Alzheimer's disease

Jump to navigation Jump to search

Template:DiseaseDisorder infobox Editor-In-Chief: C. Michael Gibson, M.S., M.D. [1]

Please Take Over This Page and Apply to be Editor-In-Chief for this topic: There can be one or more than one Editor-In-Chief. You may also apply to be an Associate Editor-In-Chief of one of the subtopics below. Please mail us [2] to indicate your interest in serving either as an Editor-In-Chief of the entire topic or as an Associate Editor-In-Chief for a subtopic. Please be sure to attach your CV and or biographical sketch.

Overview

Alzheimer's disease (AD), also called Alzheimer disease, Senile Dementia of the Alzheimer Type (SDAT) or simply Alzheimer's, is the most common form of dementia. This incurable, degenerative, and terminal disease was first described by German psychiatrist Alois Alzheimer in 1906. Generally it is diagnosed in people over 65 years of age,[1] although the less-prevalent early-onset Alzheimer's can occur much earlier. An estimated 26.6 million people worldwide had Alzheimer's in 2006; this number may quadruple by 2050.[2]

Although each sufferer experiences Alzheimer's in a unique way, there are many common symptoms.[3] The earliest observable symptoms are often mistakenly thought to be 'age-related' concerns, or manifestations of stress.[4] In the early stages, the most commonly recognised symptom is memory loss, such as difficulty in remembering recently learned facts. When a doctor or physician has been notified, and AD is suspected, the diagnosis is usually confirmed with behavioural assessments and cognitive tests, often followed by a brain scan if available.[5] As the disease advances, symptoms include confusion, irritability and aggression, mood swings, language breakdown, long-term memory loss, and the general withdrawal of the sufferer as their senses decline.[4][6] Gradually, bodily functions are lost, ultimately leading to death.[7] Individual prognosis is difficult to assess, as the duration of the disease varies. AD develops for an indeterminate period of time before becoming fully apparent, and it can progress undiagnosed for years. The mean life expectancy following diagnosis is approximately seven years.[8] Fewer than three percent of individuals live more than fourteen years after diagnosis.[9]

The cause and progression of Alzheimer's disease are not well understood. Research indicates that the disease is associated with plaques and tangles in the brain.[10] Currently-used treatments offer a small symptomatic benefit; no treatments to delay or halt the progression of the disease are as yet available. As of 2008, more than 500 clinical trials were investigating possible treatments for AD, but it is unknown if any of them will prove successful.[11] Many measures have been suggested for the prevention of Alzheimer's disease, but their value is unproven in slowing the course and reducing the severity of the disease. Mental stimulation, exercise, and a balanced diet are often recommended, as both a possible prevention and a sensible way of managing the disease.[12]

Because AD cannot be cured and is degenerative, management of patients is essential. The role of the main caregiver is often taken by the spouse or a close relative.[13] Alzheimer's disease is known for placing a great burden on caregivers; the pressures can be wide-ranging, involving social, psychological, physical, and economic elements of the caregiver's life.[14][15][16] In developed countries, AD is one of the most economically costly diseases to society.[17][18]

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 fulfills clinical criteria of diagnosis.[19] It is not yet clear if these early symptoms affect daily living activities. Recent studies show impairments in the most complex activities.[20] The most noticeable deficit is short-term memory loss and the resultant inability to acquire new information. In addition, subtle executive function problems or semantic memory impairments can also occur.[21][22] Apathy can be observed at this stage, and is the most common and persistent neuropsychiatric symptom throughout the course of the disease.[23][24][25] This stage of the disease has also been termed mild cognitive impairment,[26] 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.[27]

Early dementia

In 1994 United States ex-president R. Reagan informed the country of his AD diagnosis via a hand-written letter. Writing is usually affected in the first stages of the disease.

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.[28] Nevertheless, memory problems do not affect all memory subcapacities equally. Older memories of the patient's life (episodic memory), facts learned (declarative memory), and implicit memory (the memory of the body on how to do things, such as using a fork to eat) are affected to a much lesser degree than the capacities needed to learn new facts or make new memories.[29][30] Language problems are mainly characterised by a shrinking vocabulary and a decreased word fluency which leads to a general impoverishment of oral and written language. The Alzheimer's patient is usually capable of adequately communicating basic ideas.[31][32][33] While performing fine motor tasks such as writing, drawing or dressing, certain visoconstructional difficulties, or apraxia, may be present, which may appear as clumsiness.[34] 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.[28]

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.[28]

Speech 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.[31][35] As time passes, complex motor sequences become less coordinated, costing the patient most of their daily-living abilities.[36] Memory problems worsen, and the person may not recognize close relatives.[37] Long-term memory, which was previously left intact, is now also impaired.[38] At this stage, 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.[23][39] Often urinary incontinence develops.[40] Because of the communication deficit along with delusions, patients often resist when caregivers attempt to provide care.[41] 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.[28][42]

Advanced

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.[31] Nevertheless many patients can receive and return emotional signals long after the loss of verbal language.[43] Although aggressiveness can still be present, extreme apathy and exhaustion are much more common.[28] 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,[44] and to lose the ability to feed oneself,[45] if death from some external cause, such as infection due to pressure ulcers or pneumonia, does not occur first.[46][47]

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.[48][49] 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.[50][51] The SNPs TRPC6 and TRPC4 have been implicated in late onset Alzheimer's disease.[52][53][54]

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.[55]

Pathophysiology

Neuropathology

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 characterised 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.[56]

Both amyloid plaques and neurofibrillary tangles are clearly visible by microscopy in AD brains.[10] 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.[57]

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.[58] Plaques are made of a small peptide (39 to 43 amino acid residues) 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.[59][60] In AD, something causes APP to be divided by enzymes through a mechanism called proteolysis.[61] One of these fragments is beta-amyloid. Beta-amyloid fragments (amyloid fibrils) outside the cell form clumps that deposit outside neurons in dense formations known as senile plaques.[62][10]

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.[63] In AD, tau is changed chemically, becoming hyperphosphorylated.

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.[64] 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,[65] leading to generalised neuroinflammation.[56]

In 1991 the amyloid hypothesis was proposed,[66] 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.[67]

The tau hypothesis is supported by the long-standing observation that deposition of amyloid plaques does not correlate well with neuron loss.[68] In this model, 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.[69] 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.[70]

A majority of researchers support the alternative hypothesis that Aβ is the primary causative agent.[67] 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.[71][72] 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.[73] It is known that some types of inherited AD involve only mutations in the APP gene (although this is not the most common type—others involve genes for "pre-senilin" proteins which process APP and may also have still-unknown functions).[74] However, 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.[75]

If damage from Aβ is the primary initiating cause of AD, the exact mechanism has not been elucidated. 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.[76] It is also known that Aβ selectively builds up in the mitochondria of samples from the brains of humans with AD, and in mitochondria from transgenic mice with APP genes, and in both cases inhibits certain mitochondrial enzyme functions, and a similar decrease in glucose utilization in neurons to the one which is a known characteristic of AD. This process may also lead to the formation of damaging reactive oxygen species, calcium influx, and apoptosis. Mechanisms which involve direct damage from Aβ before it forms fibrils and plaques also address the issue that neuronal damage is not correlated as well with plaques, since in this model it is not the plaques themselves which cause the major damage, but rather the precursor Aβ protein directly, via another mechanism.[77]

Various inflammatory processes and inflammatory cytokines may also have a role in the pathology of Alzheimer's disease. However, these are general markers of tissue damage in any disease, and may also be either secondary causes of tissue damage in AD, or else bystander "marker" effects.[78]

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.[79][80] Advanced medical imaging with CT or MRI, and with SPECT or PET are generally used to help to diagnose the subtype of dementia and exclude other cerebral pathology.[81] Neuropsychological evaluation including memory testing and assessment of intellectual functioning can further characterize the dementia.[4] Medical organizations have created diagnostic criteria to ease and standardize the process for practicing physicians. Sometimes the diagnosis can be confirmed or made at postmortem when brain material is available and can be examined histologically and histochemically.[82]

Diagnostic criteria

The diagnostic criteria for Alzheimer of the NINCDS-ADRDA (National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association) are among the most used.[83] 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.[84] 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.[85][86]

Diagnostic tools

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.[87][88] On the other hand neurological examination in early AD will usually be normal, independent 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.[4] In addition, interviews with family members are also utilised 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.[89] This is especially important since a patient with AD is commonly unaware of his or her own deficits (anosognosia).[90] Many times families also have difficulties in the detection of initial dementia symptoms and in adequately communicating them to a physician.[91] Finally, supplemental testing provides extra information on some features of the disease or are used to rule out other diagnoses. Examples are blood tests, which can identify other causes for dementia different than AD,[4] which rarely may even be reversible;[92] 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.[93][94]

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.[95] Furthermore, the ability of SPECT to differentiate Alzheimer's disease from other possible causes, in a 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.[96] A new technique known as "PiB PET" has been developed for directly and clearly imaging beta-amyloid deposits in vivo using a contrasting tracer that binds selectively to the Abeta deposits.[97][98][99] Another recent objective marker of the disease is the analysis of cerebrospinal fluid for amyloid beta or tau proteins.[100] Both advances (neuroimaging and cerebrospinal fluid analysis) have led to the proposal of new diagnostic criteria.[83][4]

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.

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.[101] Different epidemiological studies have proposed relationships between certain modifiable factors, such as diet, cardiovascular risk, pharmaceutical products, or intellectual activities among others, and a population's likelihood of developing AD. Only further research, including clinical trials, will reveal whether, in fact, these factors can help to prevent AD.[102]

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. There is evidence that frequent and moderate consumption of alcohol (beer, wine or distilled spirits) reduces the risk of the disease,[103] [104] but it is still considered premature to make dietary recommendations on this basis.[105][106] Vitamins E, B, and C, or folic acid have appeared to be related to a reduced risk of AD,[107] but other studies indicate that they do not have any significant effect on the onset or course of the disease, but may have important secondary effects in conjunction with other therapies.[108] Curcumin in curry has shown some effectiveness in preventing brain damage in mouse models.[109]

Although cardiovascular risk factors, such as hypercholesterolemia, hypertension, diabetes, and smoking, are associated with a higher risk of onset and course of AD,[110][111] statins, which are cholesterol lowering drugs, have not been effective in preventing or improving the course of the disease.[112][113] However long-term usage of non-steroidal anti-inflammatory drug (NSAIDs), is associated with a reduced likelihood of developing AD in some individuals.[114][115][116]

Other pharmaceutical therapies such as female hormone replacement therapy are no longer thought to prevent dementia,[117][118] and a 2007 systematic review concluded that there was inconsistent and unconvincing evidence that ginkgo has any positive effect on dementia or cognitive impairment.[119]

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.[120][121] Bilingualism is also related to a later onset of Alzheimer.[122]

Management

There is no known cure for Alzheimer's disease. Available treatments offer relatively small symptomatic benefit but remain palliative in nature. Current treatments can be divided into pharmaceutical, psychosocial and caregiving.

Pharmaceutical

3d molecular spacefill of donepezil, an acetylcholinesterase inhibitor used in the treatment of AD symptoms
Molecular structure of memantine, a medication approved for advanced AD symptoms

Four medications, to treat the cognitive manifestations of AD, are currently approved by regulatory agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMEA). Three are acetylcholinesterase inhibitors and the other is memantine, an NMDA receptor antagonist. No drug has an indication for delaying or halting the progression of the disease.

Because reduction in the activity of the cholinergic neurons in the disease is well known,[123] acetylcholinesterase inhibitors are employed to reduce the rate at which acetylcholine (ACh) is broken down and so to increase the concentration of ACh in the brain, thereby combatting the loss of ACh caused by the death of the cholinergin neurons.[124] Cholinesterase inhibitors currently approved include donepezil (brand name Aricept),[125] galantamine (Razadyne),[126] and rivastigmine (branded as Exelon,[127] and Exelon Patch[128]). There is also evidence for the efficacy of these medications in mild to moderate Alzheimer’s disease,[129] and some evidence for their use in the advanced stage. Only donepezil is approved for treatment of advanced AD dementia.[130] The use of these drugs in mild cognitive impairment has not shown any effect in a delay of the onset of AD.[131] Most common side effects include nausea and vomiting, both of which are linked to cholinergic excess. These side effects arise in approximately ten to twenty percent of users and are mild to moderate in severity. Less common secondary effects include muscle cramps; decreased heart rate (bradycardia), decreased appetite and weight, and increased gastric acid.[132][133][134][135]

Glutamate is a useful excitatory neurotransmitter of the nervous system, although excessive amounts in the brain can lead to cell death through a process called excitotoxicity which consists of the overstimulation of glutamate receptors. Excitotoxicity occurs not only in Alzheimer's disease, but also in other neurological diseases such as Parkinson's disease and multiple sclerosis.[136] Memantine (brand names Akatinol, Axura, Ebixa/Abixa, Memox and Namenda),[137] is a noncompetitive NMDA receptor antagonist first used as an anti-influenza agent. It acts on the glutamatergic system by blocking NMDA glutamate receptors and inhibits their overstimulation by glutamate.[136] Memantine has been shown to be moderately efficacious in the treatment of moderate to severe Alzheimer’s disease. Its effects in the initial stages are unknown.[138] Reported adverse events with memantine are infrequent and mild, including hallucinations, confusion, dizziness, headache and fatigue.[139] Memantine used in combination with donepezil has been shown to be "of statistically significant but clinically marginal effectiveness".[140]

Neuroleptic anti-psychotic drugs commonly given to Alzheimer's patients with behavioural problems are modestly useful in reducing aggression and psychosis, but are associated with serious adverse effects, such as cerebrovascular events, movement difficulties or cognitive decline, that do not permit their routine use.[141][142][143]

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.[144]

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,[145] but can help to reduce some specific problem behaviors, such as incontinence.[146] There is still a lack of high quality data on the effectiveness of these techniques in other behavior problems such as wandering.[147][148]

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.[144] 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.[149] Simulated presence therapy (SPT) is based on attachment theories and is normally carried out playing a recording with voices of the closest relatives of the patient. There is preliminary evidence indicating that SPT may reduce anxiety and challenging behaviors.[150][151] 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.[152][153]

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,[154][155] although in some works these effects were transient and negative effects, such as frustration, have also been reported.[144]

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.[144]

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.[156] Many family members choose to look after their relative,[157] but two-thirds of nursing home residents have dementias.[158]

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.[159][160] 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.[161]

Clinical research

As of 2008, the safety and efficacy of more than 400 pharmaceutical treatments are being investigated in clinical trials worldwide, and approximately one-fourth of these compounds are in Phase III trials, which is the last step prior to review by regulatory agencies.[162] It is unknown as to whether any of these trials will ultimately prove successful in treating the disease.

A critical area of clinical research is focused on treating the underlying disease pathology. Reduction of amyloid beta levels is a common target of compounds under investigation. Immunotherapy or vaccination for the amyloid protein is one treatment modality under study. Unlike vaccines which seek to prevent disease, this therapy would be used to treat diagnosed patients, and is based upon the concept of training the immune system to recognize, attack, and reverse deposition of amyloid, thereby altering the course of the disease.[163] An example of such a vaccine under investigation is ACC-001.[164][165] Similar agents are bapineuzumab, an antibody designed as identical to the naturally-induced anti-amyloid antibody,[166] and MPC-7869, a selective amyloid beta-42 lowering agent.[167] Other approaches are neuroprotective agents, such as AL-108,[168] metal-protein interaction attenuation agents, such as PBT2,[169] or tumor necrosis factor-alpha receptor fusion proteins, such as etanercept.[170][171][172] There are also many basic investigations attempting to increase the knowledge on the origin and mechanisms of the disease that may lead to new treatments.

Prognosis

The early stages of Alzheimer's disease are difficult to diagnose. A definitive diagnosis is usually made once cognitive impairment compromises daily living activities, although the person may still be living independently. He will progress from mild cognitive problems, such as memory loss through increasing stages of cognitive and non-cognitive disturbances, eliminating any possibility of independent living.[28]

Life expectancy of the population with the disease is reduced.[8][173][174] The mean life expectancy following diagnosis is approximately seven years.[8] Fewer than 3% of patients live more than fourteen years.[9] Disease features significantly associated with reduced survival are an increased severity of cognitive impairment, decreased functional level, history of falls, and disturbances in the neurological examination. Other coincident diseases such as heart problems, diabetes or history of alcohol abuse are also related with shortened survival.[173][175][176] While the earlier the age at onset the higher the total survival years, life expectancy is particularly reduced when compared to the healthy population among those who are younger.[174] Men have a less favourable survival prognosis than women.[9][177]

The disease is the underlying cause of death in 70% of all cases.[8] Pneumonia and dehydration are the most frequent immediate causes of death, while cancer is a less frequent cause of death than in the general population.[8][177]

Epidemiology

AD incidence rates
after 65 years of age[178]
Age Incidence
(new affected)
per thousand
person–years
65–69  3
70–74  6
75–79  9
80–84 23
85–89 40
90–   69

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. [179] [180]

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.[181] 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 (nine percent), while vascular dementias were nearly equal.[182]

Some studies have shown a relationship between risk of developing AD and lifetime magnetic field exposure, although the mechanism is unknown.[183][184] Other research does not confirm this link.[185] The role of metals in the disease is also controversial.[186]

Two main measures are used in epidemiological studies: incidence and prevalence. Incidence is the number of new cases per unit of person–time at risk (usually number of new cases per thousand person–years); while prevalence is the total number of cases of the disease in the population at a given time.

Regarding incidence, cohort longitudinal studies (studies where a disease-free population is followed over the years) provide rates between 10–15 per thousand person–years for all dementias and 5–8 for AD,[178][187] which means that half of new dementia cases each year are AD. Advancing age is a primary risk factor for the disease and incidence rates are not equal for all ages: every five years after the age of 65, the risk of acquiring the disease approximately doubles, increasing from 3 to as much as 69 per thousand person years.[178][187] There are also sex differences in the incidence rates, women having a higher risk of developing AD particularly in the population older than 85.[187][188]

Prevalence of AD in populations is dependent upon different factors including incidence and survival. Since the incidence of AD increases with age, it is particularly important to include the mean age of the population of interest. In the United States, Alzheimer prevalence was estimated to be 1.6% in the year 2000 both overall and in the 65–74 age group, with the rate increasing to 19% in the 75–84 group and to 42% in the greater than 84 group.[189] Prevalence rates in less developed regions are lower.[190] The World Health Organization estimated that in 2005, 0.379% of people worldwide had dementia, and that the prevalence would increase to 0.441% in 2015 and to 0.556% in 2030.[191] Other studies have reached similar conclusions.[190] Another study estimated that in 2006, 0.40% of the world population (range 0.17–0.89%; absolute number 26.6 million, range 11.4–59.4 million) were afflicted by AD, and that the prevalence rate would triple and the absolute number would quadruple by the year 2050.[2]

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,[192] it was in 1901 when Alöis Alzheimer, a German psychiatrist, identified the first case of what became known as Alzheimer's disease in a fifty-year-old woman he called Auguste D. Alöis Alzheimer followed her until she died in 1906, when he first reported the case publicly.[193] In the following five years, eleven similar cases were reported in the medical literature, some of them already using the term Alzheimer's disease.[192] 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.[194]

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.[195][196] 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.[197]

Society and culture

Social costs

Because the median age of the industrialised world's population is gradually increasing, 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.[198]

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,[199] Alzheimer's disease is known for effecting a high burden on caregivers.[157]

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.[157]

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.[200]


References

  1. Brookmeyer R, Gray S, Kawas C (1998). "Projections of Alzheimer's disease in the United States and the public health impact of delaying disease onset". Am J Public Health. 88 (9): 1337–42. PMC 1509089. PMID 9736873. Unknown parameter |month= ignored (help)
  2. 2.0 2.1 2006 prevalence estimate:
  3. "What is Alzheimer's disease?". Alzheimers.org.uk. 2007. Retrieved 2008-02-21. Unknown parameter |month= ignored (help)
  4. 4.0 4.1 4.2 4.3 4.4 4.5 Waldemar G, Dubois B, Emre M; et al. (2007). "Recommendations for the diagnosis and management of Alzheimer's disease and other disorders associated with dementia: EFNS guideline". Eur. J. Neurol. 14 (1): e1–26. doi:10.1111/j.1468-1331.2006.01605.x. PMID 17222085. Unknown parameter |month= ignored (help)
  5. "Alzheimer's diagnosis of AD". Alzheimer's Research Trust. Retrieved 2008-02-29.
  6. 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): 155–160. doi:10.1002/ana.20533. PMID 15984022.
  7. "Understanding stages and symptoms of Alzheimer's disease". National Institute on Aging. 2007-10-26. Retrieved 2008-02-21.
  8. 8.0 8.1 8.2 8.3 8.4 Mölsä PK, Marttila RJ, Rinne UK (1986). "Survival and cause of death in Alzheimer's disease and multi-infarct dementia". Acta Neurol. Scand. 74 (2): 103–7. PMID 3776457. Unknown parameter |month= ignored (help); |access-date= requires |url= (help)
  9. 9.0 9.1 9.2 Mölsä PK, Marttila RJ, Rinne UK (1995). "Long-term survival and predictors of mortality in Alzheimer's disease and multi-infarct dementia". Acta Neurol. Scand. 91 (3): 159–64. PMID 7793228. Unknown parameter |month= ignored (help)
  10. 10.0 10.1 10.2 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.
  11. "Alzheimer's Disease Clinical Trials". US National Institutes of Health. Retrieved 2008-08-18.
  12. "Can Alzheimer's disease be prevented" (pdf). National Institute on Aging. 2006-08-29. Retrieved 2008-02-29.
  13. "The MetLife study of Alzheimer's disease: The caregiving experience" (PDF). MetLife Mature Market Institute. 2006. Retrieved 2008-02-12. Unknown parameter |month= ignored (help)
  14. Thompson CA, Spilsbury K, Hall J, Birks Y, Barnes C, Adamson J (2007). "Systematic review of information and support interventions for caregivers of people with dementia". BMC Geriatr. 7: 18. doi:10.1186/1471-2318-7-18. PMC 1951962. PMID 17662119.
  15. Schneider J, Murray J, Banerjee S, Mann A (1999). "EUROCARE: a cross-national study of co-resident spouse carers for people with Alzheimer's disease: I—Factors associated with carer burden". International Journal of Geriatric Psychiatry. 14 (8): 651–661. doi:10.1002/(SICI)1099-1166(199908)14:8<651::AID-GPS992>3.0.CO;2-B. PMID 10489656. Unknown parameter |month= ignored (help); |access-date= requires |url= (help)
  16. Murray J, Schneider J, Banerjee S, Mann A (1999). "EUROCARE: a cross-national study of co-resident spouse carers for people with Alzheimer's disease: II--A qualitative analysis of the experience of caregiving". International Journal of Geriatric Psychiatry. 14 (8): 662–667. doi:10.1002/(SICI)1099-1166(199908)14:8<662::AID-GPS993>3.0.CO;2-4. PMID 10489657. Unknown parameter |month= ignored (help)
  17. Bonin-Guillaume S, Zekry D, Giacobini E, Gold G, Michel JP (2005). "Impact économique de la démence (English: The economical impact of dementia)". Presse Med (in French). 34 (1): 35–41. ISSN 0755-4982. PMID 15685097. Unknown parameter |month= ignored (help)
  18. Meek PD, McKeithan K, Schumock GT (1998). "Economic considerations in Alzheimer's disease". Pharmacotherapy. 18 (2 Pt 2): 68–73, discussion 79–82. PMID 9543467.
  19. 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.
    • 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.
    • Twamley EW, Ropacki SA, Bondi MW (2006). "Neuropsychological and neuroimaging changes in preclinical Alzheimer's disease". J Int Neuropsychol Soc. 12 (5): 707–735. doi:10.1017/S1355617706060863. PMID 16961952.
  20. 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): 240–245. doi:10.1093/ageing/afj054. PMID 16513677.
  21. 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.
  22. 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.
  23. 23.0 23.1 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.
  24. 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): 733–736. doi:10.1016/j.clineuro.2006.02.003. PMID 16567037.
  25. 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.
  26. 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.
  27. 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.
  28. 28.0 28.1 28.2 28.3 28.4 28.5 Förstl H, Kurz A (1999). "Clinical features of Alzheimer's disease". European Archives of Psychiatry and Clinical Neuroscience. 249 (6): 288–290. PMID 10653284.
  29. Carlesimo GA, Oscar-Berman M (1992). "Memory deficits in Alzheimer's patients: a comprehensive review". Neuropsychology Review. 3 (2): 119–169. doi:10.1007/BF01108841. PMID 1300219.
  30. 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. doi:10.1017/S1041610295002134. PMID 8821346.
  31. 31.0 31.1 31.2 Frank EM (1994). "Effect of Alzheimer's disease on communication function". J S C Med Assoc. 90 (9): 417–423. PMID 7967534.
  32. 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.
  33. 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.
  34. Benke T (1993). "Two forms of apraxia in Alzheimer's disease". Cortex. 29 (4): 715–725. PMID 8124945.
  35. 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.
  36. 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): 446–453. PMID 16209425.
  37. 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.
  38. 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.
  39. 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.
  40. Honig LS, Mayeux R (2001). "Natural history of Alzheimer's disease". Aging (Milano). 13 (3): 171–182. PMID 11442300.
  41. 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): 527–532. PMID 17931577.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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: 920. doi:10.1016/j.neuint.2007.11.003. PMID 18164103.
  50. 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.
  51. 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.
  52. Lessard CB, Lussier MP, Cayouette S, Bourque G, Boulay G. (2005). "The overexpression of presenilin2 and Alzheimer's-disease-linked presenilin2 variants influences TRPC6-enhanced Ca2+ entry into HEK293 cells". Cell Signal. 17: 437–445. doi:10.1016/j.cellsig.2004.09.005.
  53. Poduslo SE, Huang R, Huang J, Smith S (2008). "Genome screen of late-onset Alzheimer's extended pedigrees identifies TRPC4AP by haplotype analysis". Am. J. Med. Genet. B Neuropsychiatr. Genet. doi:10.1002/ajmg.b.30767. PMID 18449908. Unknown parameter |month= ignored (help)
  54. Poduslo SE, Huang R, Huang J, Smith S (2008-04-30 [Epub ahead of print]). "Genome screen of late-onset Alzheimer's extended pedigrees identifies TRPC4AP by haplotype analysis". Am J Med Genet B Neuropsychiatr Genet. PMID 18449908. Check date values in: |date= (help)
  55. Selkoe Dennis J (1999-06-24). "Translating cell biology into therapeutic advances in Alzheimer's disease" (PDF). Nature. 399 (SUPP).
  56. 56.0 56.1 Wenk GL (2003). "Neuropathologic changes in Alzheimer's disease". Journal of Clinical Psychiatry. 64 Suppl 9: 7–10. PMID 12934968.
  57. 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. doi:10.1093/cercor/4.2.138. PMID 8038565.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. Hernández F, Avila J (2007). "Tauopathies". Cellular Molecular Life Sciences. 64 (17): 2219–2233. doi:10.1007/s00018-007-7220-x. PMID 17604998.
  64. 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.
  65. 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.
  66. 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. doi:10.1016/0165-6147(91)90609-V. PMID 1763432.
  67. 67.0 67.1 Mudher A, Lovestone S (2002). "Alzheimer's disease-do tauists and baptists finally shake hands?". Trends in Neuroscience. 25 (1): 22–26. doi:10.1016/S0166-2236(00)02031-2. PMID 11801334.
  68. 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.
  69. Goedert M, Spillantini MG, Crowther RA (1991). "Tau proteins and neurofibrillary degeneration". Brain Pathology. 1 (4): 279–286. doi:10.1111/j.1750-3639.1991.tb00671.x. PMID 1669718.
  70. Chun W, Johnson GV (2007). "The role of tau phosphorylation and cleavage in neuronal cell death". Frontiers of Bioscience. 12: 733–756. PMID 17127334.
  71. 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.
  72. 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.
  73. 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. doi:10.1056/NEJM199511093331902. PMID 7566000.
  74. "Alzheimer disease". US National Library of Medicine. 2008-04-25. Retrieved 2008-05-02.
  75. 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.
    • 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.
    • 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. doi:10.1126/science.274.5284.99. PMID 8810256.
  76. Yankner BA, Duffy LK, Kirschner DA (1990). "Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides". Science. 250 (4978): 279–282. doi:10.1126/science.2218531. PMID 2218531.
  77. Chen, X, Yan, SD (2006). "Mitochondrial Aβ: A Potential Cause of Metabolic Dysfunction in Alzheimer's Disease". IUBMB Life. 58 (12): 686–694. doi:10.1080/15216540601047767. PMID 17424907.
  78. Greig NH, Mattson MP, Perry T, Chan SL, Giordano T, Sambamurti K, Rogers JT, Ovadia H, Lahiri DK (2004). "New therapeutic strategies and drug candidates for neurodegenerative diseases: p53 and TNF-alpha inhibitors, and GLP-1 receptor agonists". Ann N Y Acad Sci. 1035 (Dec): 290–315. doi:10.1196/annals.1332.018. PMID 15681814.
  79. Mendez MF (2006). "The accurate diagnosis of early-onset dementia". International Journal of Psychiatry Medicine. 36 (4): 401–412. PMID 17407994.
  80. 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.
  81. "Dementia: Quick reference guide" (PDF). National Institute for Health and Clinical Excellence. 2006. ISBN 1-84629-312-X. Retrieved 2008-02-22. Unknown parameter |month= ignored (help)
  82. 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.
  83. 83.0 83.1 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.
  84. 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.
  85. American Psychiatric Association (2000). Diagnostic and Statistical Manual of Mental disorders, 4th Edition Text Revision. Washington DC.
  86. Ito N (1996). "Clinical aspects of dementia". Hokkaido Igaku Zasshi (in Japanese). 71 (3): 315–320. PMID 8752526.
  87. Tombaugh TN, McIntyre NJ (1992). "The mini-mental state examination: a comprehensive review". J Am Geriatr Soc. 40 (9): 922–935. PMID 1512391.
  88. Pasquier F (1999). "Early diagnosis of dementia: neuropsychology". J. Neurol. 246 (1): 6–15. PMID 9987708.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. Geldmacher DS, Whitehouse PJ (1997). "Differential diagnosis of Alzheimer's disease". Neurology. 48 (5 Suppl 6): S2–9. PMID 9153154.
  94. 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.
  95. 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.
  96. 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.
  97. Kemppainen NM, Aalto S, Karrasch M, Någren K, Savisto N, Oikonen V, Viitanen M, Parkkola R, Rinne JO (2008). "Cognitive reserve hypothesis: Pittsburgh Compound B and fluorodeoxyglucose positron emission tomography in relation to education in mild Alzheimer's disease". Ann. Neurol. 63 (1): 112–8. doi:10.1002/ana.21212. PMID 18023012.
  98. Ikonomovic MD, Klunk WE, Abrahamson EE, Mathis CA, Price JC, Tsopelas ND, Lopresti BJ, Ziolko S, Bi W, Paljug WR, Debnath ML, Hope CE, Isanski BA, Hamilton RL, Dekosky ST (March 2008). "Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer's disease". Brain. doi:doi:10.1093/brain/awn016 Check |doi= value (help). PMID 18339640.
  99. Jack CR, Lowe VJ, Senjem ML; et al. (2008). "11C PiB and structural MRI provide complementary information in imaging of Alzheimer's disease and amnestic mild cognitive impairment". Brain. 131 (Pt 3): 665–80. doi:10.1093/brain/awm336. PMID 18263627.
  100. 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.
  101. 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. doi:10.1007/s11910-007-0057-8. PMID 17764625.
  102. 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.
  103. Alcohol:
    • Mulkamal KJ; et al. (2003-03-19). "Prospective study of alcohol consumption and risk of dementia in older adults". Journal of the American Medical Association. 289: 1405–1413.
    • Ganguli M; et al. (2005). "Alcohol consumption and cognitive function in late life: A longitudinal community study". Neurology. 65: 1210–1217.
    • Huang W; et al. (2002). "Alcohol consumption and incidence of dementia in a community sample aged 75 years and older". Journal of Clinical Epidemiology. 55 (10): 959–964.
    • Rodgers B; et al. (2005). "Non-linear relationships between cognitive function and alcohol consumption in young, middle-aged and older adults: The PATH Through Life Project". Addiction. 100 (9): 1280–1290.
    • Anstey KJ; et al. (2005). "Lower cognitive test scores observed in alcohol are associated with demographic, personality, and biological factors: The PATH Through Life Project". Addiction. 100 (9): 1291–1301, .Espeland, M.; et al. (2006). "Association between alcohol intake and domain-specific cognitive function in older women". Neuroepidemiology. 1 (27): 1–12.
    • Stampfer MJ; et al. (2005). "Effects of moderate alcohol consumption on cognitive function in women". New England Journal of Medicine. 352: 245–253.
    • Ruitenberg A; et al. (2002). "Alcohol consumption and risk of dementia: the Rotterdam Study". Lancet. 359 (9303): 281–286.
    • Scarmeas N; et al. (2006-04-18). "Mediterranean diet and risk for Alzheimer's disease". Annals of Neurology.
  104. Mediterranean diet:
    • Scarmeas N, Stern Y, Mayeux R, Luchsinger JA (2006). "Mediterranean diet, Alzheimer disease, and vascular mediation". Arch. Neurol. 63 (12): 1709–1717. doi:10.1001/archneur.63.12.noc60109. PMID 17030648.
    • Scarmeas N, Luchsinger JA, Mayeux R, Stern Y (2007). "Mediterranean diet and Alzheimer disease mortality". Neurology. 69 (11): 1084–93. doi:10.1212/01.wnl.0000277320.50685.7c. PMID 17846408.
    • Barberger-Gateau P, Raffaitin C, Letenneur L, Berr C, Tzourio C, Dartigues JF, Alpérovitch A (2007). "Dietary patterns and risk of dementia: the Three-City cohort study". Neurology. 69 (20): 1921–1930. doi:10.1212/01.wnl.0000278116.37320.52. PMID 17998483.
    • Dai Q, Borenstein AR, Wu Y, Jackson JC, Larson EB (2006). "Fruit and vegetable juices and Alzheimer's disease: the Kame Project". American Journal of Medicine. 119 (9): 751–759. doi:10.1016/j.amjmed.2006.03.045. PMID 16945610.
    • Savaskan E, Olivieri G, Meier F, Seifritz E, Wirz-Justice A, Müller-Spahn F (2003). "Red wine ingredient resveratrol protects from beta-amyloid neurotoxicity". Gerontology. 49 (6): 380–383. doi:10.1159/000073766. PMID 14624067.
  105. Luchsinger JA, Mayeux R (2004 Oct). "Dietary factors and Alzheimer's disease". Lancet Neurology. 3 (10): 579–587. PMID 15380154. Available data do not permit definitive conclusions regarding diet and AD or specific recommendations on diet modification for the prevention of AD. Check date values in: |date= (help)
  106. Kawas CH (2006 Jul-Sep). "Medications and diet: protective factors for AD?". Alzheimer Dis Assoc Disord. 20 (3 Suppl 2): S89–96. PMID 16917203. Evidence regarding dietary and supplemental intake of vitamins E, C, and folate, and studies of alcohol and wine intake are also reviewed. At present, there is insufficient evidence to make public health recommendations, but these studies can provide potentially important clues and new avenues for clinical and laboratory research. Check date values in: |date= (help)
  107. Vitamins:
    • Morris MC, Schneider JA, Tangney CC (2006). "Thoughts on B-vitamins and dementia". J. Alzheimers Dis. 9 (4): 429–33. PMID 16917152.
    • 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.
  108. 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.
    • 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.
    • 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, randomised, double-blind, placebo-controlled study in Taiwanese patients". Clin Ther. 29 (10): 2204–14. doi:10.1016/j.clinthera.2007.10.012. PMID 18042476.
    • 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.
  109. 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.
    • 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.
  110. Rosendorff C, Beeri MS, Silverman JM (2007). "Cardiovascular risk factors for Alzheimer's disease". Am J Geriatr Cardiol. 16 (3): 143–9. doi:10.1111/j.1076-7460.2007.06696.x. PMID 17483665.
  111. 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): 1524–1528. doi:10.1001/archinte.163.13.1524. PMID 12860573.
  112. Reiss AB, Wirkowski E (2007). "Role of HMG-CoA reductase inhibitors in neurological disorders : progress to date". Drugs. 67 (15): 2111–2120. PMID 17927279.
  113. Kuller LH (2007). "Statins and dementia". Current Atherosclerosis Reports. 9 (2): 154–161. doi:10.1007/s11883-007-0012-9. PMID 17877925.
  114. 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.
  115. "Long-term use of ibuprofen may reduce the risk of developing Alzheimer's disease, a large US study reports". BBC. 2008-05-05.
  116. "Ibuprofen Linked to Reduced Alzheimer's Risk". Washington Post. 2008-05-05.
  117. 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.
  118. Mori K, Takeda M (2007). "Hormone replacement Up-to-date. Hormone replacement therapy and brain function". Clin Calcium (in Japanese). 17 (9): 1349–1354. doi:CliCa070913491354 Check |doi= value (help). PMID 17767023.
  119. 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.
  120. 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): 2508–2516. doi:10.1056/NEJMoa022252. PMID 12815136.
  121. 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): 406–412. doi:10.1016/S1474-4422(06)70417-3. PMID 16632311.
  122. 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.
  123. Geula C, Mesulam MM (1995). "Cholinesterases and the pathology of Alzheimer disease". Alzheimer Dis Assoc Disord. 9 Suppl 2: 23–8. PMID 8534419.
  124. Stahl SM (2000). "The new cholinesterase inhibitors for Alzheimer's disease, Part 2: illustrating their mechanisms of action". J Clin Psychiatry. 61 (11): 813–814. PMID 11105732.
  125. "Donepezil". US National Library of Medicine (Medline). 2007-01-08. Retrieved 2008-03-20.
  126. "Galantamine". US National Library of Medicine (Medline). 2007-01-08. Retrieved 2008-03-20.
  127. "Rivastigmine". US National Library of Medicine (Medline). 2007-01-08. Retrieved 2008-03-20.
  128. "Rivastigmine Transdermal". US National Library of Medicine (Medline). 2007-01-08. Retrieved 2008-03-20.
  129. Birks J (2006). "Cholinesterase inhibitors for Alzheimer's disease". Cochrane Database Syst Rev (1): CD005593. doi:10.1002/14651858.CD005593. PMID 16437532.
  130. 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.
  131. 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.
  132. "Aricept and Aricept ODT Product Insert" (PDF). Eisai and Pfizer. Retrieved 2008-01-30.
  133. "Razadyne ER U.S. Full Prescribing Information" (PDF). Ortho-McNeil Neurologics. Retrieved 2008-02-19.
  134. "Exelon ER U.S. Prescribing Information" (PDF). Novartis Pharmaceuticals. Retrieved 2008-02-19.
  135. "Exelon U.S. Prescribing Information" (PDF). Novartis Pharmaceuticals. Retrieved 2008-02-21.
  136. 136.0 136.1 Lipton SA (2006). "Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond". Nat Rev Drug Discov. 5 (2): 160–170. doi:10.1038/nrd1958. PMID 16424917.
  137. "Memantine". US National Library of Medicine (Medline). 2004-01-04. Retrieved 2008-03-22.
  138. Areosa Sastre A, McShane R, Sherriff F (2004). "Memantine for dementia". Cochrane Database Syst Rev (4): CD003154. doi:10.1002/14651858.CD003154.pub2. PMID 15495043.
  139. "Namenda Prescribing Information" (PDF). Forest Pharmaceuticals. Retrieved 2008-02-19.
  140. Raina P, Santaguida P, Ismaila A; et al. (2008). "Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline". Annals of Internal Medicine. 148 (5): 379–397. PMID 18316756.
  141. Ballard C, Waite J (2006). "The effectiveness of atypical antipsychotics for the treatment of aggression and psychosis in Alzheimer's disease". Cochrane Database Syst Rev (1): CD003476. doi:10.1002/14651858.CD003476.pub2. PMID 16437455.
  142. Ballard C, Lana MM, Theodoulou M; et al. (2008). "A Randomised, Blinded, Placebo-Controlled Trial in Dementia Patients Continuing or Stopping Neuroleptics (The DART-AD Trial)". PLoS Med. 5 (4): e76. doi:10.1371/journal.pmed.0050076. PMID 18384230.
  143. Sink KM, Holden KF, Yaffe K (2005). "Pharmacological treatment of neuropsychiatric symptoms of dementia: a review of the evidence". JAMA. 293 (5): 596–608. doi:10.1001/jama.293.5.596. PMID 15687315.
  144. 144.0 144.1 144.2 144.3 "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.
  145. 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): 861–869. doi:10.1191/0269215505cr911oa. PMID 16323385.
  146. 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): 1154–1166. PMID 11342679.
  147. 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.
  148. 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.
  149. 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.
  150. 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.
  151. Camberg L, Woods P, Ooi WL; et al. (1999). "Evaluation of Simulated Presence: a personalised approach to enhance well-being in persons with Alzheimer's disease". J Am Geriatr Soc. 47 (4): 446–452. PMID 10203120.
  152. Neal M, Briggs M (2003). "Validation therapy for dementia". Cochrane Database Syst Rev (3): CD001394. doi:10.1002/14651858.CD001394. PMID 12917907.
  153. Chung JC, Lai CK, Chung PM, French HP (2002). "Snoezelen for dementia". Cochrane Database Syst Rev (4): CD003152. doi:10.1002/14651858.CD003152. PMID 12519587.
  154. 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.
  155. 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: 248–254. doi:10.1192/bjp.183.3.248. PMID 12948999.
  156. Gaugler JE, Kane RL, Kane RA, Newcomer R (2005). "Early community-based service utilization and its effects on institutionalization in dementia caregiving". Gerontologist. 45 (2): 177–85. PMID 15799982. Retrieved 2008-05-30. Unknown parameter |month= ignored (help)
  157. 157.0 157.1 157.2 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.
  158. "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.
  159. "Treating behavioral and psychiatric symptoms". Alzheimer's Association. 2006. Retrieved 2006-09-25.
  160. 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. doi:10.1016/S1353-8292(97)00024-5. PMID 10671009.
  161. 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. doi:10.1016/j.clnu.2003.09.015. PMID 15297089.
  162. "Clinical Trials. Found 459 studies with search of: alzheimer". US National Institutes of Health. Retrieved 2008-03-23.
  163. Vaccination:
  164. "Study Evaluating ACC-001 in Mild to Moderate Alzheimers Disease Subjects". Clinical Trial. [FDA clinicaltrials.gov]. 2008-03-11.
  165. "Study Evaluating Safety, Tolerability, and Immunogenicity of ACC-001 in Subjects With Alzheimer's Disease".
  166. "Bapineuzumab in Patients With Mild to Moderate Alzheimer's Disease/ Apo_e4 non-carriers". Clinical Trial. US National Institutes of Health. 2008-02-29. Retrieved 2008-03-23.
  167. "Efficacy Study of MPC-7869 to Treat Patients With Alzheimer's". Clinical Trial. US National Institutes of Health. 2007-12-11. Retrieved 2008-03-23.
  168. "Safety, Tolerability and Efficacy Study to Evaluate Subjects With Mild Cognitive Impairment". Clinical Trial. US National Institutes of Health. 2008-03-11. Retrieved 2008-03-23.
  169. "Study Evaluating the Safety, Tolerability and Efficacy of PBT2 in Patients With Early Alzheimer's Disease". Clinical Trial. US National Institutes of Health. 2008-01-13. Retrieved 2008-03-23.
  170. 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.
  171. Griffin WS (2008). "Perispinal etanercept: potential as an Alzheimer therapeutic". J Neuroinflammation. 5: 3. doi:10.1186/1742-2094-5-3. PMID 18186919.
  172. Tobinick E (2007). "Perispinal etanercept for treatment of Alzheimer's disease". Curr Alzheimer Res. 4 (5): 550–552. doi:10.2174/156720507783018217. PMID 18220520.
  173. 173.0 173.1 Bowen JD, Malter AD, Sheppard L; et al. (1996). "Predictors of mortality in patients diagnosed with probable Alzheimer's disease". Neurology. 47 (2): 433–9. PMID 8757016. Unknown parameter |month= ignored (help)
  174. 174.0 174.1 Dodge HH, Shen C, Pandav R, DeKosky ST, Ganguli M (2003). "Functional transitions and active life expectancy associated with Alzheimer disease". Arch. Neurol. 60 (2): 253–9. PMID 12580712. Unknown parameter |month= ignored (help)
  175. Larson EB, Shadlen MF, Wang L; et al. (2004). "Survival after initial diagnosis of Alzheimer disease". Ann. Intern. Med. 140 (7): 501–9. PMID 15068977. Unknown parameter |month= ignored (help)
  176. Jagger C, Clarke M, Stone A (1995). "Predictors of survival with Alzheimer's disease: a community-based study". Psychol Med. 25 (1): 171–7. PMID 7792352. Unknown parameter |month= ignored (help)
  177. 177.0 177.1 Ganguli M, Dodge HH, Shen C, Pandav RS, DeKosky ST (2005). "Alzheimer disease and mortality: a 15-year epidemiological study". Arch. Neurol. 62 (5): 779–84. doi:10.1001/archneur.62.5.779. PMID 15883266. Unknown parameter |month= ignored (help)
  178. 178.0 178.1 178.2 Bermejo-Pareja F, Benito-León J, Vega S, Medrano MJ, Román GC (2008). "Incidence and subtypes of dementia in three elderly populations of central Spain". J. Neurol. Sci. 264 (1–2): 63–72. doi:10.1016/j.jns.2007.07.021. PMID 17727890. Unknown parameter |month= ignored (help)
  179. Gorelick P (2004). "Risk factors for vascular dementia and Alzheimer disease". Stroke. 35 (11 Suppl 1): 2620–2622. doi:10.1161/01.STR.0000143318.70292.47. PMID 15375299.
  180. 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. doi:10.1001/archneur.60.8.1119. PMID 12925369.
  181. "Neurological disorders: Public health challenges" (PDF). World Health Organization.
  182. 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): 1992–1997. PMID 10599770.
  183. 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.
  184. 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): 687–694. doi:10.1097/01.ede.0000142147.49297.9d. PMID 15475717.
  185. 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): 108–114. doi:10.1136/oem.2005.024190. PMID 17043077.
  186. Role of metals:
  187. 187.0 187.1 187.2 Di Carlo A, Baldereschi M, Amaducci L; et al. (2002). "Incidence of dementia, Alzheimer's disease, and vascular dementia in Italy. The ILSA Study". J Am Geriatr Soc. 50 (1): 41–8. PMID 12028245. Unknown parameter |month= ignored (help)
  188. Andersen K, Launer LJ, Dewey ME; et al. (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. Unknown parameter |month= ignored (help)
  189. 2000 U.S. estimates:
  190. 190.0 190.1 Ferri CP, Prince M, Brayne C; et al. (2005). "Global prevalence of dementia: a Delphi consensus study" (PDF). Lancet. 366 (9503): 2112–7. doi:10.1016/S0140-6736(05)67889-0. PMID 16360788. Retrieved 2008-06-13. Unknown parameter |month= ignored (help)
  191. World Health Organization (2006). Neurological Disorders: Public Health Challenges. Switzerland: World Health Organization. pp. 204–207. ISBN 978-92-4-156336-9.
  192. 192.0 192.1 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. doi:10.1016/S0197-4580(98)00052-9. PMID 9661992.
  193. Auguste D.:
    • Alzheimer Alöis (1907). "Uber eine eigenartige Erkrankung der Hirnrinde" (in Template:De icon). 64 (1–2): 146–148.
    • 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. Unknown parameter |origdate= ignored (|orig-year= 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.
  194. 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.
  195. Boller F, Forbes MM (1998). "History of dementia and dementia in history: an overview". Journal of Neurological Science. 158 (2): 125–133. doi:10.1016/S0022-510X(98)00128-2. PMID 9702682.
  196. 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.
  197. 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.
  198. 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.
  199. O’Donovan ST. "Dementia caregiving burden and breakdown" (PDF). Forum of Consultant Nurses, Midwives and Allied Health Professionals. Retrieved 2008-02-29.
  200. "The MetLife Study of Alzheimer's Disease: The Caregiving Experience" (PDF). MetLife Mature Market Institute. August 2006. Retrieved 2008-02-12.

Further reading

Template:Diseases of the nervous system

Template:SIB


af:Alzheimer se siekte ar:ألزهايمر ast:Alzheimer bg:Болест на Алцхаймер ca:Malaltia d'Alzheimer cs:Alzheimerova choroba da:Alzheimers sygdom de:Alzheimer-Krankheit eo:Alchajmero eu:Alzheimer fa:آلزایمر gl:Alzheimer ko:알츠하이머병 hy:Ալցհայմերի հիվանդություն hr:Alzheimerova bolest id:Alzheimer is:Alsheimer it:Morbo di Alzheimer he:אלצהיימר lb:Alzheimer lt:Alzheimerio liga hu:Alzheimer-kór ms:Alzheimer nl:Ziekte van Alzheimer no:Alzheimers sykdom simple:Alzheimer's disease sk:Alzheimerova choroba sr:Алцхајмерова болест su:Panyakit Alzheimer fi:Alzheimerin tauti sv:Alzheimers sjukdom uk:Хвороба Альцгеймера yi:אלצהיימערס קרענק


Template:WikiDoc Sources