Jump to content

Alzheimer's disease: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
m Undid revision 191075273 by LeadSongDog (talk) Doesn't match data
Line 532: Line 532:


==Treatment==
==Treatment==
There is currently no cure for Alzheimer's disease. Currently available medications offer relatively small symptomatic benefit for some patients and some medications do slow disease symptom progression.<ref name="pmid18044110">{{cite journal |author=Shah S, Reichman WE |title=Treatment of Alzheimer's disease across the spectrum of severity |journal=Clin Interv Aging |volume=1 |issue=2 |pages=131–42 |year=2006 |pmid=18044110 |doi=}}</ref>
There is currently no cure for Alzheimer's disease. Currently available medications offer relatively small symptomatic benefit for some patients and some medications do slow disease progression.<ref name="pmid18044110">{{cite journal |author=Shah S, Reichman WE |title=Treatment of Alzheimer's disease across the spectrum of severity |journal=Clin Interv Aging |volume=1 |issue=2 |pages=131–42 |year=2006 |pmid=18044110 |doi=}}</ref>


===Pharmaceutical===
===Pharmaceutical===

Revision as of 05:07, 13 February 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 a neurodegenerative disease that, in its most common form, occurs in people over 65 years old and is the most common cause of dementia afflicting 24 million people worldwide.[1]

Clinical signs of Alzheimer's disease include progressive cognitive deterioration, declining ability to perform activities of daily living, and neuropsychiatric symptoms or behavioral changes. Plaques which contain misfolded peptides called amyloid beta (Aβ) are formed in the brain many years before the clinical signs of Alzheimer's are observed. Together, these plaques and neurofibrillary tangles form the pathological hallmarks of the disease. These features can only be discovered at autopsy to confirm the clinical diagnosis. At this time medications can help reduce the symptoms of the disease, but they cannot change the course of the underlying pathology.

The ultimate cause of Alzheimer's is unknown. Genetic factors are clearly indicated as dominant mutations in three different genes that account for the small number of cases of familial, early-onset AD that have been identified. For the more common form of late onset AD, ApoE is the only clearly established susceptibility gene. All four genes can contain mutations or variants that confer increased risk for AD, but they account for only 30% of the genetic picture of AD. Mutations in any of these four genes lead to the excessive accumulation in the brain of Aβ, the main component of the senile plaques that are prevalent in the brains of AD patients.[2][3]

Symptoms

Disease course can be divided into stages, with a different pattern of cognitive and functional impairment occurring at each stage.

Predementia stage

Careful neuropsychological testing can reveal mild cognitive difficulties up to eight years before a person fullfils clinical criteria of diagnosis.[4][5][6] It is not yet clear if these difficulties affect daily living activities. Recent studies show impairments in the most complex activities.[7] The most noticeable deficit for most people is short-term memory loss and the consequent problems to acquire new information, but subtle executive problems or semantic memory impairments can also occur.[8][9] Apathy can be seen at this stage, which is the most common and most persistent neuropsychiatric symptom through the disease.[10][11][12] This stage of the disease has also been termed mild cognitive impairment,[13] 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.[14]

Early stage

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.[15] 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.[16][17] 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.[18][19][20] While performing fine motor tasks such as writing, drawing or dressing, certain visoconstructional difficulties, or apraxia, may present, which may appear as clumsiness.[21] 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.[15]

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.[15] 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.[18][22] As time passes, complex motor sequences become less coordinated, costing the patient most of his daily-living abilities.[23] Memory problems worsen, and the person may not recognize close relatives.[24] Long-term memory, which was previously left intact, is now also impaired.[25] The patient is usually almost completely unaware of its 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.[10][26] Often urinary incontinence develops.[27] 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.[15][28]

Advanced stage

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.[18] Nevertheless many patients can receive and return emotional signals long after the loss of verbal language.[29] Although aggressiveness can still present, extreme apathy and exhaustion are much more common.[15] 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[30] and to lose the ability to feed oneself[31] if death from some external cause (such as infection due to pressure ulcers or pneumonia) does not occur first.[32][33]

Etiology

Most cases of Alzheimer's disease are sporadic, i.e., do not exhibit familial inheritance. Nonetheless, at least 80% of sporadic AD cases most likely 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. 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.[34]

On the other hand, 5 to 10% 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 <60 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 20 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.

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.[35]

Both amyloid plaques and neurofibrillary tangles are clearly visible by microscopy in AD brains.[36] Plaques are dense, mostly insoluble deposits of 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.[37]

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.[38] 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.[39][40] In AD, something causes APP to be divided by enzymes through a mechanism called proteolysis.[41] 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.[42][36] 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.[43] 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.[44] 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.[45]

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.[46] 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,[47] leading to generalized neuroinflammation.[35]

Research after 2000 includes hypotheses centered on the effects of the misfolded and aggregated proteins, amyloid beta and tau. 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.[48] The tau hypothesis is supported by the long-standing observation that deposition of amyloid plaques does not correlate well with neuron loss,[49] but a majority of researchers support the alternative hypothesis that Aβ is the primary causative agent.[48] 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.[50][51] 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.[52] 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.[53] 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.[54][55][56]

Diagnosis

Dementia is by definition a clinical condition, and thus can be confidently diagnosed with careful testing. A definitive diagnosis of Alzheimer's disease as a particular cause of dementia must await microscopic examination of brain tissue; which generally occurs at autopsy and less often with a pre-mortem brain biopsy.[57] Alzheimer's disease is usually a clinically diagnosed condition based on the presence of characteristic neurological and neuropsychological features and the absence of alternative diagnoses. In this process, determination of neurological characteristics is made utilizing patient history and clinical observation,[58] while neuropsychological evaluation includes memory testing and assessment of intellectual functioning.[59] Medical organizations have created diagnostic criteria to ease and standardize the process for practicing physicians.

Diagnostic criteria

The diagnostic criteria for Alzheimer of the NINCDS-ADRDA (NINCDS and the ADRDA) are among the most used.[57] 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.[60] 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 DSM-IV-TR criteria published by the American Psychiatric Association.[61][62]

Diagnostic tools

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.[63][64] 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.[59] 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.[65] This is especially important since a patient with AD is commonly unaware of his or her own deficits (anosognosia). [66] Many times families also have difficulties in the detection of initial dementia symptoms and in adequately communicating them to a physician.[67] 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;[59] some of which may even be reversible;[68] 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.[69][70]

Increasingly, the functional neuroimaging modalities of SPECT and 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.[71] 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.[72] Another recent objective marker of the disease is the analysis of cerebrospinal fluid for amyloid beta or tau proteins.[73] Both advances (neuroimaging and cerebrospinal fluid analysis) have led to the proposal of new diagnostic criteria.[74][59]

Prevention

Although aging itself cannot be prevented, the senescence of it can be mitigated. The evidence relating certain behaviors, dietary intakes, environmental exposures, and diseases to the likelihood of developing Alzheimer's varies in quality and its acceptance by the medical community.[75]

Reducing cardiovascular risk factors, such as hypercholesterolemia, hypertension, diabetes, smoking, lack of physical activity, and high dietary saturated fats are implicated in concomitant reduction in risk for the onset and course of Alzheimer's disease.[76]

There are several intellectual or physical activities that may delay the onset or reduce the severity of Alzheimer's disease. Intellectual stimulation, such as playing chess or completing crossword puzzles,[77] regular physical exercise,[78] and regular social interaction[79] all have a positive effect on the disease.

Diet

A Mediterranean diet, which includes high consumption of fruit and vegetables, bread, wheat and other cereals, olive oil, olive leaf, fish, and red wine, may affect the risk and course of the disease. The diet is considered to be high in dietary fiber and monounsaturated fat and low in saturated fat.[80][81] There is some evidence that B-vitamins and folic acid have a positive effect on the prevention and course of the disease.[82] Recent data from research actually indicates that neither dietary folate nor vitamins B-12 and B-6 have any significant effect on the development of the disease.[83][84] Another study has ruled out vitamins C and E, individually or in combination, from being correlated with the onset of AD.[85] In addition, high intake of folate may be associated with cognitive decline in elderly individuals.[86]

Curcumin in Curry (from the yellow spice Turmeric) may delay disease onset and severity through a possible interaction with metallic ions.[87][88][89] A large clinical study is underway to determine if the compound has a significant effect in humans.[90] Other foods or nutrients, such as Omega-3 fatty acids, especially Docosahexaenoic acid (DHA, often found in fish oil),[91] fresh fruit and vegetables high in the Polyphenol antioxidant (for example, blueberries, apples, grapes, broccoli, and legumes).[92][93] and the moderate consumption of red wine[94][95] may all individually or together reduce the risk and development of Alzheimer's disease.

Pharmaceuticals

Cholesterol-lowering drugs (statins) can reduce the cardiovascular risk, but have not shown much evidence that they have an effect in preventing or reducing the course of the disease.[96][97] Other pharmaceutical therapies such as female Hormone replacement therapy is no longer thought to prevent dementia.[98][99] Long-term usage of non-steroidal anti-inflammatory drugs (NSAIDs), used to reduce joint inflammation and pain, are associated with a reduced likelihood of developing AD, according to some observational studies.[100]

A 2007 systematic review concluded that there was inconsistent and unconvincing evidence that Ginkgo has any positive effect on dementia or cognitive impairment.[101] A large, randomized clinical study is now underway, which examines the effect of Ginkgo to prevent dementia. Results are expected in early 2008. [102]

Treatment

There is currently no cure for Alzheimer's disease. Currently available medications offer relatively small symptomatic benefit for some patients and some medications do slow disease progression.[103]

Pharmaceutical

There is an observed reduction in activity of the cholinergic neurons in the disease. 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). AChE-inhibitors seem to modestly moderate symptoms but do not alter the course of the underlying dementing process.[104] Examples currently marketed include donepezil (Trade name Aricept), galantamine (trade names Reminylin and Nivalin, U.S. trade name Razadyne) and rivastigmine (Exelon). The three products come in an oral form taken once or twice a day. Rivastigmine is also available as a once-daily transdermal patch.[105][106][107]

There is some question as to the effectiveness of cholinesterase inhibitors. Different articles have criticized the design of studies reporting benefit from these drugs, concluding that they have doubtful clinical utility, are costly, and confer many side effects.[108][109][110]

Recent evidence of the involvement of glutamatergic neuronal excitotoxicity in Alzheimer's disease led to the development and introduction of memantine. Memantine is a novel NMDA receptor antagonist, and has been shown to be moderately clinically efficacious.[111][112][103] Memantine is marketed as Akatinol, Axura, Ebixa and Namenda.

Psychosocial intervention

Cognitive and behavioral interventions and rehabilitation strategies may be used as an adjunct to pharmacological treatment, especially in the early to moderately advanced stages of disease. Treatment modalities include counseling, psychotherapy (if cognitive functioning is adequate), reminiscent therapy, reality orientation therapy, and behavioral reinforcements as well as cognitive rehabilitation training.[113][114][115]

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.[116] 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.[117]

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.[118][119][120]

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.[121][122][123]

Simvastatin, a statin, stimulates brain vascular endothelial cells to create a beta-amyloid ejector.[124] The use of this statin may be have a causal relationship to decreased development of the disease.[125]

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.[126] A further study, administering to a single AD patient via perispinal infusion, showed rapid and significant improvement in Alzheimer's symptoms.[127] 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.[128]

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.[129] 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.[130]

Adults with damaged blood vessels in the brain or atrophy in their temporal lobe are more likely to develop Alzheimer's disease. It is known that blood vessel damage in the brain is more likely to occur in patients with high blood pressure, high cholesterol or diabetes. Prevention of these conditions can lower the risk of developing the disease, as well as heart attack and stroke,[131][132][133][134] Head injury and smoking are also risk factors for AD;[135][136] while the role of metals in the etiology of the disease is more controversial.[137][138][139]

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.[15] It is important to note there is also an important individual variability in the presentation and development of the symtoms; 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.[140][141][142][143]

Social issues

Alzheimer's is a major public health challenge since the median age of the industrialized world's population is increasing gradually.[144] Indeed, 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.

The role of family caregivers has also become more prominent, as 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. Home-based care may entail tremendous economic, emotional, and even psychological costs as well (see elderly care). Family caregivers often give up time from work and forego pay to spend 47 hours per week on average with an affected loved one who frequently cannot be left alone. From a survey of patients with long term care insurance, direct and indirect costs of caring for an Alzheimer's patient average $77,500 per year.[145]

History

Auguste D.

Although the origin of the concept of dementia goes as far back as the ancient Greek and Roman philosophers and physicians;[146] it was in 1901 when Alois 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. Alois Alzheimer followed her until she died in 1906, when he first reported the case publicly.[147][148][149] In the following five years eleven similar cases were reported in the medical literature; some of them already using the term Alzheimer's disease.[146] 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.[150]

Accordingly; 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 held in that year 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.[151][152] The term senile dementia of the Alzheimer type (SDAT) was often 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.[153]

Cultural references

Since Alzheimer's is such a prevalent disease, many notable people have lived with it. Well known examples are United States president Ronald Reagan and Irish writer Iris Murdoch, both of whom have articles focused on how their language deteriorated with the disease.[154][155] Other cases are the retired footballer Ferenc Puskas,[156] the former British Prime Minister Harold Wilson,[157] the actress Rita Hayworth,[158] the actor Charlton Heston,[159] 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.

Alzheimer has also been portrayed in many films such as Iris (2001),[160] The Notebook (2004),[161] Thanmathra (2005),[162] or Memories of tomorrow (Ashita no Kioku) (2006);[163] documentaries, such as Malcolm and Barbara...A Love Story (1999) and Malcolm and Barbara...Love’s Farewell (2007) both featuring Malcolm Pointon;[164][165] or TV series; being either the center of the main plot or a secondary issue.

See also

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): 2112–7. doi:10.1016/S0140-6736(05)67889-0. PMID 16360788.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. ^ Bertram L, Mullin K, Parkinson M, Hsiao M, Moscarillo TJ, Wagner SL, Becker KD, Velicelebi G, Blacker D, Tanzi RE (2007). "Is alpha-T catenin (VR22) an Alzheimer's disease risk gene?". J. Med. Genet. 44 (1): e63. doi:10.1136/jmg.2005.039263. PMID 17209133.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  3. ^ Bertram L, Tanzi RE (2004). "The current status of Alzheimer's disease genetics: what do we tell the patients?". Pharmacol. Res. 50 (4): 385–96. doi:10.1016/j.phrs.2003.11.018. PMID 15304236.
  4. ^ 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". Arch. Neurol. 52 (5): 485–90. PMID 7733843.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  5. ^ 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): 2341–7. PMID 15623697.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  6. ^ 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)
  7. ^ Perneczky R, Pohl C, Sorg C; et al. (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}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  8. ^ Rapp MA, Reischies FM (2005). "Attention and executive control predict Alzheimer disease in late life: results from the Berlin Aging Study (BASE)". Am J Geriatr Psychiatry. 13 (2): 134–41. doi:10.1176/appi.ajgp.13.2.134. PMID 15703322.
  9. ^ Spaan PE, Raaijmakers JG, Jonker C (2003). "Alzheimer's disease versus normal ageing: a review of the efficiency of clinical and experimental memory measures". J Clin Exp Neuropsychol. 25 (2): 216–33. PMID 12754679.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  10. ^ 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". Am J Geriatr Psychiatry. 13 (6): 460–8. 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).
  11. ^ Robert PH, Berr C, Volteau M; et al. (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}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  12. ^ 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): 1596–602. doi:10.1212/01.wnl.0000260968.92345.3f. PMID 17485646.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  13. ^ 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)
  14. ^ Petersen RC (2007). "The current status of mild cognitive impairment--what do we tell our patients?". Nat Clin Pract Neurol. 3 (2): 60–1. doi:10.1038/ncpneuro0402. PMID 17279076.
  15. ^ a b c d e f Förstl H, Kurz A (1999). "Clinical features of Alzheimer's disease". Eur Arch Psychiatry Clin Neurosci. 249 (6): 288–90. PMID 10653284. Cite error: The named reference "pmid10653284" was defined multiple times with different content (see the help page).
  16. ^ Carlesimo GA, Oscar-Berman M (1992). "Memory deficits in Alzheimer's patients: a comprehensive review". Neuropsychol Rev. 3 (2): 119–69. PMID 1300219.
  17. ^ Jelicic M, Bonebakker AE, Bonke B (1995). "Implicit memory performance of patients with Alzheimer's disease: a brief review". Int Psychogeriatr. 7 (3): 385–92. PMID 8821346.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  18. ^ a b c Frank EM (1994). "Effect of Alzheimer's disease on communication function". J S C Med Assoc. 90 (9): 417–23. PMID 7967534. Cite error: The named reference "pmid7967534" was defined multiple times with different content (see the help page).
  19. ^ Becker JT, Overman AA (2002). "[The semantic memory deficit in Alzheimer's disease]". Rev Neurol (in Spanish; Castilian). 35 (8): 777–83. PMID 12402233.{{cite journal}}: CS1 maint: unrecognized language (link)
  20. ^ 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): 441–59. PMID 7617154.
  21. ^ Benke T (1993). "Two forms of apraxia in Alzheimer's disease". Cortex. 29 (4): 715–25. PMID 8124945.
  22. ^ Forbes KE, Shanks MF, Venneri A (2004). "The evolution of dysgraphia in Alzheimer's disease". Brain Res. Bull. 63 (1): 19–24. doi:10.1016/j.brainresbull.2003.11.005. PMID 15121235.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  23. ^ 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". J Int Neuropsychol Soc. 11 (4): 446–53. PMID 16209425.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  24. ^ Mendez MF, Martin RJ, Smyth KA, Whitehouse PJ (1992). "Disturbances of person identification in Alzheimer's disease. A retrospective study". J. Nerv. Ment. Dis. 180 (2): 94–6. PMID 1737981.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  25. ^ Sartori G, Snitz BE, Sorcinelli L, Daum I (2004). "Remote memory in advanced Alzheimer's disease". Arch Clin Neuropsychol. 19 (6): 779–89. doi:10.1016/j.acn.2003.09.007. PMID 15288331.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  26. ^ 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". Am J Geriatr Psychiatry. 14 (5): 438–45. doi:10.1097/01.JGP.0000218218.47279.db. PMID 16670248.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  27. ^ Honig LS, Mayeux R (2001). "Natural history of Alzheimer's disease". Aging (Milano). 13 (3): 171–82. PMID 11442300.
  28. ^ Gold DP, Reis MF, Markiewicz D, Andres D (1995). "When home caregiving ends: a longitudinal study of outcomes for caregivers of relatives with dementia". J Am Geriatr Soc. 43 (1): 10–6. PMID 7806732.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  29. ^ Bär M, Kruse A, Re S (2003). "[Situations of emotional significance in residents suffering from dementia]". Z Gerontol Geriatr (in German). 36 (6): 454–62. doi:10.1007/s00391-003-0191-0. PMID 14685735.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  30. ^ Souren LE, Franssen EH, Reisberg B (1995). "Contractures and loss of function in patients with Alzheimer's disease". J Am Geriatr Soc. 43 (6): 650–5. PMID 7775724.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  31. ^ 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): 637–41. PMID 12675103.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  32. ^ Wada H, Nakajoh K, Satoh-Nakagawa T; et al. (2001). "Risk factors of aspiration pneumonia in Alzheimer's disease patients". Gerontology. 47 (5): 271–6. PMID 11490146. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  33. ^ 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)
  34. ^ Alzheimer's Disease Genetics Fact Sheet from the National Institute of Health
  35. ^ a b Wenk GL (2003). "Neuropathologic changes in Alzheimer's disease". J Clin Psychiatry. 64 Suppl 9: 7–10. PMID 12934968.
  36. ^ a b 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): 1984–9. PMID 15184601.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  37. ^ 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". Cereb. Cortex. 4 (2): 138–50. PMID 8038565.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  38. ^ 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 Med. 4 (1–2): 21–36. doi:10.1385/NMM:4:1-2:21. PMID 14528050.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  39. ^ Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J (2006). "Synapse formation and function is modulated by the amyloid precursor protein". J. Neurosci. 26 (27): 7212–21. doi:10.1523/JNEUROSCI.1450-06.2006. PMID 16822978.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  40. ^ 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. Neurobiol. 70 (1): 1–32. PMID 12927332.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  41. ^ Hooper NM (2005). "Roles of proteolysis and lipid rafts in the processing of the amyloid precursor protein and prion protein". Biochem. Soc. Trans. 33 (Pt 2): 335–8. doi:10.1042/BST0330335. PMID 15787600.
  42. ^ Ohnishi S, Takano K (2004). "Amyloid fibrils from the viewpoint of protein folding". Cell. Mol. Life Sci. 61 (5): 511–24. doi:10.1007/s00018-003-3264-8. PMID 15004691.
  43. ^ Hernández F, Avila J (2007). "Tauopathies". Cell. Mol. Life Sci. 64 (17): 2219–33. doi:10.1007/s00018-007-7220-x. PMID 17604998.
  44. ^ Goedert M, Spillantini MG, Crowther RA (1991). "Tau proteins and neurofibrillary degeneration". Brain Pathol. 1 (4): 279–86. PMID 1669718.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  45. ^ Chun W, Johnson GV (2007). "The role of tau phosphorylation and cleavage in neuronal cell death". Front. Biosci. 12: 733–56. PMID 17127334.
  46. ^ Walker LC, Rosen RF (2006). "Alzheimer therapeutics-what after the cholinesterase inhibitors?". Age Ageing. 35 (4): 332–5. doi:10.1093/ageing/afl009. PMID 16644763.
  47. ^ Shen ZX (2004). "Brain cholinesterases: II. The molecular and cellular basis of Alzheimer's disease". Med. Hypotheses. 63 (2): 308–21. doi:10.1016/j.mehy.2004.02.031. PMID 15236795.
  48. ^ a b Mudher A, Lovestone S (2002). "Alzheimer's disease-do tauists and baptists finally shake hands?". Trends Neurosci. 25 (1): 22–6. PMID 11801334.
  49. ^ Schmitz C, Rutten BP, Pielen A; et al. (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}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  50. ^ Nistor M, Don M, Parekh M; et al. (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}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  51. ^ Lott IT, Head E (2005). "Alzheimer disease and Down syndrome: factors in pathogenesis". Neurobiol. Aging. 26 (3): 383–9. doi:10.1016/j.neurobiolaging.2004.08.005. PMID 15639317.
  52. ^ Yankner BA, Duffy LK, Kirschner DA (1990). "Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides". Science. 250 (4978): 279–82. PMID 2218531.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  53. ^ 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". N. Engl. J. Med. 333 (19): 1242–7. PMID 7566000.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  54. ^ 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): 523–7. doi:10.1038/373523a0. PMID 7845465.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  55. ^ 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". J. Neurosci. 16 (18): 5795–811. PMID 8795633.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  56. ^ 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)
  57. ^ a b 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)
  58. ^ Lopera F (2001). "Clinical history in the study of a patient with dementia]". Rev Neurol (in Spanish; Castilian). 32 (12): 1187–91. PMID 11708253.{{cite journal}}: CS1 maint: unrecognized language (link)
  59. ^ a b c d 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. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid17222085" was defined multiple times with different content (see the help page).
  60. ^ 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". Arch. Neurol. 51 (12): 1198–204. PMID 7986174.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  61. ^ 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)
  62. ^ Ito N (1996). "Clinical aspects of dementia". Hokkaido Igaku Zasshi (in Japanese). 71 (3): 315–20. PMID 8752526.
  63. ^ Tombaugh TN, McIntyre NJ (1992). "The mini-mental state examination: a comprehensive review". J Am Geriatr Soc. 40 (9): 922–35. PMID 1512391.
  64. ^ Pasquier F (1999). "Early diagnosis of dementia: neuropsychology". J. Neurol. 246 (1): 6–15. PMID 9987708.
  65. ^ 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 Dis Assoc Disord. 19 (4): 186–94. PMID 16327345.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  66. ^ Antoine C, Antoine P, Guermonprez P, Frigard B (2004). "Awareness of deficits and anosognosia in Alzheimer's disease". Encephale (in French). 30 (6): 570–7. PMID 15738860.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  67. ^ Cruz VT, Pais J, Teixeira A, Nunes B (2004). "The initial symptoms of Alzheimer disease: caregiver perception". Acta Med Port (in Portuguese). 17 (6): 435–44. PMID 16197855.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  68. ^ Sellal F, Becker H (2007). "Potentially reversible dementia". Presse Med (in French). 36 (2 Pt 2): 289–98. doi:10.1016/j.lpm.2006.11.005. PMID 17258677.
  69. ^ Geldmacher DS, Whitehouse PJ (1997). "Differential diagnosis of Alzheimer's disease". Neurology. 48 (5 Suppl 6): S2–9. PMID 9153154.
  70. ^ Potter GG, Steffens DC (2007). "Contribution of depression to cognitive impairment and dementia in older adults". Neurologist. 13 (3): 105–17. doi:10.1097/01.nrl.0000252947.15389.a9. PMID 17495754.
  71. ^ 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): 376–8. doi:10.1097/01.rlu.0000222736.81365.63. PMID 16785801.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  72. ^ Dougall NJ, Bruggink S, Ebmeier KP (2004). "Systematic review of the diagnostic accuracy of 99mTc-HMPAO-SPECT in dementia". The American journal of geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry. 12 (6): 554–70. doi:10.1176/appi.ajgp.12.6.554. PMID 15545324.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  73. ^ 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): 423–31. doi:10.1358/dot.2007.43.6.1067341. PMID 17612711.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  74. ^ 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 Neurol. 6 (8): 734–46. doi:10.1016/S1474-4422(07)70178-3. PMID 17616482.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  75. ^ Small GW (2002). "What we need to know about age related memory loss". BMJ. 324 (7352): 1502–5. PMID 12077041.
  76. ^ 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)
  77. ^ 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)
  78. ^ Larson E, Wang L, Bowen J, McCormick W, Teri L, Crane P, Kukull W (2006). "Exercise is associated with reduced risk for incident dementia among persons 65 years of age and older". Ann Intern Med. 144 (2): 73–81. PMID 16418406.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  79. ^ 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. Retrieved 2008-01-27.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  80. ^ Scarmeas N, Stern Y, Mayeux R, Luchsinger J (2006). "Mediterranean diet, Alzheimer disease, and vascular mediation". Arch Neurol. 63 (12): 1709–17. PMID 17030648.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  81. ^ 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. Retrieved 2008-01-27.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  82. ^ Morris M, Schneider J, Tangney C (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)
  83. ^ 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. Retrieved 2008-01-27.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  84. ^ 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. Retrieved 2008-01-27.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  85. ^ Gray Shelly L., Anderson Melissa L., Crane Paul K., Breitner John C. S., McCormick Wayne, Bowen James D., Teri Linda, Larson Eric (2008). "Antioxidant Vitamin Supplement Use and Risk of Dementia or Alzheimer's Disease in Older Adults". Journal of the American Geriatrics Society. 56 (2): 291–295. doi:10.1111/j.1532-5415.2007.01531.x.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  86. ^ Morris MC, Evans DA, Bienias JL, Tangney CC, Hebert LE, Scherr PA, Schneider JA (2005). "Dietary folate and vitamin B12 intake and cognitive decline among community-dwelling older persons". Arch. Neurol. 62 (4): 641–5. doi:10.1001/archneur.62.4.641. PMID 15824266.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  87. ^ 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". J. Neurochem. 102 (4): 1095–104. doi:10.1111/j.1471-4159.2007.04613.x. PMID 17472706.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  88. ^ Giselle P. Lim1, Teresa Chu1, Fusheng Yang, Walter Beech1, Sally A. Frautschy1, and Greg M. Cole1 (2001). "The Curry Spice Curcumin Reduces Oxidative Damage and Amyloid Pathology in an Alzheimer Transgenic Mouse". The Journal of Neuroscience. 21 (4): 8370–8377. PMID 11606625.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: numeric names: authors list (link)
  89. ^ Baum L, Ng A (2004). "Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models". J. Alzheimers Dis. 6 (4): 367–77, discussion 443–9. PMID 15345806.
  90. ^ Ringman JM, Frautschy SA, Cole GM, Masterman DL, Cummings JL (2005). "A potential role of the curry spice curcumin in Alzheimer's disease". Curr Alzheimer Res. 2 (2): 131–6. PMID 15974909.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  91. ^ 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): 1921–30. doi:10.1212/01.wnl.0000278116.37320.52. PMID 17998483.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  92. ^ Dai Q, Borenstein AR, Wu Y, Jackson JC, Larson EB (2006). "Fruit and vegetable juices and Alzheimer's disease: the Kame Project". Am. J. Med. 119 (9): 751–9. doi:10.1016/j.amjmed.2006.03.045. PMID 16945610.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  93. ^ Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, Ehrhart J, Zeng J, Mori T, Arendash GW, Shytle D, Town T, Tan J (2006). "ADAM10 activation is required for green tea (-)-epigallocatechin-3-gallate-induced alpha-secretase cleavage of amyloid precursor protein". J. Biol. Chem. 281 (24): 16419–27. doi:10.1074/jbc.M600617200. PMID 16624814.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  94. ^ 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): 380–3. doi:10.1159/000073766. PMID 14624067.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  95. ^ Anekonda TS (2006). "Resveratrol--a boon for treating Alzheimer's disease?". Brain Res Rev. 52 (2): 316–26. doi:10.1016/j.brainresrev.2006.04.004. PMID 16766037.
  96. ^ Reiss AB, Wirkowski E (2007). "Role of HMG-CoA reductase inhibitors in neurological disorders : progress to date". Drugs. 67 (15): 2111–20. PMID 17927279.
  97. ^ Kuller LH (2007). "Statins and dementia". Curr Atheroscler Rep. 9 (2): 154–61. PMID 17877925.
  98. ^ 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.
  99. ^ 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)
  100. ^ 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)
  101. ^ 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 2008-01-23.
  102. ^ DeKosky ST, Fitzpatrick A, Ives DG, Saxton J, Williamson J, Lopez OL, Burke G, Fried L, Kuller LH, Robbins J, Tracy R, Woolard N, Dunn L, Kronmal R, Nahin R, Furberg C (2006). "The Ginkgo Evaluation of Memory (GEM) study: design and baseline data of a randomized trial of Ginkgo biloba extract in prevention of dementia". Contemp Clin Trials. 27 (3): 238–53. doi:10.1016/j.cct.2006.02.007. PMID 16627007. Retrieved 2008-01-23.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  103. ^ a b Shah S, Reichman WE (2006). "Treatment of Alzheimer's disease across the spectrum of severity". Clin Interv Aging. 1 (2): 131–42. PMID 18044110. Cite error: The named reference "pmid18044110" was defined multiple times with different content (see the help page).
  104. ^ Musiał A, Bajda M, Malawska B (2007). "Recent developments in cholinesterases inhibitors for Alzheimer's disease treatment". Curr. Med. Chem. 14 (25): 2654–79. PMID 17979717.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  105. ^ "Razadyne ER U.S. Product Insert" (PDF). Novartis. Retrieved 2008-01-30.
  106. ^ "Exelon ER U.S. Product Insert" (PDF). Novartis. Retrieved 2008-01-30.
  107. ^ "Aricept and Aricept ODT Product Insert" (PDF). Eisai Inc. and Pfizer Inc. Retrieved 2008-01-30.
  108. ^ 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 (2004). "Long-term donepezil treatment in 565 patients with Alzheimer's disease (AD2000): randomised double-blind trial". Lancet. 363 (9427): 2105–15. PMID 15220031.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  109. ^ 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)
  110. ^ Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt H, van den Bussche H (2005). "Cholinesterase inhibitors for patients with Alzheimer's disease: systematic review of randomised clinical trials". BMJ. 331 (7512): 321–7. PMID 16081444.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  111. ^ 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.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  112. ^ Daiello LA (2007). "Current issues in dementia pharmacotherapy". Am J Manag Care. 13 Suppl 8: S198–202. PMID 18095783.
  113. ^ Olazarán J, Muñiz R, Reisberg B, Peña-Casanova J, del Ser T, Cruz-Jentoft AJ, Serrano P, Navarro E, García de la Rocha ML, Frank A, Galiano M, Fernández-Bullido Y, Serra JA, González-Salvador MT, Sevilla C (2004). "Benefits of cognitive-motor intervention in MCI and mild to moderate Alzheimer disease". Neurology. 63 (12): 2348–53. PMID 15623698.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  114. ^ Clare L, Woods RT, Moniz Cook ED, Orrell M, Spector A (2003). "Cognitive rehabilitation and cognitive training for early-stage Alzheimer's disease and vascular dementia". Cochrane Database Syst Rev (4): CD003260. doi:10.1002/14651858.CD003260. PMID 14583963.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  115. ^ "Practice Guideline for the Treatment of Patients with Alzheimer's disease and Other Dementias" (PDF). American Psychiatric Association. October 2007. Retrieved 2007-12-28.
  116. ^ "Treating behavioral and psychiatric symptoms". Retrieved 2008-02-12.
  117. ^ Dunne TE, Neargarder SA, Cipolloni PB, Cronin-Golomb A (2004). "Visual contrast enhances food and liquid intake in advanced Alzheimer's disease". Clin Nutr. 23 (4): 533–8. PMID 15297089.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  118. ^ 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 Dis Assoc Disord. 21 (4): 292–9. doi:10.1097/WAD.0b013e31815d1048. PMID 18090435.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  119. ^ 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)
  120. ^ Christensen DD (2007). "Alzheimer's disease: progress in the development of anti-amyloid disease-modifying therapies". CNS Spectr. 12 (2): 113–6, 119–23. PMID 17277711.
  121. ^ Hawkes CA, McLaurin J (2007). "Immunotherapy as treatment for Alzheimer's disease". Expert Rev Neurother. 7 (11): 1535–48. doi:10.1586/14737175.7.11.1535. PMID 17997702.
  122. ^ Solomon B (2007). "Clinical immunologic approaches for the treatment of Alzheimer's disease". Expert Opin Investig Drugs. 16 (6): 819–28. doi:10.1517/13543784.16.6.819. PMID 17501694.
  123. ^ Woodhouse A, Dickson TC, Vickers JC (2007). "Vaccination strategies for Alzheimer's disease: A new hope?". Drugs Aging. 24 (2): 107–19. PMID 17313199.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  124. ^ Whitfield JF (2007). "The road to LOAD: late-onset Alzheimer's disease and a possible way to block it". Expert Opin. Ther. Targets. 11 (10): 1257–60. doi:10.1517/14728222.11.10.1257. PMID 17907956.
  125. ^ 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)
  126. ^ 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)
  127. ^ Tobinick EL, Gross H (2008). "Rapid cognitive improvement in Alzheimer's disease following perispinal etanercept administration". J Neuroinflammation. 5: 2. doi:10.1186/1742-2094-5-2. PMID 18184433.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  128. ^ Landreth G (2006). "PPARgamma agonists as new therapeutic agents for the treatment of Alzheimer's disease". Exp. Neurol. 199 (2): 245–8. doi:10.1016/j.expneurol.2006.04.006. PMID 16733054.
  129. ^ Gorelick P (2004). "Risk factors for vascular dementia and Alzheimer disease". Stroke. 35 (11 Suppl 1): 2620–2. PMID 15375299.
  130. ^ Hebert L, Scherr P, Bienias J, Bennett D, Evans D (2003). "Alzheimer disease in the U.S. population: prevalence estimates using the 2000 census". Arch Neurol. 60 (8): 1119–22. PMID 12925369.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  131. ^ Kehoe P, Wilcock G (2007). "Is inhibition of the renin-angiotensin system a new treatment option for Alzheimer's disease?". Lancet neurology. 6 (4): 373–8. PMID 17362841.
  132. ^ Kofman OS, MacMillan VH (1970). "Diffuse Cerebral Atrophy". Applied Therapeutics. 12 (4): 24–26. PMID 5446326.
  133. ^ Crisby M, Carlson L, Winblad B (2002). "Statins in the prevention and treatment of Alzheimer disease". Alzheimer disease and associated disorders. 16 (3): 131–6. PMID 12218642.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  134. ^ Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, Staufenbiel M, Huang LE, Song W (2006). "Hypoxia facilitates Alzheimer's disease pathogenesis by up-regulating BACE1 gene expression". Proc. Natl. Acad. Sci. U.S.A. 103 (49): 18727–32. doi:10.1073/pnas.0606298103. PMID 17121991.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  135. ^ Mayeux R, Ottman R, Tang M, Noboa-Bauza L, Marder K, Gurland B, Stern Y (1993). "Genetic susceptibility and head injury as risk factors for Alzheimer's disease among community-dwelling elderly persons and their first-degree relatives". Ann. Neurol. 33 (5): 494–501. PMID 8498827.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  136. ^ Anstey KJ, von Sanden C, Salim A, O'kearney R (2007). "Smoking as a risk factor for dementia and cognitive decline: a meta-analysis of prospective studies". Am. J. Epidemiol. 166 (4): 367–78. doi:10.1093/aje/kwm116. PMID 17573335.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  137. ^ Rondeau V, Commenges D, Jacqmin-Gadda H, Dartigues JF (2000). "Relation between aluminum concentrations in drinking water and Alzheimer's disease: an 8-year follow-up study". Am. J. Epidemiol. 152 (1): 59–66. PMID 10901330.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  138. ^ Shcherbatykh I, Carpenter DO (2007). "The role of metals in the etiology of Alzheimer's disease". J. Alzheimers Dis. 11 (2): 191–205. PMID 17522444.
  139. ^ Santibáñez M, Bolumar F, García AM (2007). "Occupational risk factors in Alzheimer's disease: a review assessing the quality of published epidemiological studies". Occup Environ Med. 64 (11): 723–32. doi:10.1136/oem.2006.028209. PMID 17525096.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  140. ^ 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.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  141. ^ 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.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  142. ^ Kaldjian LC (2004). "Survival versus prognosis in Alzheimer disease" (PDF). Ann. Intern. Med. 141 (7): 575, author reply 575. PMID 15466779. Retrieved 2008-02-12.
  143. ^ "Alzheimer's Disease Information Page". National Institute of Neurological Disorders and Stroke (NINDS). 2008-02-07. Retrieved 2008-02-12.
  144. ^ Sloane P, Zimmerman S, Suchindran C, Reed P, Wang L, Boustani M, Sudha S. "The public health impact of Alzheimer's disease, 2000-2050: potential implication of treatment advances". Annu Rev Public Health. 23: 213–31. PMID 11910061.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  145. ^ "The MetLife Study of Alzheimer's Disease: The Caregiving Experience" (PDF). MetLife Mature Market Institute. August 2006. Retrieved 2008-02-12.
  146. ^ a b Berchtold NC, Cotman CW (1998). "Evolution in the conceptualization of dementia and Alzheimer's disease: Greco-Roman period to the 1960s". Neurobiol. Aging. 19 (3): 173–89. PMID 9661992.
  147. ^ Alzheimer, A. (1907). "Uber eine eigenartige Erkrankung der Hirnrinde". Allg. Z. Psychiat. Psych.-Gerichtl. Med. 64 (1–2): 146–148.
  148. ^ Maurer, Konrad (2003). Alzheimer: the life of a physician and the career of a disease. New York: Columbia University Press. ISBN 0-231-11896-1. {{cite book}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  149. ^ Hochberg, Fred H.; Rottenberg, David (1977). Neurological classics in modern translation. New York: Hafner Press. ISBN 0-02-851180-8.{{cite book}}: CS1 maint: multiple names: authors list (link)
  150. ^ Kraepelin, Emil (1910). Textbook of Psychiatry.
  151. ^ Boller F, Forbes MM (1998). "History of dementia and dementia in history: an overview". J. Neurol. Sci. 158 (2): 125–33. PMID 9702682.
  152. ^ Katzman (1978). Alzheimer's disease: senile dementia and related disorders. New York: Raven Press. {{cite book}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  153. ^ 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): 1497–9. PMID 3531918.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  154. ^ 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): 250–60. doi:10.1093/brain/awh341. PMID 15574466.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  155. ^ 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.
  156. ^ "Hungary legend Puskas dies at 79". BBC. 2006-11-17. Retrieved 2008-01-25.
  157. ^ "Harold Wilson". 10 Downing Street. Retrieved 2008-01-25.
  158. ^ "Chicago Rita Hayworth Gala". Alzheimer’s Association. 2007. Retrieved 2008-01-25.
  159. ^ "Charlton Heston has Alzheimer's symptoms". CNN. 2002-08-09. Retrieved 2008-01-25.
  160. ^ "Iris". IMDB. Retrieved 2008-01-24.
  161. ^ "The Notebook". IMDB. Retrieved 2008-01-24.
  162. ^ "Thanmathra". Webindia123.com. Retrieved 2008-01-24.
  163. ^ "Ashita no kioku". IMDB. Retrieved 2008-01-24.
  164. ^ "Malcolm and Barbara...A Love Story". Dfgdocs. Retrieved 2008-01-24.
  165. ^ "Alzheimer's film-maker to face ITV lawyers". Media Guardian. 2007-08-07. Retrieved 2008-01-24.

Further reading

Template:Link FA