellectual activities such as playing chess or regular social interaction have been linked to a reduced risk of AD in epidemiological studies, although no causal relationship has been found.

Different epidemiological studies have shown relationships between certain modifiable factors, such as diet, cardiovascular risk, pharmaceutical products, or intellectual activities among others, and a population's likelihood of developing Alzheimer. Only further research, including clinical trials, will reveal whether, in fact, these factors can help to prevent AD.[86] At present contradictory results in global studies, incapacity to prove causal relationships between risk factors and the disease, and possible secondary effects indicate a lack of specific measures to prevent or delay the onset of AD.[87]

The components of a Mediterranean diet, which include fruit and vegetables, bread, wheat and other cereals, olive oil, fish, and red wine, may all individually or together reduce the risk and course of Alzheimer's disease.[88] Vitamins E, B, and C, or folic acid have appeared to be related to a reduced risk of Alzheimer,[89] but other studies indicate that they do not have any significant effect on the onset or course of the disease, while at the same time may have important secondary effects in conjunction with other therapies. [90] Curcumin in curry has shown some effectiveness in preventing brain damage in mouse models.[91]

Although cardiovascular risk factors, such as hypercholesterolemia, hypertension, diabetes, and smoking, are associated with a higher risk of onset and course of AD,[92][93] statins, which are cholesterol lowering drugs, have not been effective in preventing or improving the course of the disease.[94][95] However long-term usage of non-steroidal anti-inflammatory drug (NSAIDs), is associated with a reduced likelihood of developing AD.[96] Other pharmaceutical therapies such as female hormone replacement therapy are no longer thought to prevent dementia,[97][98] while a 2007 systematic review concluded that there was inconsistent and unconvincing evidence that ginkgo has any positive effect on dementia or cognitive impairment.[99]

Intellectual activities such as playing chess, completing crossword puzzles or regular social interaction, may also delay the onset or reduce the severity of Alzheimer's disease.[100][101] Bilingualism is also related to a later onset of Alzheimer.[102]

Treatment

There is currently no cure for Alzheimer's disease. Currently available medications offer relatively small symptomatic benefit for some patients and some may even slow disease progression but in essence remain palliative care.

Pharmaceutical

3d molecular spacefill of donepezil, an acetylcholinesterase inhibitor used in the treatment of AD.

AS of 2008 only four medications are approved for AD by regulatory agencies as the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMEA) and the Japanese PMDA. Three are acetylcholinesterase inhibitors and the other is memantine, an NMDA receptor antagonist.

A reduction in the activity of the cholinergic neurons in the disease is well known.[103] Acetylcholinesterase inhibitors reduce the rate at which acetylcholine (ACh) is broken down and hence increase the concentration of ACh in the brain, combatting the loss of ACh caused by the death of the cholinergin neurons.[104] Cholinesterase inhibitors currently approved include donepezil (Brand name Aricept), galantamine (Razadyne), and rivastigmine (Marketed as Exelon and Exelon Patch). [105][106][107][108] There is substantial evidence for the efficacy of these medications in mild to moderate Alzheimer’s disease,[109] and some evidence for their use in the advanced stage, being only donepezil approved for this stage.[110] Their use in mild cognitive impairment has not been associated with any delay in the onset of AD.[111] Donepezil and galantamine are taken orally while rivastigmine has oral forms and a once-daily transdermal patch.[112]

Involvement of glutamatergic neuronal excitotoxicity in Alzheimer's disease led to the development and introduction of memantine (Brand names Akatinol, Axura, Ebixa/Abixa, Memox and Namenda). Memantine is a novel NMDA receptor antagonist, and has been shown to be moderately clinically efficacious.[113]

Psychosocial intervention

A specifically designed room for sensory integration therapy, or snoezelen; an emotion-oriented psychosocial intervention for people with dementia.

Psychosocial interventions are used as an adjunct to pharmaceutical treatment and can be classified within behavior, emotion, cognition or stimulation oriented approaches. Research on efficacy is unavailable and rarely specific to the disease, focusing instead on dementia.[114]

Behavioral interventions attempt to identify and reduce the antecedents and consequences of problem behaviors. This approach has not shown success in the overall functioning of patients,[115] but can help to reduce some specific problem behaviors, such as incontinence.[116] There is still a lack of high quality data on the effectiveness of these techniques in other behavior problems such as wandering.[117][118]

Emotion-oriented interventions include reminiscence therapy, validation therapy, supportive psychotherapy, sensory integration or snoezelen, and simulated presence therapy. Supportive psychotherapy has received little or no formal scientific study, but some clinicians find it useful in helping mildly impaired patients adjust to their illness.[114] Reminiscence therapy (RT) involves the discussion of past experiences individually or in group, many times with the aid of photographs, household items, music and sound recordings, or other familiar items from the past. Although there are few quality studies on the effectiveness of RT it may be beneficial for cognition and mood.[119] Simulated presence therapy (SPT) is based on attachment theories and is normally carried out playing a recording with voices of the closests relatives of the patient. There is preliminary evidence indicating that SPT may reduce anxiety and challenging behaviors.[120][121] Finally, validation therapy is based on acceptance of the reality and personal truth of another's experience, while sensory integration is based on exercises aimed to stimulate senses. There is little evidence to support the usefulness of these therapies.[122][123]

The aim of cognition-oriented treatments, which include reality orientation and cognitive retraining is the restoration of cognitive deficits. Reality orientation consists in the presentation of information about time, place or person in order to ease the understanding of the person about its surroundings and his place in them. On the other hand cognitive retraining tries to improve impaired capacities by exercitation of mental abilities. Both have shown some efficacy improving cognitive capacities,[124][125] although in some works these effects were transient and negative effects, such as frustation, have also been reported.[114]

Stimulation-oriented treatments include art, music and pet therapies, exercise, and any other kind of recreational activities for patients. Stimulation has modest support for improving behavior, mood, and, to a lesser extent, function. Nevertheless, as important as these effects are, the main support for the use of stimulation therapies is the improvement in the patient daily life routine they suppose.[114]

Treatments in clinical development

Several potential treatments for Alzheimer's disease are currently under investigation, including two compounds being studied in phase 3 clinical trials. Tarenflurbil (MPC-7869, formerly R-flubiprofen) is a gamma secretase modulator sometimes called a selective amyloid beta 42 lowering agent. It is believed to reduce the production of the toxic amyloid beta in favor of shorter forms of the peptide.[126]

Vaccines or immunotherapy for Alzheimer's, unlike typical vaccines, would be used to treat diagnosed patients rather than for disease prevention. Ongoing efforts are based on the idea that, by training the immune system to recognize and attack beta-amyloid, the immune system might reverse deposition of amyloid and thus stop the disease. Initial results using this approach in animals were promising, and clinical trials of the drug candidate AN-1792 showed results in 20% of patients. In 2002 it was reported that 6% of multi-dosed participants (18 of 300) developed symptoms resembling meningoencephalitis, and the trials were stopped. Participants in the halted trials continued to be followed, and 20% "developed high levels of antibodies to beta-amyloid" and some showed slower progression of the disease, maintaining memory-test levels while placebo-patients worsened. Micro-cererebral hemorrhages during passive immunisation and meningoencephalitis with active immunisation still remain potent threats to this strategy.[127]

Simvastatin, a statin, stimulates brain vascular endothelial cells to create a beta-amyloid ejector.[128] The use of this statin may be have a causal relationship to decreased development of the disease.[129]

Several other pharmaceuticals are under investigation to treat Alzheimer's disease. A 2006 pilot study showed small but significant improvements in various cognitive rating scales in patients with Alzheimer's disease after treatment with etanercept, Tumor necrosis factor-alpha receptor fusion protein, which binds to tumor necrosis factor-alpha, and decreases its role in inflammation of nervous tissue.[130] A further study, administering to a single AD patient via perispinal infusion, showed rapid and significant improvement in Alzheimer's symptoms.[131] Laboratory studies with cells and animals continually fuel the pipeline of potential treatments. Some currently approved drugs such as statins and thiazolidinediones have also been under investigation for the treatment and prevention of Alzheimer’s.[132]

Caregiving

Since there is no cure for Alzheimer's, caregiving is an essential part of the treatment. Due to the eventual inability for the sufferer to self-care, Alzheimer's has to be carefully care-managed. Home care in the familiar surroundings of home may delay onset of some symptoms and delay or eliminate the need for more professional and costly levels of care.[citation needed] Many family members choose to look after their relative,[12] but two-thirds of nursing home residents have dementias.[133]

Modifications to the living environment and lifestyle of the Alzheimer's patient can improve functional performance and ease caretaker burden. Assessment by an occupational therapist is often indicated. Adherence to simplified routines and labeling of household items to cue the patient can aid with activities of daily living, while placing safety locks on cabinets, doors, and gates and securing hazardous chemicals can prevent accidents and wandering. Changes in routine or environment can trigger or exacerbate agitation, whereas well-lit rooms, adequate rest, and avoidance of excess stimulation all help prevent such episodes.[134][135] Appropriate social and visual stimulation can improve function by increasing awareness and orientation. For instance, boldly colored tableware aids those with severe AD, helping people overcome a diminished sensitivity to visual contrast to increase food and beverage intake.[136]

Prognosis

As the disease progresses the patient will advance from mild cognitive impairment, when the disease has not yet been diagnosed, to mild and advanced stages of dementia, finally reaching a severe stage of dementia.[22] It is important to note there is also an important individual variability in the presentation and development of the symptoms; being sometimes difficult to classify the person in one of the described stages. Once Alzheimer's has been diagnosed, the average life expectancy of patients living with the disease is approximately 7 years, while less than 3% of the patients live more than 14 years.[137][138][139][6]

Social costs

Because the median age of the industrialized world's population is increasing gradually, Alzheimer's is a major public health challenge. Much of the concern about the solvency of governmental social safety nets is founded on estimates of the costs of caring for baby boomers, assuming that they develop Alzheimer's in the same proportions as earlier generations. For this reason, money spent informing the public of available effective prevention methods may yield disproportionate benefits.[140]

Caregiving burden

The role of family caregivers has become more prominent in both reducing the social cost of care and improving the quality of life of the patient. Home-based care also can have economic, emotional, and psychological costs to the patient's family. Although family members in particular often express the desire to care for the sufferer to the end,[11] Alzheimer's disease is known for effecting a high burden on caregivers.[12]

Alzheimer's disease can incur a variety of stresses on the caregivers: typical complaints are stress, depression, and an inability to cope. Reasons for these complaints can include: high-demands on the caregiver's concentration, as Alzheimer's sufferers have a decreasing regard for their own safety (and can wander when unattended, for example); the lack of gratitude received when the sufferer is unaware of the help being given; and the lack of satisfaction when the sufferer's condition does not abate. Alzheimer's sufferers can be verbally and physically aggressive, and can stubbornly refuse to be helped. Aggression in particular can lead to a temptation to retaliate, which can put both the sufferer and carer at risk. It is additionally stressful for caregivers who are friends and family to witness a sufferer lose his or her identity, and eventually be unable to recognise them.[12]

Family caregivers often give up time from work and forego pay to spend 47 hours per week on average with the person with AD. From a 2006 survey of US patients with long term care insurance, direct and indirect costs of caring for an Alzheimer's patient average $77,500 per year.[141]

Epidemiology

Alzheimer's disease is the most frequent type of dementia in the elderly and affects almost half of all patients with dementia. Correspondingly, advancing age is the primary risk factor for the disease. Among people aged 65, 2–3% show signs of the disease, while 25–50% of people aged 85 have symptoms of Alzheimer's and an even greater number have some of the pathological hallmarks of the disease without the characteristic symptoms. Every five years after the age of 65, the probability of having the disease doubles.[142] The share of Alzheimer's patients over the age of 85 is the fastest growing segment of the Alzheimer's disease population in the US, although current estimates suggest the 75–84 population has about the same number of patients as the over 85 population.[143]

The World Health Organization estimates that globally the total disability adjusted life years (DALY) for AD and other dementias exceeded eleven million in 2005, with a projected 3.4% annual increase.[144]

A study in Denmark found that women aged 65 are at significantly higher risk (22 percent) of developing AD by age 95 than their male counterparts (9 percent), while vascular dementias were nearly equal.[145]

Some studies have shown a relationship between Alzheimer's Disease and magnetic field exposure, although the mechanism is unknown.[146][147] Other research does not confirm this link.[148] The role of metals in the disease is also controversial.[149]

History

Auguste D, first described patient with AD.

Although the concept of dementia goes as far back as the ancient Greek and Roman philosophers and physicians,[150] it was in 1901 when Alöis Alzheimer, a German psychiatrist, identified the first case of what became known as Alzheimer's disease. The patient was a 50-year-old woman called Auguste D. Alöis Alzheimer followed her until she died in 1906, when he first reported the case publicly.[151] In the following five years, 11 similar cases were reported in the medical literature, some of them already using the term Alzheimer's disease.[150] The official consideration of the disease as a distinctive entity is attributed to Emil Kraepelin, who included Alzheimer’s disease or presenile dementia as a subtype of senile dementia in the eighth edition of his Textbook of Psychiatry, published in 1910.[152]

For most of the twentieth century, the diagnosis of Alzheimer's disease was reserved for individuals between the ages of 45 and 65 who developed symptoms of dementia. The terminology changed after 1977 when a conference concluded that the clinical and pathological manifestations of presenile and senile dementia were almost identical, although the authors also added that this did not rule out the possibility of different etiologies. This eventually led to the use of Alzheimer's disease independently of onset age of the disease.[153][154] The term senile dementia of the Alzheimer type (SDAT) was used for a time to describe the condition in those over 65, with classical Alzheimer's disease being used for those younger. Eventually, the term Alzheimer's disease was formally adopted in medical nomenclature to describe individuals of all ages with a characteristic common symptom pattern, disease course, and neuropathology.[155]

Cultural references

As a result of the prevalence of the disease, many notable people have contracted it. Well-known examples are former United States President Ronald Reagan and Irish writer Iris Murdoch, both of whom have scientific articles on how their cognitive capacities deteriorated with the disease.[156][157] Other cases include the retired footballer Ferenc Puskas,[158] the former British Prime Minister Harold Wilson,[159] the actress Rita Hayworth,[160] the actor Charlton Heston,[161] etc. The media attention to the cases of these prominent individuals has helped to make society even more aware of the problem of the disease.

Alzheimers has also been portrayed in films such as Iris (2001),[162] (based on John Baley's memoir of his wife Iris Murdoch)[163] The Notebook (2004),[164] Thanmathra (2005),[165] Memories of tomorrow (Ashita no Kioku) (2006),[166] and Away From Her (2006), (based on Alice Munro's short story The Bear Came Over the Mountain), in documentaries, such as Malcolm and Barbara: A Love Story (1999) and Malcolm and Barbara: Love’s Farewell (2007) both featuring Malcolm Pointon,[167] and in television series.

References

  1. ^ Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, Hall K, Hasegawa K, Hendrie H, Huang Y, Jorm A, Mathers C, Menezes PR, Rimmer E, Scazufca M (2005). "Global prevalence of dementia: a Delphi consensus study". Lancet. 366 (9503): 2112–2117. doi:10.1016/S0140-6736(05)67889-0. PMID 16360788.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. ^ a b Waldemar G, Dubois B, Emre M, Georges J, McKeith IG, Rossor M, Scheltens P, Tariska P, Winblad B (2007). "Recommendations for the diagnosis and management of Alzheimer's disease and other disorders associated with dementia: EFNS guideline". European Journal of Neurology. 14 (1): E1–26. doi:10.1111/j.1468-1331.2006.01605.x. PMID 17222085.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  3. ^ Tabert MH, Liu X, Doty RL, Serby M, Zamora D, Pelton GH, Marder K, Albers MW, Stern Y, Devanand DP (2005). "A 10-item smell identification scale related to risk for Alzheimer's disease". Ann. Neurol. 58 (1): 155–60. doi:10.1002/ana.20533. PMID 15984022.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  4. ^ "Understanding Stages and Symptoms of Alzheimer's Disease". National Institute on Aging. 2007-10-26. Retrieved 2008-02-21.
  5. ^ "What is Alzheimer's disease?". www.alzheimers.org.uk. August 2007. Retrieved 2008-02-21.
  6. ^ a b "Alzheimer's Disease Information Page". National Institute of Neurological Disorders and Stroke (NINDS). 2008-02-07. Retrieved 2008-02-12.
  7. ^ "Alzheimer's Diagnosis of AD". Alzheimer's Reearch Trust. Retrieved 2008-02-29.
  8. ^ a b c Tiraboschi P, Hansen LA, Thal LJ, Corey-Bloom J (2004). "The importance of neuritic plaques and tangles to the development and evolution of AD". Neurology. 62 (11): 1984–1989. PMID 15184601.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  9. ^ "The Search for AD Prevention Strategies". National Institute on Aging. 2006-08-29. Retrieved 2008-02-29.
  10. ^ a b O’Donovan ST. "Dementia caregiving burden and breakdown" (PDF). Forum of Consultant Nurses, Midwives and Allied Health Professionals. Retrieved 2008-02-29. {{cite journal}}: Cite journal requires |journal= (help)
  11. ^ a b c d Selwood A, Johnston K, Katona C, Lyketsos C, Livingston G (2007). "Systematic review of the effect of psychological interventions on family caregivers of people with dementia". Journal of Affective Disorders. 101 (1–3): 75–89. doi:10.1016/j.jad.2006.10.025. PMID 17173977.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  12. ^ Preclinical phase
    • Linn RT, Wolf PA, Bachman DL, Knoefel JE, Cobb JL, Belanger AJ, Kaplan EF, D'Agostino RB (1995). "The 'preclinical phase' of probable Alzheimer's disease. A 13-year prospective study of the Framingham cohort". Archives of Neurology. 52 (5): 485–490. PMID 7733843.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Saxton J, Lopez OL, Ratcliff G, Dulberg C, Fried LP, Carlson MC, Newman AB, Kuller L (2004). "Preclinical Alzheimer disease: neuropsychological test performance 1.5 to 8 years prior to onset". Neurology. 63 (12): 2341–2347. PMID 15623697.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Twamley EW, Ropacki SA, Bondi MW (2006). "Neuropsychological and neuroimaging changes in preclinical Alzheimer's disease". J Int Neuropsychol Soc. 12 (5): 707–35. doi:10.1017/S1355617706060863. PMID 16961952.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  13. ^ Perneczky R, Pohl C, Sorg C, Hartmann J, Komossa K, Alexopoulos P, Wagenpfeil S, Kurz A (2006). "Complex activities of daily living in mild cognitive impairment: conceptual and diagnostic issues". Age Ageing. 35 (3): 240–5. doi:10.1093/ageing/afj054. PMID 16513677.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  14. ^ Rapp MA, Reischies FM (2005). "Attention and executive control predict Alzheimer disease in late life: results from the Berlin Aging Study (BASE)". American Journal of Geriatric Psychiatry. 13 (2): 134–141. doi:10.1176/appi.ajgp.13.2.134. PMID 15703322.
  15. ^ Spaan PE, Raaijmakers JG, Jonker C (2003). "Alzheimer's disease versus normal ageing: a review of the efficiency of clinical and experimental memory measures". Journal of Clinical Experimental Neuropsychology. 25 (2): 216–233. PMID 12754679.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  16. ^ a b Craig D, Mirakhur A, Hart DJ, McIlroy SP, Passmore AP (2005). "A cross-sectional study of neuropsychiatric symptoms in 435 patients with Alzheimer's disease". American Journal of Geriatric Psychiatry. 13 (6): 460–468. doi:10.1176/appi.ajgp.13.6.460. PMID 15956265.{{cite journal}}: CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid15956265" was defined multiple times with different content (see the help page).
  17. ^ Robert PH, Berr C, Volteau M, Bertogliati C, Benoit M, Sarazin M, Legrain S, Dubois B (2006). "Apathy in patients with mild cognitive impairment and the risk of developing dementia of Alzheimer's disease: a one-year follow-up study". Clin Neurol Neurosurg. 108 (8): 733–6. doi:10.1016/j.clineuro.2006.02.003. PMID 16567037.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  18. ^ Palmer K, Berger AK, Monastero R, Winblad B, Bäckman L, Fratiglioni L (2007). "Predictors of progression from mild cognitive impairment to Alzheimer disease". Neurology. 68 (19): 1596–1602. doi:10.1212/01.wnl.0000260968.92345.3f. PMID 17485646.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  19. ^ Small BJ, Gagnon E, Robinson B (2007). "Early identification of cognitive deficits: preclinical Alzheimer's disease and mild cognitive impairment". Geriatrics. 62 (4): 19–23. PMID 17408315.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  20. ^ Petersen RC (2007). "The current status of mild cognitive impairment—what do we tell our patients?". Nat Clin Pract Neurol. 3 (2): 60–61. doi:10.1038/ncpneuro0402. PMID 17279076.
  21. ^ a b c d e f Förstl H, Kurz A (1999). "Clinical features of Alzheimer's disease". European Archives of Psychiatry and Clinical Neuroscience. 249 (6): 288–290. PMID 10653284. Cite error: The named reference "pmid10653284" was defined multiple times with different content (see the help page).
  22. ^ Carlesimo GA, Oscar-Berman M (1992). "Memory deficits in Alzheimer's patients: a comprehensive review". Neuropsychology Review. 3 (2): 119–169. PMID 1300219.
  23. ^ Jelicic M, Bonebakker AE, Bonke B (1995). "Implicit memory performance of patients with Alzheimer's disease: a brief review". International Psychogeriatrics. 7 (3): 385–392. PMID 8821346.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  24. ^ a b c Frank EM (1994). "Effect of Alzheimer's disease on communication function". J S C Med Assoc. 90 (9): 417–423. PMID 7967534. Cite error: The named reference "pmid7967534" was defined multiple times with different content (see the help page).
  25. ^ Becker JT, Overman AA (2002). "The semantic memory deficit in Alzheimer's disease". Reviews of Neurology (in Spanish; Castilian). 35 (8): 777–783. PMID 12402233.{{cite journal}}: CS1 maint: unrecognized language (link)
  26. ^ Hodges JR, Patterson K (1995). "Is semantic memory consistently impaired early in the course of Alzheimer's disease? Neuroanatomical and diagnostic implications". Neuropsychologia. 33 (4): 441–459. PMID 7617154.
  27. ^ Benke T (1993). "Two forms of apraxia in Alzheimer's disease". Cortex. 29 (4): 715–725. PMID 8124945.
  28. ^ Forbes KE, Shanks MF, Venneri A (2004). "The evolution of dysgraphia in Alzheimer's disease". Brain Research Bulletin. 63 (1): 19–24. doi:10.1016/j.brainresbull.2003.11.005. PMID 15121235.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  29. ^ Galasko D, Schmitt F, Thomas R, Jin S, Bennett D (2005). "Detailed assessment of activities of daily living in moderate to severe Alzheimer's disease". Journal of the International Neuropsychology Society. 11 (4): 446–53. PMID 16209425.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  30. ^ Mendez MF, Martin RJ, Smyth KA, Whitehouse PJ (1992). "Disturbances of person identification in Alzheimer's disease. A retrospective study". Journal of Nervous and Mental Disease. 180 (2): 94–96. PMID 1737981.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  31. ^ Sartori G, Snitz BE, Sorcinelli L, Daum I (2004). "Remote memory in advanced Alzheimer's disease". Archives of Clinical Neuropsychology. 19 (6): 779–789. doi:10.1016/j.acn.2003.09.007. PMID 15288331.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  32. ^ Tatsch MF, Bottino CM, Azevedo D, Hototian SR, Moscoso MA, Folquitto JC, Scalco AZ, Louzã MR (2006). "Neuropsychiatric symptoms in Alzheimer disease and cognitively impaired, nondemented elderly from a community-based sample in Brazil: prevalence and relationship with dementia severity". American Journal of Geriatric Psychiatry. 14 (5): 438–45. doi:10.1097/01.JGP.0000218218.47279.db. PMID 16670248.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  33. ^ Honig LS, Mayeux R (2001). "Natural history of Alzheimer's disease". Aging (Milano). 13 (3): 171–82. PMID 11442300.
  34. ^ Volicer L, Bass EA, Luther SL (2007). "Agitation and resistiveness to care are two separate behavioral syndromes of dementia". J Am Med Dir Assoc. 2007 Oct;8(8):527-32. 8 (8): 527–32. PMID 17931577.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  35. ^ Gold DP, Reis MF, Markiewicz D, Andres D (1995). "When home caregiving ends: a longitudinal study of outcomes for caregivers of relatives with dementia". Journal of the American Geriatric Society. 43 (1): 10–16. PMID 7806732.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  36. ^ Bär M, Kruse A, Re S (2003). "Situations of emotional significance in residents suffering from dementia". Z Gerontol Geriatr (in Template:De icon). 36 (6): 454–462. doi:10.1007/s00391-003-0191-0. PMID 14685735.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unrecognized language (link)
  37. ^ Souren LE, Franssen EH, Reisberg B (1995). "Contractures and loss of function in patients with Alzheimer's disease". Journal of the American Geriatric Society. 43 (6): 650–655. PMID 7775724.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  38. ^ Berkhout AM, Cools HJ, van Houwelingen HC (1998). "The relationship between difficulties in feeding oneself and loss of weight in nursing-home patients with dementia". Age Ageing. 27 (5): 637–641. PMID 12675103.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  39. ^ Wada H, Nakajoh K, Satoh-Nakagawa T, Suzuki T, Ohrui T, Arai H, Sasaki H (2001). "Risk factors of aspiration pneumonia in Alzheimer's disease patients". Gerontology. 47 (5): 271–276. PMID 11490146.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  40. ^ Gambassi G, Landi F, Lapane KL, Sgadari A, Mor V, Bernabei R (1999). "Predictors of mortality in patients with Alzheimer's disease living in nursing homes". J Neurol Neurosurg Psychiatr. 67 (1): 59–65. PMID 10369823.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  41. ^ Itzhaki RF, Cosby SL, Wozniak MA (2008). "Herpes simplex virus type 1 and Alzheimer's disease: the autophagy connection". J. Neurovirol. 14 (1): 1–4. doi:10.1080/13550280701802543. PMID 18300070.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  42. ^ Carter CJ (2007). "Interactions between the products of the Herpes simplex genome and Alzheimer's disease susceptibility genes: Relevance to pathological-signalling cascades". Neurochem Int. doi:10.1016/j.neuint.2007.11.003. PMID 18164103.
  43. ^ Waring SC, Rosenberg RN (2008). "Genome-wide association studies in Alzheimer disease". Arch. Neurol. 65 (3): 329–34. doi:10.1001/archneur.65.3.329. PMID 18332245.
  44. ^ Li Y, Grupe A, Rowland C; et al. (2008). "Evidence that common variation in NEDD9 is associated with susceptibility to late-onset Alzheimer's and Parkinson's disease". Hum. Mol. Genet. 17 (5): 759–67. doi:10.1093/hmg/ddm348. PMID 18063669. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  45. ^ a b Wenk GL (2003). "Neuropathologic changes in Alzheimer's disease". Journal of Clinical Psychiatry. 64 Suppl 9: 7–10. PMID 12934968.
  46. ^ Bouras C, Hof PR, Giannakopoulos P, Michel JP, Morrison JH (1994). "Regional distribution of neurofibrillary tangles and senile plaques in the cerebral cortex of elderly patients: a quantitative evaluation of a one-year autopsy population from a geriatric hospital". Cerebral Cortex. 4 (2): 138–150. PMID 8038565.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  47. ^ Hashimoto M, Rockenstein E, Crews L, Masliah E (2003). "Role of protein aggregation in mitochondrial dysfunction and neurodegeneration in Alzheimer's and Parkinson's diseases". Neuromolecular Medicine. 4 (1–2): 21–36. doi:10.1385/NMM:4:1-2:21. PMID 14528050.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  48. ^ Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J (2006). "Synapse formation and function is modulated by the amyloid precursor protein". Journal of Neuroscience. 26 (27): 7212–7221. doi:10.1523/JNEUROSCI.1450-06.2006. PMID 16822978.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  49. ^ Turner PR, O'Connor K, Tate WP, Abraham WC (2003). "Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory". Prog. Neurobiology. 70 (1): 1–32. PMID 12927332.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  50. ^ Hooper NM (2005). "Roles of proteolysis and lipid rafts in the processing of the amyloid precursor protein and prion protein". Biochemical Society Transactions. 33 (Pt 2): 335–338. doi:10.1042/BST0330335. PMID 15787600.
  51. ^ Ohnishi S, Takano K (2004). "Amyloid fibrils from the viewpoint of protein folding". Cellular Molecular Life Sciences. 61 (5): 511–524. doi:10.1007/s00018-003-3264-8. PMID 15004691.
  52. ^ Hernández F, Avila J (2007). "Tauopathies". Cellular Molecular Life Sciences. 64 (17): 2219–2233. doi:10.1007/s00018-007-7220-x. PMID 17604998.
  53. ^ Goedert M, Spillantini MG, Crowther RA (1991). "Tau proteins and neurofibrillary degeneration". Brain Pathology. 1 (4): 279–286. PMID 1669718.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  54. ^ Chun W, Johnson GV (2007). "The role of tau phosphorylation and cleavage in neuronal cell death". Frontiers of Bioscience. 12: 733–756. PMID 17127334.
  55. ^ Walker LC, Rosen RF (2006). "Alzheimer therapeutics-what after the cholinesterase inhibitors?". Age Ageing. 35 (4): 332–335. doi:10.1093/ageing/afl009. PMID 16644763.
  56. ^ Shen ZX (2004). "Brain cholinesterases: II. The molecular and cellular basis of Alzheimer's disease". Medical Hypotheses. 63 (2): 308–321. doi:10.1016/j.mehy.2004.02.031. PMID 15236795.
  57. ^ Hardy J, Allsop D (1991). "Amyloid deposition as the central event in the aetiology of Alzheimer's disease". Trends Pharmacol. Sci. 12 (10): 383–8. PMID 1763432.
  58. ^ a b Mudher A, Lovestone S (2002). "Alzheimer's disease-do tauists and baptists finally shake hands?". Trends in Neuroscience. 25 (1): 22–26. PMID 11801334.
  59. ^ Schmitz C, Rutten BP, Pielen A, Schäfer S, Wirths O, Tremp G, Czech C, Blanchard V, Multhaup G, Rezaie P, Korr H, Steinbusch HW, Pradier L, Bayer TA (2004). "Hippocampal neuron loss exceeds amyloid plaque load in a transgenic mouse model of Alzheimer's disease". Am. J. Pathol. 164 (4): 1495–502. PMID 15039236.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  60. ^ Nistor M, Don M, Parekh M, Sarsoza F, Goodus M, Lopez GE, Kawas C, Leverenz J, Doran E, Lott IT, Hill M, Head E (2007). "Alpha- and beta-secretase activity as a function of age and beta-amyloid in Down syndrome and normal brain". Neurobiol. Aging. 28 (10): 1493–506. doi:10.1016/j.neurobiolaging.2006.06.023. PMID 16904243.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  61. ^ Lott IT, Head E (2005). "Alzheimer disease and Down syndrome: factors in pathogenesis". Neurobiology of Aging. 26 (3): 383–389. doi:10.1016/j.neurobiolaging.2004.08.005. PMID 15639317.
  62. ^ Yankner BA, Duffy LK, Kirschner DA (1990). "Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides". Science. 250 (4978): 279–282. PMID 2218531.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  63. ^ Polvikoski T, Sulkava R, Haltia M, Kainulainen K, Vuorio A, Verkkoniemi A, Niinistö L, Halonen P, Kontula K (1995). "Apolipoprotein E, dementia, and cortical deposition of beta-amyloid protein". New England Journal of Medicine. 333 (19): 1242–1247. PMID 7566000.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  64. ^ Beta-amyloid precursor protein
    • Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F (1995). "Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein". Nature. 373 (6514): 523–527. doi:10.1038/373523a0. PMID 7845465.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Masliah E, Sisk A, Mallory M, Mucke L, Schenk D, Games D (1996). "Comparison of neurodegenerative pathology in transgenic mice overexpressing V717F beta-amyloid precursor protein and Alzheimer's disease". Journal of Neuroscience. 16 (18): 5795–5811. PMID 8795633.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G (1996). "Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice". Science. 274 (5284): 99–102. PMID 8810256.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  65. ^ Mendez MF (2006). "The accurate diagnosis of early-onset dementia". International Journal of Psychiatry Medicine. 36 (4): 401–412. PMID 17407994.
  66. ^ Klafki HW, Staufenbiel M, Kornhuber J, Wiltfang J (2006). "Therapeutic approaches to Alzheimer's disease". Brain. 129 (Pt 11): 2840–2855. doi:10.1093/brain/awl280. PMID 17018549.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  67. ^ "Dementia: Quick reference guide" (PDF). National Institute for Health and Clinical Excellence. 2006. ISBN 1-84629-312-X. Retrieved 2008-02-22. {{cite web}}: Unknown parameter |month= ignored (help)
  68. ^ a b c d Waldemar G, Dubois B, Emre M, Georges J, McKeith IG, Rossor M, Scheltens P, Tariska P, Winblad B (2007). "Recommendations for the diagnosis and management of Alzheimer's disease and other disorders associated with dementia: EFNS guideline". European Journal of Neurology. 14 (1): e1–26. doi:10.1111/j.1468-1331.2006.01605.x. PMID 17222085.{{cite journal}}: CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid17222085" was defined multiple times with different content (see the help page).
  69. ^ McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM (1984). "Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease". Neurology. 34 (7): 939–44. PMID 6610841.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  70. ^ a b Dubois B, Feldman HH, Jacova C, Dekosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, Meguro K, O'brien J, Pasquier F, Robert P, Rossor M, Salloway S, Stern Y, Visser PJ, Scheltens P (2007). "Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS-ADRDA criteria". Lancet Neurology. 6 (8): 734–746. doi:10.1016/S1474-4422(07)70178-3. PMID 17616482.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  71. ^ Blacker D, Albert MS, Bassett SS, Go RC, Harrell LE, Folstein MF (1994). "Reliability and validity of NINCDS-ADRDA criteria for Alzheimer's disease. The National Institute of Mental Health Genetics Initiative". Archives of Neurology. 51 (12): 1198–1204. PMID 7986174.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  72. ^ American Psychiatric Association (2000). Diagnostic and Statistical Manual of Mental disorders, 4th Edition Text Revision. Washington DC. {{cite book}}: Cite has empty unknown parameter: |1= (help)CS1 maint: location missing publisher (link)
  73. ^ Ito N (1996). "Clinical aspects of dementia". Hokkaido Igaku Zasshi (in Japanese). 71 (3): 315–320. PMID 8752526.
  74. ^ Tombaugh TN, McIntyre NJ (1992). "The mini-mental state examination: a comprehensive review". J Am Geriatr Soc. 40 (9): 922–935. PMID 1512391.
  75. ^ Pasquier F (1999). "Early diagnosis of dementia: neuropsychology". J. Neurol. 246 (1): 6–15. PMID 9987708.
  76. ^ Harvey PD, Moriarty PJ, Kleinman L, Coyne K, Sadowsky CH, Chen M, Mirski DF (2005). "The validation of a caregiver assessment of dementia: the Dementia Severity Scale". Alzheimer Disease and Associated Disorders. 19 (4): 186–194. PMID 16327345.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  77. ^ Antoine C, Antoine P, Guermonprez P, Frigard B (2004). "Awareness of deficits and anosognosia in Alzheimer's disease". Encephale (in French). 30 (6): 570–577. PMID 15738860.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  78. ^ Cruz VT, Pais J, Teixeira A, Nunes B (2004). "The initial symptoms of Alzheimer disease: caregiver perception". Acta Med Port (in Portuguese). 17 (6): 435–444. PMID 16197855.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  79. ^ Clarfield AM (2003). "The decreasing prevalence of reversible dementias: an updated meta-analysis". Arch. Intern. Med. 163 (18): 2219–29. doi:10.1001/archinte.163.18.2219. PMID 14557220.
  80. ^ Geldmacher DS, Whitehouse PJ (1997). "Differential diagnosis of Alzheimer's disease". Neurology. 48 (5 Suppl 6): S2–9. PMID 9153154.
  81. ^ Potter GG, Steffens DC (2007). "Contribution of depression to cognitive impairment and dementia in older adults". Neurologist. 13 (3): 105–117. doi:10.1097/01.nrl.0000252947.15389.a9. PMID 17495754.
  82. ^ Bonte FJ, Harris TS, Hynan LS, Bigio EH, White CL (2006). "Tc-99m HMPAO SPECT in the differential diagnosis of the dementias with histopathologic confirmation". Clinical nuclear medicine. 31 (7): 376–378. doi:10.1097/01.rlu.0000222736.81365.63. PMID 16785801.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  83. ^ Dougall NJ, Bruggink S, Ebmeier KP (2004). "Systematic review of the diagnostic accuracy of 99mTc-HMPAO-SPECT in dementia". American Journal of Geriatric Psychiatry. 12 (6): 554–570. doi:10.1176/appi.ajgp.12.6.554. PMID 15545324.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  84. ^ Marksteiner J, Hinterhuber H, Humpel C (2007). "Cerebrospinal fluid biomarkers for diagnosis of Alzheimer's disease: beta-amyloid(1-42), tau, phospho-tau-181 and total protein". Drugs Today. 43 (6): 423–431. doi:10.1358/dot.2007.43.6.1067341. PMID 17612711.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  85. ^ Szekely CA, Breitner JC, Zandi PP (2007). "Prevention of Alzheimer's disease". Int Rev Psychiatry. 19 (6): 693–706. doi:10.1080/09540260701797944. PMID 18092245.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  86. ^ Prevention recommendations not supported:
    • Kawas CH (2006). "Medications and diet: protective factors for AD?". Alzheimer Dis Assoc Disord. 20 (3 Suppl 2): S89–96. PMID 16917203.
    • Luchsinger JA, Mayeux R (2004). "Dietary factors and Alzheimer's disease". Lancet Neurol. 3 (10): 579–87. doi:10.1016/S1474-4422(04)00878-6. PMID 15380154.
    • Luchsinger JA, Noble JM, Scarmeas N (2007). "Diet and Alzheimer's disease". Curr Neurol Neurosci Rep. 7 (5): 366–72. PMID 17764625.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  87. ^ Mediterranean diet:
  88. ^ Vitamins:
    • Morris MC, Schneider JA, Tangney CC (2006). "Thoughts on B-vitamins and dementia". J. Alzheimers Dis. 9 (4): 429–33. PMID 16917152.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Alzheimer's Association. "Vitamin E".
    • Landmark K (2006). "[Could intake of vitamins C and E inhibit development of Alzheimer dementia?]". Tidsskr. Nor. Laegeforen. (in Norwegian). 126 (2): 159–61. PMID 16415937.
    • Luchsinger JA, Tang MX, Miller J, Green R, Mayeux R (2007). "Relation of higher folate intake to lower risk of Alzheimer disease in the elderly". Arch. Neurol. 64 (1): 86–92. doi:10.1001/archneur.64.1.86. PMID 17210813.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  89. ^ Vitamins only of secondary benefit:
    • Morris MC, Evans DA, Schneider JA, Tangney CC, Bienias JL, Aggarwal NT (2006). "Dietary folate and vitamins B-12 and B-6 not associated with incident Alzheimer's disease". J. Alzheimers Dis. 9 (4): 435–43. PMID 16917153.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Malouf M, Grimley EJ, Areosa SA (2003). "Folic acid with or without vitamin B12 for cognition and dementia". Cochrane Database Syst Rev (4): CD004514. doi:10.1002/14651858.CD004514. PMID 14584018.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Sun Y, Lu CJ, Chien KL, Chen ST, Chen RC (2007). "Efficacy of multivitamin supplementation containing vitamins B6 and B12 and folic acid as adjunctive treatment with a cholinesterase inhibitor in Alzheimer's disease: a 26-week, randomized, double-blind, placebo-controlled study in Taiwanese patients". Clin Ther. 29 (10): 2204–14. doi:10.1016/j.clinthera.2007.10.012. PMID 18042476.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Boothby LA, Doering PL (2005). "Vitamin C and vitamin E for Alzheimer's disease". Ann Pharmacother. 39 (12): 2073–80. doi:10.1345/aph.1E495. PMID 16227450.
    • Gray SL, Anderson ML, Crane PK, Breitner JC, McCormick W, Bowen JD, Teri L, Larson E (2008). "Antioxidant vitamin supplement use and risk of dementia or Alzheimer's disease in older adults". J Am Geriatr Soc. 56 (2): 291–295. doi:10.1111/j.1532-5415.2007.01531.x. PMID 18047492.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  90. ^ Curcumin in diet:
    • Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ (2007). "Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model". Journal of Neurochemistry. 102 (4): 1095–1104. doi:10.1111/j.1471-4159.2007.04613.x. PMID 17472706.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM (2001). "The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse". Journal of Neuroscience. 21 (21): 8370–8377. PMID 11606625.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  91. ^ Rosendorff C, Beeri MS, Silverman JM (2007). "Cardiovascular risk factors for Alzheimer's disease". Am J Geriatr Cardiol. 16 (3): 143–9. PMID 17483665.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  92. ^ Gustafson D, Rothenberg E, Blennow K, Steen B, Skoog I (2003). "An 18-year follow-up of overweight and risk of Alzheimer disease". Arch. Intern. Med. 163 (13): 1524–8. doi:10.1001/archinte.163.13.1524. PMID 12860573.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  93. ^ Reiss AB, Wirkowski E (2007). "Role of HMG-CoA reductase inhibitors in neurological disorders : progress to date". Drugs. 67 (15): 2111–2120. PMID 17927279.
  94. ^ Kuller LH (2007). "Statins and dementia". Current Atherosclerosis Reports. 9 (2): 154–161. PMID 17877925.
  95. ^ Szekely CA, Breitner JC, Fitzpatrick AL, Rea TD, Psaty BM, Kuller LH, Zandi PP (2008). "NSAID use and dementia risk in the Cardiovascular Health Study: role of APOE and NSAID type". Neurology. 70 (1): 17–24. doi:10.1212/01.wnl.0000284596.95156.48. PMID 18003940.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  96. ^ Craig MC, Murphy DG (2007). "Estrogen: effects on normal brain function and neuropsychiatric disorders". Climacteric. 10 Suppl 2: 97–104. doi:10.1080/13697130701598746. PMID 17882683.
  97. ^ Mori K, Takeda M (2007). "Hormone replacement Up-to-date. Hormone replacement therapy and brain function". Clin Calcium (in Japanese). 17 (9): 1349–54. doi:CliCa070913491354. PMID 17767023. {{cite journal}}: Check |doi= value (help)
  98. ^ Birks J, Grimley Evans J (2007). "Ginkgo biloba for cognitive impairment and dementia". Cochrane Database Syst Rev (2): CD003120. doi:10.1002/14651858.CD003120.pub2. PMID 17443523. Retrieved 2008-02-22.
  99. ^ Verghese J, Lipton R, Katz M, Hall C, Derby C, Kuslansky G, Ambrose A, Sliwinski M, Buschke H (2003). "Leisure activities and the risk of dementia in the elderly". N Engl J Med. 348 (25): 2508–16. PMID 12815136.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  100. ^ Bennett DA, Schneider JA, Tang Y, Arnold SE, Wilson RS (2006). "The effect of social networks on the relation between Alzheimer's disease pathology and level of cognitive function in old people: a longitudinal cohort study". Lancet Neurol. 5 (5): 406–12. doi:10.1016/S1474-4422(06)70417-3. PMID 16632311.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  101. ^ Bialystok E, Craik FIM, Freedman M (2007). "Bilingualism as a protection against the onset of symptoms of dementia". Neuropsychologia. 42 (2): 459–464. doi:10.1016/j.neuropsychologia.2006.10.009.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  102. ^ Geula C, Mesulam MM (1995). "Cholinesterases and the pathology of Alzheimer disease". Alzheimer Dis Assoc Disord. 9 Suppl 2: 23–8. PMID 8534419.
  103. ^ Stahl SM (2000). "The new cholinesterase inhibitors for Alzheimer's disease, Part 2: illustrating their mechanisms of action". J Clin Psychiatry. 61 (11): 813–4. PMID 11105732.
  104. ^ . US National Library of Medicine (Medline). 2007-01-08 http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a697032.html. Retrieved 2008-03-20. {{cite web}}: Missing or empty |title= (help); Text "Donepezil" ignored (help)
  105. ^ . US National Library of Medicine (Medline). 2007-01-08 http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a699058.html. Retrieved 2008-03-20. {{cite web}}: Missing or empty |title= (help); Text "Galantamine" ignored (help)
  106. ^ . US National Library of Medicine (Medline). 2007-01-08 http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a602009.html. Retrieved 2008-03-20. {{cite web}}: Missing or empty |title= (help); Text "Rivastigmine" ignored (help)
  107. ^ . US National Library of Medicine (Medline). 2007-01-08 http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a607078.html. Retrieved 2008-03-20. {{cite web}}: Missing or empty |title= (help); Text "Rivastigmine Transdermal" ignored (help)
  108. ^ Birks J (2006). "Cholinesterase inhibitors for Alzheimer's disease". Cochrane Database Syst Rev (1): CD005593. doi:10.1002/14651858.CD005593. PMID 16437532.
  109. ^ Birks J, Harvey RJ (2006). "Donepezil for dementia due to Alzheimer's disease". Cochrane Database Syst Rev (1): CD001190. doi:10.1002/14651858.CD001190.pub2. PMID 16437430.
  110. ^ Raschetti R, Albanese E, Vanacore N, Maggini M (2007). "Cholinesterase inhibitors in mild cognitive impairment: a systematic review of randomised trials". PLoS Med. 4 (11): e338. doi:10.1371/journal.pmed.0040338. PMID 18044984.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  111. ^ Rivastigmine transdermal patch:
  112. ^ Memantine (NMDA receptor antagonist):
    • Shah S, Reichman WE (2006). "Treatment of Alzheimer's disease across the spectrum of severity". Clinical Interventions in Aging. 1 (2): 131–142. PMID 18044110.
    • Areosa Sastre A, McShane R, Sherriff F (2004). "Memantine for dementia". Cochrane Database of Systematic Reviews (online) (4): CD003154. doi:10.1002/14651858.CD003154.pub2. PMID 15495043.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Daiello LA (2007). "Current issues in dementia pharmacotherapy". American Journal of Managed Care. 13 Suppl 8: S198–202. PMID 18095783.
    • "Namenda Prescribing Information" (PDF). Forest Pharmaceuticals. Retrieved 2008-02-19.
  113. ^ a b c d "Practice Guideline for the Treatment of Patients with Alzheimer's disease and Other Dementias" (PDF). American Psychiatric Association. October 2007. doi:10.1176/appi.books.9780890423967.152139. Retrieved 2007-12-28. Cite error: The named reference "pracGuideAPA" was defined multiple times with different content (see the help page).
  114. ^ Bottino CM, Carvalho IA, Alvarez AM; et al. (2005). "Cognitive rehabilitation combined with drug treatment in Alzheimer's disease patients: a pilot study". Clin Rehabil. 19 (8): 861–9. PMID 16323385. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  115. ^ Doody RS, Stevens JC, Beck C; et al. (2001). "Practice parameter: management of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology". Neurology. 56 (9): 1154–66. PMID 11342679. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  116. ^ Hermans DG, Htay UH, McShane R (2007). "Non-pharmacological interventions for wandering of people with dementia in the domestic setting". Cochrane Database Syst Rev (1): CD005994. doi:10.1002/14651858.CD005994.pub2. PMID 17253573.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  117. ^ Robinson L, Hutchings D, Dickinson HO; et al. (2007). "Effectiveness and acceptability of non-pharmacological interventions to reduce wandering in dementia: a systematic review". Int J Geriatr Psychiatry. 22 (1): 9–22. doi:10.1002/gps.1643. PMID 17096455. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  118. ^ Woods B, Spector A, Jones C, Orrell M, Davies S (2005). "Reminiscence therapy for dementia". Cochrane Database Syst Rev (2): CD001120. doi:10.1002/14651858.CD001120.pub2. PMID 15846613.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  119. ^ Peak JS, Cheston RI (2002). "Using simulated presence therapy with people with dementia". Aging Ment Health. 6 (1): 77–81. doi:10.1080/13607860120101095. PMID 11827626.
  120. ^ Camberg L, Woods P, Ooi WL; et al. (1999). "Evaluation of Simulated Presence: a personalized approach to enhance well-being in persons with Alzheimer's disease". J Am Geriatr Soc. 47 (4): 446–52. PMID 10203120. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  121. ^ Neal M, Briggs M (2003). "Validation therapy for dementia". Cochrane Database Syst Rev (3): CD001394. doi:10.1002/14651858.CD001394. PMID 12917907.
  122. ^ Chung JC, Lai CK, Chung PM, French HP (2002). "Snoezelen for dementia". Cochrane Database Syst Rev (4): CD003152. PMID 12519587.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  123. ^ Spector A, Orrell M, Davies S, Woods B (2000). "WITHDRAWN: Reality orientation for dementia". Cochrane Database Syst Rev (3): CD001119. doi:10.1002/14651858.CD001119.pub2. PMID 17636652.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  124. ^ Spector A, Thorgrimsen L, Woods B; et al. (2003). "Efficacy of an evidence-based cognitive stimulation therapy programme for people with dementia: randomised controlled trial". Br J Psychiatry. 183: 248–54. PMID 12948999. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  125. ^ Tarenflurbil:
    • Galasko DR, Graff-Radford N, May S, Hendrix S, Cottrell BA, Sagi SA, Mather G, Laughlin M, Zavitz KH, Swabb E, Golde TE, Murphy MP, Koo EH (2007). "Safety, tolerability, pharmacokinetics, and Abeta levels after short-term administration of R-flurbiprofen in healthy elderly individuals". Alzheimer Disease and Associated Disorders. 21 (4): 292–9. doi:10.1097/WAD.0b013e31815d1048. PMID 18090435.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLendon DC, Ozols VV, Jessing KW, Zavitz KH, Koo EH, Golde TE (2003). "NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo". J. Clin. Invest. 112 (3): 440–9. doi:10.1172/JCI200318162. PMID 12897211.{{cite journal}}: CS1 maint: multiple names: authors list (link)
    • Christensen DD (2007). "Alzheimer's disease: progress in the development of anti-amyloid disease-modifying therapies". CNS Spectrum. 12 (2): 113–116, 119–123. PMID 17277711.
  126. ^ Vaccination:
    • Hawkes CA, McLaurin J (2007). "Immunotherapy as treatment for Alzheimer's disease". Expert Reviews of Neurotherapy. 7 (11): 1535–1548. doi:10.1586/14737175.7.11.1535. PMID 17997702.
    • Solomon B (2007). "Clinical immunologic approaches for the treatment of Alzheimer's disease". Expert Opin Investig Drugs. 16 (6): 819–828. doi:10.1517/13543784.16.6.819. PMID 17501694.
    • Woodhouse A, Dickson TC, Vickers JC (2007). "Vaccination strategies for Alzheimer's disease: A new hope?". Drugs Aging. 24 (2): 107–119. PMID 17313199.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  127. ^ Whitfield JF (2007). "The road to LOAD: late-onset Alzheimer's disease and a possible way to block it". Expert Opinion on Theraputic Targets. 11 (10): 1257–1260. doi:10.1517/14728222.11.10.1257. PMID 17907956.
  128. ^ Li G, Larson EB, Sonnen JA, Shofer JB, Petrie EC, Schantz A, Peskind ER, Raskind MA, Breitner JC, Montine TJ (2007). "Statin therapy is associated with reduced neuropathologic changes of Alzheimer disease". Neurology. 69 (9): 878–85. doi:10.1212/01.wnl.0000277657.95487.1c. PMID 17724290.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  129. ^ Tobinick E, Gross H, Weinberger A, Cohen H (2006). "TNF-alpha modulation for treatment of Alzheimer's disease: a 6-month pilot study". MedGenMed. 8 (2): 25. PMID 16926764.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  130. ^ Tobinick EL, Gross H (2008). "Rapid cognitive improvement in Alzheimer's disease following perispinal etanercept administration". Journal of Neuroinflammation. 5: 2. doi:10.1186/1742-2094-5-2. PMID 18184433.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  131. ^ Landreth G (2006). "PPARgamma agonists as new therapeutic agents for the treatment of Alzheimer's disease". Experimental Neurology. 199 (2): 245–248. doi:10.1016/j.expneurol.2006.04.006. PMID 16733054.
  132. ^ "Practice Guideline for the Treatment of Patients with Alzheimer's disease and Other Dementias" (PDF). American Psychiatric Association. October 2007. doi:10.1176/appi.books.9780890423967.152139. Retrieved 2007-12-28.
  133. ^ "Treating behavioral and psychiatric symptoms". Alzheimer's Association. 2006. Retrieved 2006-09-25.
  134. ^ Wenger GC, Burholt V, Scott A (1998). "Dementia and help with household tasks: a comparison of cases and non-cases". Health Place. 4 (1): 33–44. PMID 10671009.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  135. ^ Dunne TE, Neargarder SA, Cipolloni PB, Cronin-Golomb A (2004). "Visual contrast enhances food and liquid intake in advanced Alzheimer's disease". Clinical Nutrition. 23 (4): 533–538. PMID 15297089.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  136. ^ Mölsä PK, Marttila RJ, Rinne UK (1986). "Survival and cause of death in Alzheimer's disease and multi-infarct dementia". Acta Neurologica Scandinavica. 74 (2): 103–107. PMID 3776457.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  137. ^ Mölsä PK, Marttila RJ, Rinne UK (1995). "Long-term survival and predictors of mortality in Alzheimer's disease and multi-infarct dementia". Acta Neurologica Scandinavica. 91 (3): 159–164. PMID 7793228.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  138. ^ Kaldjian LC (2004). "Survival versus prognosis in Alzheimer disease" (PDF). Annals of Internal Medicine. 141 (7): 575, author reply 575. PMID 15466779. Retrieved 2008-02-12.
  139. ^ Sloane PD, Zimmerman S, Suchindran C, Reed P, Wang L, Boustani M, Sudha S (2002). "The public health impact of Alzheimer's disease, 2000–2050: potential implication of treatment advances". Annual Review of Public Health. 23: 213–231. doi:10.1146/annurev.publhealth.23.100901.140525. PMID 11910061.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  140. ^ "The MetLife Study of Alzheimer's Disease: The Caregiving Experience" (PDF). MetLife Mature Market Institute. August 2006. Retrieved 2008-02-12.
  141. ^ Gorelick P (2004). "Risk factors for vascular dementia and Alzheimer disease". Stroke. 35 (11 Suppl 1): 2620–2622. PMID 15375299.
  142. ^ Hebert L, Scherr P, Bienias J, Bennett D, Evans D (2003). "Alzheimer disease in the U.S. population: prevalence estimates using the 2000 census". Archives of Neurology. 60 (8): 1119–1122. PMID 12925369.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  143. ^ "Neurological disorders: Public health challenges" (PDF). World Health Organization.
  144. ^ Andersen K, Launer LJ, Dewey ME, Letenneur L, Ott A, Copeland JR, Dartigues JF, Kragh-Sorensen P, Baldereschi M, Brayne C, Lobo A, Martinez-Lage JM, Stijnen T, Hofman A (1999). "Gender differences in the incidence of AD and vascular dementia: The EURODEM Studies. EURODEM Incidence Research Group". Neurology. 53 (9): 1992–7. PMID 10599770.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  145. ^ Davanipour Z, Tseng CC, Lee PJ, Sobel E (2007). "A case-control study of occupational magnetic field exposure and Alzheimer's disease: results from the California Alzheimer's Disease Diagnosis and Treatment Centers". BMC Neurol. 7: 13. doi:10.1186/1471-2377-7-13. PMID 17559686. Retrieved 2008-02-26.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  146. ^ Qiu C, Fratiglioni L, Karp A, Winblad B, Bellander T (2004). "Occupational exposure to electromagnetic fields and risk of Alzheimer's disease". Epidemiology. 15 (6): 687–94. PMID 15475717.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  147. ^ Seidler A, Geller P, Nienhaus A, Bernhardt T, Ruppe I, Eggert S, Hietanen M, Kauppinen T, Frölich L (2007). "Occupational exposure to low frequency magnetic fields and dementia: a case-control study". Occup Environ Med. 64 (2): 108–14. doi:10.1136/oem.2005.024190. PMID 17043077.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  148. ^ Role of metals:
  149. ^ a b Berchtold NC, Cotman CW (1998). "Evolution in the conceptualization of dementia and Alzheimer's disease: Greco-Roman period to the 1960s". Neurobiology of Aging. 19 (3): 173–189. PMID 9661992.
  150. ^ Auguste D.:
    • Alzheimer Alöis (1907). "Uber eine eigenartige Erkrankung der Hirnrinde" (in Template:De icon). 64 (1–2): 146–148. {{cite journal}}: Cite journal requires |journal= (help)CS1 maint: unrecognized language (link)
    • Alöis Alzheimer (1987). "About a peculiar disease of the cerebral cortex. (Translated by L. Jarvik and H. Greenson)". Alzheimer Disease and Associated Disorders. 1 (1): 3–8. PMID 3331112. {{cite journal}}: Unknown parameter |origdate= ignored (|orig-date= suggested) (help)
    • Maurer Ulrike, Maurer Konrad (2003). Alzheimer: the life of a physician and the career of a disease. New York: Columbia University Press. p. 270. ISBN 0-231-11896-1.
    • Hochberg Fred H., Rottenberg David (1977). Neurological classics in modern translation. New York: Hafner Press. ISBN 0-02-851180-8.
  151. ^ Kraepelin Emil, Diefendorf A. Ross (translated by) (2007-01-17). Clinical Psychiatry: A Textbook For Students And Physicians (Reprint). Kessinger Publishing. p. 568. ISBN 1-4325-0833-4.
  152. ^ Boller F, Forbes MM (1998). "History of dementia and dementia in history: an overview". Journal of Neurological Science. 158 (2): 125–133. PMID 9702682.
  153. ^ Katzman Robert, Terry Robert D, Bick Katherine L (editors) (1978). Alzheimer's disease: senile dementia and related disorders. New York: Raven Press. p. 595. ISBN 0-89004-225-X. {{cite book}}: |author= has generic name (help)CS1 maint: multiple names: authors list (link)
  154. ^ Amaducci LA, Rocca WA, Schoenberg BS (1986). "Origin of the distinction between Alzheimer's disease and senile dementia: how history can clarify nosology". Neurology. 36 (11): 1497–1499. PMID 3531918.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  155. ^ Garrard P, Maloney LM, Hodges JR, Patterson K (2005). "The effects of very early Alzheimer's disease on the characteristics of writing by a renowned author". Brain. 128 (Pt 2): 250–260. doi:10.1093/brain/awh341. PMID 15574466.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  156. ^ Sherman FT (2004). "Did President Reagan have mild cognitive impairment while in office? Living longer with Alzheimer's Disease". Geriatrics. 59 (9): 11, 15. PMID 15461232.
  157. ^ "Hungary legend Puskas dies at 79". BBC. 2006-11-17. Retrieved 2008-01-25.
  158. ^ "Harold Wilson 1964-70 and 1974-6 Labour". 10 Downing Street. Retrieved 2008-03-08.
  159. ^ "Chicago Rita Hayworth Gala". Alzheimer’s Association. 2007. Retrieved 2008-01-25.
  160. ^ "Charlton Heston has Alzheimer's symptoms". CNN. 2002-08-09. Retrieved 2008-01-25.
  161. ^ "Iris". IMDB. 2002-01-18. Retrieved 2008-01-24.
  162. ^ Baley John. Iris: A Memoir and Elegy for Iris.
  163. ^ "The Notebook". IMDB. Retrieved 2008-02-22. (based on Nicholas Sparks 1996 novel of the same name)
  164. ^ "Thanmathra". Webindia123.com. Retrieved 2008-01-24.
  165. ^ "Ashita no kioku". IMDB. Retrieved 2008-01-24. {{cite web}}: Unknown parameter |originallanguage= ignored (help) (based on Hiroshi Ogiwara's novel of the same name)
  166. ^ Malcolm and Barbara:

Further reading

Template:Link FA