3372 PART 13 Neurologic Disorders
■ EPIDEMIOLOGY
The most important risk factors for AD are increasing age and a positive family history. In the United States, approximately 10% of people
over age 65 years have AD, including 3% of people age 65–74 years, 17%
of people age 75–84 years, and 32% of people age 85 years and older. A
positive family history of dementia suggests a genetic contribution to
AD, which is usually attributable to the ApoE ε4 risk allele. Autosomal
dominant inheritance occurs in only 1–2% of patients and is typically
accompanied by a multigenerational history of early-onset dementia.
Female sex is a risk factor independent of the greater longevity of
females, and women who carry a single ApoE ε4 allele are more susceptible than are male ε4 carriers. A history of mild-to-severe traumatic
brain injury increases the risk for AD. AD is more common in groups
with low educational attainment, but education influences test-taking
ability, and it is clear that AD can affect persons of all intellectual levels.
One study found that the capacity to express complex written language
in early adulthood correlated with a decreased risk for AD. Similarly,
illiteracy and low educational attainment are risk factors for dementia.
Numerous environmental factors, including aluminum, mercury, and
viruses, have been proposed as causes of AD, but rigorous studies have
failed to demonstrate a significant role for any of these exposures. Similarly, several studies suggest that the use of nonsteroidal anti-inflammatory
agents is associated with a decreased risk of AD, but this risk has not been
confirmed in large prospective studies. Vascular disease, and stroke in
particular, seems to lower the threshold for the clinical expression of
AD. Also, in many patients with AD, amyloid angiopathy can lead to
microhemorrhages, large lobar hemorrhages, ischemic infarctions most
often in the subcortical white matter, or in rare cases an inflammatory
leukoencephalopathy. Diabetes increases the risk of AD threefold. Elevated homocysteine and cholesterol levels; hypertension; obesity; hearing loss; tobacco use; diminished serum levels of folic acid; low dietary
intake of fruits, vegetables, and red wine; sleep disorders; low levels of
exercise; and air pollution exposure are all being explored as potential
risk factors for dementia in general and AD in particular.
■ PATHOLOGY
At autopsy, the earliest and most severe degeneration is usually
found in the medial temporal lobe (entorhinal/perirhinal cortex and
hippocampus), inferolateral temporal cortex, and nucleus basalis of
Meynert. The characteristic microscopic findings are neuritic plaques
and NFTs (Fig. 431-2). These lesions accumulate in small numbers
during normal brain aging but dominate the picture in AD. The
overall burden of AD neuropathologic changes can be graded based
on the topography of Aβ plaques, the density of neuritic plaques, and
the spatial extent of NFTs present. Increasing evidence suggests that
soluble amyloid species called oligomers may cause cellular dysfunction and represent the early toxic molecule in AD. Eventually, further
amyloid polymerization and fibril formation lead to neuritic plaques,
which contain a central core of amyloid, proteoglycans, ApoE, αantichymotrypsin, and other proteins. Aβ is a protein of 39–42 amino
acids that is derived proteolytically from a larger transmembrane protein, amyloid precursor protein (APP), when APP is cleaved by β and γ
secretases (Fig. 431-3). The normal function of the Aβ peptides remains
uncertain. APP has neurotrophic and neuroprotective properties.
The plaque core is surrounded by a halo, which contains dystrophic,
tau-immunoreactive neurites and activated microglia. The accumulation of Aβ in cerebral arterioles is termed amyloid angiopathy. NFTs are
composed of silver-staining neuronal cytoplasmic fibrils composed of
abnormally phosphorylated tau protein; they appear as paired helical
filaments by electron microscopy. Tau binds to and stabilizes microtubules, supporting axonal transport of organelles, glycoproteins, neurotransmitters, and other important cargoes throughout the neuron. Once
hyperphosphorylated, tau can no longer bind properly to microtubules
and redistributes from the axon to throughout the neuronal cytoplasm
and distal dendrites, compromising function. Other theories emphasize that abnormal conformations of tau induce misfolding of native
(unfolded) tau into pathologic conformations and that this prionlike templating process is responsible for tau spreading (Chap. 424).
Finally, patients with AD often show comorbid DLB, TDP-43, or vascular pathology. Most prevailing rodent models of AD involve expression
of mutant transgenes that leads to Aβ42 accumulation in the absence of
tauopathy. Even in these models, diminishing neuronal tau ameliorates
cognitive deficits and nonconvulsive seizures while Aβ42 continues
to accumulate, raising hope for tau-lowering therapies in humans.
Biochemically, AD is associated with a decrease in the cortical levels
of several proteins and neurotransmitters, especially acetylcholine, its
A B
FIGURE 431-2 Neuropathology of Alzheimer’s disease. A. Early neurofibrillary degeneration, consisting of NFTs and neuropil threads, preferentially affects the medial
temporal lobes, especially the stellate pyramidal neurons that compose the layer 2 islands of entorhinal cortex, as shown using Gallyas silver staining. B. Higher
magnification view reveals the fibrillar nature of tangles (arrows) and the complex structure of neuritic plaques (arrowheads), whose major component is Aβ (inset shows
immunohistochemistry for Aβ). Scale bars are 500 μM in A, 50 μM in B, and 20 μM in B inset.
3373Alzheimer’s Disease CHAPTER 431
synthetic enzyme choline acetyltransferase, and nicotinic cholinergic
receptors. Reduction of acetylcholine reflects degeneration of cholinergic neurons in the nucleus basalis of Meynert, located just below the
thalamus and adjacent to the third ventricle, that project throughout
the cortex. There is also noradrenergic and serotonergic depletion due
to degeneration of upper brainstem nuclei such as the locus coeruleus
(norepinephrine) and dorsal raphe (serotonin), where tau-immunoreactive neuronal cytoplasmic inclusions can be identified in early adult
life, even in individuals lacking entorhinal cortex NFTs.
■ GENETIC CONSIDERATIONS
Several genes play an important role in the pathogenesis of AD.
One is the APP gene on chromosome 21. Adults with trisomy 21
(Down’s syndrome) consistently develop the typical neuropathologic hallmarks of AD if they survive beyond age 40 years, and many
develop a progressive dementia superimposed on their baseline deficits. The extra dose of the APP gene on chromosome 21 is the initiating
cause of AD in adult Down’s syndrome and results in excess cerebral
amyloid production. Supporting this hypothesis, some families with
early-age-of-onset familial AD (FAD) have point mutations in APP.
Although very rare, these families were the first examples of single-gene
autosomal dominant transmission of AD.
Investigation of large families with multigenerational FAD led to the
discovery of two additional AD-causing genes, the presenilins. Presenilin-1
(PSEN-1) is on chromosome 14 and encodes presenilin-1 protein (also
known as S182). Mutations in this gene cause an early-age-of-onset
AD, with onset typically before age 60 and often before age 50, transmitted in an autosomal dominant, highly penetrant fashion. More than
100 different mutations have been found in the PSEN-1 gene in families
from a wide range of ethnic backgrounds. Presenilin-2 (PSEN-2) is
on chromosome 1 and encodes the presenilin-2 protein (also known
as STM2). A mutation in the PSEN-2 gene was first found in a group
of American families with Volga German ethnic background. Mutations in PSEN-1 are much more common than those in PSEN-2. The
presenilins are highly homologous and encode similar proteins that
at first appeared to have seven transmembrane domains (hence the
designation STM), but subsequent studies have suggested eight such
domains, with a ninth submembrane region. Both presenilins are cytoplasmic neuronal proteins that are widely expressed throughout the
nervous system. They are homologous to a cell-trafficking protein, sel
12, found in the nematode Caenorhabditis elegans. Prior to symptom
Nontoxic
Cell
membrane
Step 1: Cleavage by either α or β secretase
Step 2: Cleavage by γ secretase
β Secretase product α Secretase product
APP β α
γ
Toxic Nontoxic
Amyloidogenic
Aβ42 Aβ40 P3
FIGURE 431-3 Amyloid precursor protein (APP) is catabolized by α, β, and γ
secretases. A key initial step is the digestion by either β secretase (BACE) or α
secretase (ADAM10 or ADAM17 [TACE]), producing smaller nontoxic products.
Cleavage of the β secretase product by γ secretase (Step 2) results in either
the toxic Aβ42 or the nontoxic Aβ40 peptide; cleavage of the α secretase product
by γ secretase produces the nontoxic P3 peptide. Excess production of Aβ42 is a
key initiator of cellular damage in Alzheimer’s disease (AD). Therapeutics for AD
have focused on attempts to reduce accumulation of Aβ42 by antagonizing β or γ
secretases, promoting α secretase, or clearing Aβ42 that has already formed by use
of specific antibodies.
onset, patients with mutations in the presenilin genes have elevated
CSF levels of Aβ42, and PSEN-1 mutations produce increased Aβ42 in
the media in cell culture. PSEN-1 is involved in the cleavage of APP at
the γ secretase site and mutations in either gene (PSEN-1 or APP) may
disturb γ secretase cleavage. Mutations in PSEN-1 are the most common cause of early-age-of-onset FAD, representing 40–70% of all cases.
Mutations in PSEN-1 tend to produce AD with an earlier age of onset
(mean onset 45 years) and a shorter, more rapidly progressive course
(mean duration 6–7 years) than mutations in PSEN-2 (mean onset 53
years; duration 11 years). Although some carriers of PSEN-2 mutations
have had onset of dementia after the age of 70 years, mutations in the
presenilins rarely lead to late-age-of-onset AD. Clinical genetic testing
for these uncommon mutations is available but likely to be revealing
only in early-age-of-onset FAD and should be performed in association
with formal genetic counseling.
The APOE gene on chromosome 19 is involved in the pathogenesis
of AD. The protein product, ApoE, participates in cholesterol transport (Chap. 407), and the gene has three alleles: ε2, ε3, and ε4. The
ApoE ε4 allele confers increased risk of AD in the general population,
including sporadic and late-age-of-onset familial forms. Approximately
24–30% of the nondemented white population has at least one ε4
allele (12–15% allele frequency), and about 2% are ε4/ε4 homozygotes.
Among patients with AD, 40–65% have at least one ε4 allele, a highly
significant elevation compared with controls. The increased risk associated with a single ε4 allele is especially prominent in women. The risk
of AD in ApoE ε4 carriers also varies by racial and ethnic background,
with increased risk in East Asians and decreased risk in blacks and
Hispanics compared with whites. Additionally, many patients with
AD have no ε4 allele, and ε4 carriers may never develop AD. Therefore, ε4 is neither necessary nor sufficient to cause AD. Nevertheless,
the ApoE ε4 allele represents the most important genetic risk factor
for sporadic AD and acts as a dose-dependent disease modifier, with
each ApoE ε4 allele associated with an approximately 10-year earlier
age of onset. The association between ApoE ε4 and AD is strongest in
patients 60–85 years of age and is weaker in younger patients and in
the very old. The precise mechanisms through which ApoE ε4 confers
AD risk or hastens onset remain unclear, but ε4 leads to less efficient
amyloid clearance and production of toxic fragments from cleavage
of the molecule. ApoE can be identified in neuritic plaques and may
also be involved in NFT formation, because it binds to tau protein.
ApoE ε4 decreases neurite outgrowth in dorsal root ganglion neuronal
cultures, perhaps indicating a deleterious role in the brain’s response
to injury. Increasing evidence suggests that the ε2 allele may reduce
AD risk. Use of ApoE testing in AD diagnosis remains controversial
because its predictive value remains unclear and many individuals
with the ε4 allele will never develop dementia. Cognitively normal ε4
heterozygotes and homozygotes may show decreased posterior cerebral
cortical metabolic function by PET imaging, suggesting presymptomatic abnormalities due to AD or an inherited vulnerability of the
AD-targeted network. In demented persons who meet clinical criteria
for AD, finding an ε4 allele increases the reliability of diagnosis; however, the absence of an ε4 allele cannot be considered evidence against
AD. Nevertheless, ApoE ε4 remains the single most important biologic
marker associated with AD risk, and studies of ε4’s functional role and
diagnostic utility are progressing rapidly. ApoE genotyping is available
in some straight-to-consumer genetic testing platforms. The ε4 allele
is associated with increased risk for cerebral amyloid angiopathy
(CAA), DLB, and vascular dementia, while its association with FTD is
uncertain. Some evidence suggests that ε4 may worsen the expression
of non-AD degenerative disorders, head trauma, and other brain injuries. Additional genes are also likely to be involved in AD, especially
as minor-risk alleles for sporadic forms of the disease. Genome-wide
association studies have identified more than 20 additional common
genetic variants that, individually, have small (i.e., odds ratios ~1.1–1.2
or 0.8–0.9) impacts on the risk of AD. Implicated genes converge in
biologic pathways related to innate immunity, lipid metabolism, and
synaptic function. Examples include the clusterin (CLU), phosphatidylinositol-binding clathrin assembly protein (PICALM), and complement component (3b/4b) receptor 1 (CR1) genes, among others. CLU
3374 PART 13 Neurologic Disorders
may play a role in synapse turnover, PICALM participates in clathrinmediated endocytosis, and CR1 may be involved in amyloid clearance
or synapse loss through the complement pathway. TREM2 is a gene
involved with inflammation that increases the likelihood of dementia.
Homozygous mutation carriers develop a frontal dementia with bone
cysts (Nasu-Hakola disease), whereas heterozygotes are predisposed
to the development of AD. TREM2 risk alleles are rare but have strong
effects, with odds ratios estimated at 3–4 for developing clinical AD.
Polygenic hazard scores that integrate the presence of multiple risk and
protective alleles may be useful in predicting an individual’s lifetime
risk of developing AD. The vast majority of AD genetic studies have
focused on white populations of European descent, and much less is
known about the genetics of AD in nonwhite populations.
TREATMENT
Alzheimer’s Disease
The management of AD is challenging and gratifying despite the
absence of a cure or a robust pharmacologic treatment. The primary focus is on long-term amelioration of associated behavioral
and neurologic problems, as well as providing caregiver support,
though many potential disease-modifying therapies are currently
being tested in human trials.
PATIENT AND CAREGIVER EDUCATION
Building rapport with the patient, family members, and other caregivers is essential. In the early stages of AD, memory aids such as
notebooks and posted daily reminders can be helpful. Family members should emphasize activities that are pleasant while curtailing
those that increase stress on the patient. Kitchens, bathrooms,
stairways, and bedrooms need to be made safe, and eventually
patients will need to stop driving. Loss of independence and change
of environment may worsen confusion, agitation, and anger. Communication and repeated calm reassurance are necessary. Caregiver
“burnout” is common, often resulting in nursing home placement
of the patient or new health problems for the caregiver. Respite
breaks for the caregiver help to maintain a successful long-term
therapeutic milieu. Use of adult day-care centers can be helpful.
Local and national support groups, such as the Alzheimer’s Association
and the Family Caregiver Alliance, are valuable resources. Internet
access to these resources has become available to clinicians and
families in recent years.
NEUROTRANSMITTER-BASED THERAPIES
Donepezil (target dose, 10 mg daily), rivastigmine (target dose, 6 mg
twice daily or 9.5-mg patch daily), galantamine (target dose 24 mg
daily, extended-release), and memantine (target dose, 10 mg twice
daily) are approved by the U.S. Food and Drug Administration
(FDA) for the treatment of AD. Due to hepatotoxicity, tacrine is
no longer used. Dose escalations for each of these medications
must be carried out over 4–6 weeks to minimize side effects. The
pharmacologic action of donepezil, rivastigmine, and galantamine
is inhibition of the cholinesterases, primarily acetylcholinesterase,
with a resulting increase in cerebral acetylcholine levels. Memantine appears to act by blocking overexcited N-methyl-d-aspartate
(NMDA) glutamate receptors. Double-blind, placebo-controlled,
crossover studies with cholinesterase inhibitors (in mild-to-severe
AD dementia) and memantine (in moderate-to-severe AD dementia) have shown them to be associated with modestly improved
caregiver ratings of patients’ functioning and with an apparent
decreased rate of decline in cognitive test scores over periods of up
to 3 years. The average patient on an anticholinesterase inhibitor
maintains his or her mini-mental state examination (MMSE) score
for close to a year, whereas a placebo-treated patient declines 2–3
points over the same time period. Memantine, used in conjunction with cholinesterase inhibitors or by itself, slows cognitive
deterioration and decreases caregiver burden for patients with
moderate-to-severe AD but is not approved for mild AD. Neither
cholinesterase inhibitors nor memantine has proven efficacious in
patients with MCI. Cholinesterase inhibitors are relatively easy to
administer, and their major side effects are gastrointestinal symptoms (nausea, diarrhea, cramps), altered sleep with unpleasant or
vivid dreams, bradycardia (usually benign), and muscle cramps.
Potential side effects associated with memantine include constipation, dizziness, headache, and somnolence. A common approach to
AD drug therapy is to initiate a cholinesterase inhibitor for a patient
diagnosed with mild AD dementia, and to add memantine when
patients enter the moderate stage of disease. Cholinesterase inhibitors may also be effective in treating delusions and hallucinations,
while memantine can reduce agitation.
THERAPIES TARGETING AMYLOID-a
AD drug development over the past two decades has focused on
the prevention or clearance of Aβ pathology. In June 2021, aducanumab, a monoclonal antibody targeting the N-terminus of the
Aβ peptide, was granted accelerated approval by the FDA based
on reduction in Aβ plaques (measured by PET) in two phase 3,
double-blinded, randomized placebo-controlled trials. However, a
clinical benefit over placebo (measured by slower decline on cognitive and functional scales) was observed with high-dose treatment
(10 mg/kg) in only one of the two trials, and lower antibody doses
did not show a benefit vs. placebo in either trial. Data interpretation was further complicated by differences in dosing between
trials and early termination of both trials based on a prespecified
futility analysis, which ultimately proved erroneous. Given these
circumstances, continued FDA approval will be contingent upon
verification of clinical benefit in confirmatory trials.
According to the FDA label, aducanumab should only be considered for treatment of patients with MCI or early dementia due
to AD, mirroring the early clinical stage of patients enrolled in the
phase 3 clinical trials. Patients with pre-clinical (i.e., asymptomatic, biomarker-positive) AD or patients with moderate-severe AD
dementia should not be treated until data on safety or efficacy in these
populations is available. Expert recommendations further stipulate
that biomarker confirmation of Aβ based on CSF or PET be required
prior to initiating treatment, since clinical diagnosis in isolation is not
sufficient to ensure the presence of Aβ plaques. Patients with confounding neurological or psychiatric conditions, unstable medical illnesses,
evidence of prior brain hemorrhages (including multiple microhemorrhages), or active anticoagulant treatment should be excluded.
Aducanumab is administered as an intravenous infusion every
4 weeks, with gradual dose titration from 1 mg/kg to 10 mg/kg over
seven infusions. Amyloid-related imaging abnormalities (ARIA)
are the most common adverse effect, occurring in 41% of patients
treated with high-dose aducanumab vs. 10% in the placebo groups.
ARIA can manifest as vasogenic edema (ARIA-E) or cortical microhemorrhages and superficial siderosis (ARIA-H). Of ARIA cases in
the phase 3 trials, 74% were asymptomatic and detected by safety
MRIs. The most common symptoms associated with ARIA were
headache, altered mental status, dizziness, visual disturbances, and
nausea. Symptoms were usually mild and transient, though severe
cases with focal neurologic deficits have been described. Most cases
occurred within the first eight infusions, though ARIA can occur at
any time. Baseline and safety surveillance MRI scans (at minimum
following the 7th and 12th infusions) are required for aducanumab
treatment, and an MRI is also indicated in treated patients in whom
ARIA is suspected clinically. Therefore, patients with contraindications to MRI cannot safely receive this therapy. ApoE genotyping
may also be considered to inform risk-benefit discussions prior to
treatment, because ARIA-E is more common in ApoE ε4 carriers
(43%) compared to noncarriers (20%). It is highly recommended
that the antibody only be prescribed by clinicians who have adequate training and access to the resources needed to safely deliver
this complex therapy.
ADDITIONAL THERAPIES
Mild-to-moderate depression is common in the early stages of AD
and may respond to antidepressants or cholinesterase inhibitors.
3375Alzheimer’s Disease CHAPTER 431
Selective serotonin reuptake inhibitors (SSRIs) are commonly used
due to their low anticholinergic side effects (for example, escitalopram, target dose 5–10 mg daily). Seizures can be treated with
levetiracetam unless the patient had a different regimen that was
effective prior to the onset of AD. Agitation, insomnia, hallucinations, and belligerence are especially troublesome characteristics of
some AD patients, and these behaviors can lead to nursing home
placement. The newer generation of atypical antipsychotics, such as
risperidone, quetiapine, and olanzapine, are being used in low doses
to treat these neuropsychiatric symptoms. The few controlled studies comparing drugs against behavioral intervention in the treatment of agitation suggest mild efficacy with significant side effects
related to sleep, gait, and cardiovascular complications, including
an increased risk of death. All antipsychotics carry a black box FDA
warning for use in elderly patients with dementia and thus should
be prescribed only with caution; however, careful, daily, nonpharmacologic behavior management is often not available, rendering
medications necessary for some patients. Medications with strong
anticholinergic effects should be vigilantly avoided, including prescription and over-the-counter sleep aids (e.g., diphenhydramine)
or incontinence therapies (e.g., oxybutynin).
Several commonly used medications and supplements, including estrogen hormone replacement therapy, statins, vitamin E,
and ginkgo biloba, appeared to be associated with a decreased
risk of AD in epidemiologic or observational studies, but did not
show efficacy in prospective, randomized, double-blinded, placebocontrolled trials. Many vitamins and dietary supplements are marketed directly to consumers as “memory enhancing” or protective
against AD without clinical evidence. Patients and families may
come across anecdotal reports of “miraculous” responses to aggressive treatments such as anti-interferon intrathecal infusions, intravenous immunoglobulin, antibiotics (purportedly to treat Lyme
disease or another questionable infection), metal chelation, and
stem cell therapies, but there is no scientific evidence to support use
of any of these approaches to treating AD, and significant concern
for harm.
EXPERIMENTAL THERAPIES
The design of AD clinical trials has been transformed by the availability of PET and CSF biomarkers of Aβ and tau. Many trials now
require biomarker evidence of AD for trial inclusion. Biomarkers
help assess target engagement (e.g., changes in CSF or PET Aβ in
an antiamyloid trial) or modification of downstream disease pathophysiology (e.g., changes in CSF or PET tau in an antiamyloid trial),
with the pivotal trials leading to approval of aducanumab being
emblematic of this novel approach. Increasingly, many trials have
shifted toward enrolling patients in the asymptomatic (preclinical)
or very early symptomatic stages of AD, using positive biomarkers
as the primary inclusion criterion. Primary (biomarker-negative)
and secondary (biomarker-positive but no symptoms) prevention
trials are underway in autosomal dominant mutation carriers, ApoE
ε4 homozygotes, and even in the normally aging population.
Beyond aducanumab, several additional anti-Aβ monoclonal
antibodies (e.g., lecanemab, gantenerumab, and donanemab) have
shown evidence of robust amyloid plaque lowering on PET and are
currently being evaluated in clinical trials across the continuum
from preclinical disease to mild dementia due to AD. As with aducanumab, ARIA-E and ARIA-H represent a safety concern for this
class of drugs. Active vaccination against Aβ is another approach
that aims to promote immune-mediated clearance of amyloid
pathology. The first Aβ42 vaccine trial in humans was aborted after
a minority of patients developed meningoencephalitis, but subsequent trials with less immunogenic formulations have shown more
favorable safety profiles.
Oral drugs that inhibit β and γ secretase reduce the cleavage of
APP to Aβ42 and showed promise in ameliorating pathology and
behavioral changes in AD transgenic mice. Unfortunately, placebocontrolled trials failed to show clinical efficacy, and trials of β
secretase inhibitors in particular, consistently found significant
worsening of cognition in treated patients vs. placebo, though fortunately this effect proved transient after discontinuing the drug. It is
unclear whether toxicity of β and γ secretase inhibitors was directly
related to changes in Aβ metabolism or to “off-target” drug effects.
Monoclonal antibodies directed against phosphorylated tau are
in earlier stages of development. These antibodies aim to prevent the transsynaptic spread of tau and have proven effective in
tau-transgenic mice. Safety profiles in human studies have proven
favorable thus far. Additional therapeutic approaches targeting tau
include: active immunization; inhibition of tau phosphorylation,
acetylation, and aggregation; microtubule stabilization; and lowering of
tau expression via antisense oligonucleotides or small interfering RNA.
Other druggable pathways represented in the AD drug development
pipeline include neuroinflammation, metabolism/bioenergetics,
synaptic plasticity, neuroprotection, and neurotransmitter-based
treatment of neuropsychiatric symptoms.
A general approach to the symptomatic management of dementia
is presented in Chap. 25.
OTHER CAUSES OF DEMENTIA
FTD (Chap. 432), vascular dementia (Chap. 433), DLB (Chap. 434),
and prion diseases (Chap. 438) are covered in dedicated chapters.
Prion diseases such as CJD are rare neurodegenerative conditions
(prevalence ~1 per million) that produce dementia. CJD is a rapidly
progressive disorder associated with dementia, focal cortical signs,
rigidity, and myoclonus, causing death <1 year after first symptoms
appear. The rapidity of progression seen with CJD is uncommon in AD
so that the distinction between the two disorders is usually straightforward, although AD can on occasion present as a rapidly progressive
dementia. In general, corticobasal degeneration (CBD) (Chap. 432)
and DLB (Chap. 426), more rapid degenerative dementias with
prominent movement abnormalities, are more likely to be mistaken
for CJD. The differential diagnosis for CJD includes other rapidly progressive dementing conditions such as viral or bacterial encephalitides,
Hashimoto’s encephalopathy, central nervous system (CNS) vasculitis,
lymphoma, or paraneoplastic/autoimmune syndromes (Chap. 94). The
markedly abnormal periodic complexes on EEG and cortical ribboning
and basal ganglia hyperintensities on diffusion-weighted imaging or
fluid-attenuated inversion recovery MRI are diagnostic features of CJD,
although rarely, prolonged focal or generalized seizures can produce a
similar imaging appearance.
Huntington’s disease (HD) (Chap. 436) is an autosomal dominant
degenerative brain disorder. Clinical hallmarks of HD include chorea,
behavioral disturbance, and executive impairment. Symptoms typically
begin in the fourth or fifth decade of life, but there is a wide range, from
childhood to >70 years. Memory is frequently not impaired until late
in the disease, but attention, judgment, self-awareness, and executive
functions are often deficient at an early stage. Depression, apathy, social
withdrawal, irritability, and intermittent disinhibition are common.
Delusions and obsessive-compulsive behavior may occur. Disease
duration is variable but typically lasts ~15 years.
Normal-pressure hydrocephalus is a relatively uncommon but treatable syndrome. The clinical, physiologic, and neuroimaging characteristics of NPH must be carefully distinguished from those of other
dementias associated with gait impairment. Historically, many patients
treated for NPH have suffered from other dementias, particularly AD,
vascular dementia, DLB, and progressive supranuclear palsy (PSP)
(Chap. 432). For NPH, the clinical triad includes an abnormal gait
(ataxic or apractic), dementia (usually mild to moderate, with an
emphasis on executive impairment), and urinary urgency or incontinence. Neuroimaging reveals enlarged lateral ventricles (hydrocephalus) with little or no cortical atrophy, although the Sylvian fissures may
appear propped open (so-called boxcarring), which can be mistaken
for perisylvian atrophy. Crowding of dorsal frontal-parietal gyri helps
distinguish NPH from other movement disorders, such as PSP and
CBD, in which dorsal atrophy with sulcal widening is common. NPH
is a communicating hydrocephalus with a patent aqueduct of Sylvius
3376 PART 13 Neurologic Disorders
(see Fig. 29-3), in contrast to aqueductal stenosis, in which the
aqueduct is small. Lumbar puncture opening pressure falls in the
high-normal range, and the CSF protein, glucose, and cell counts are
normal. NPH may be caused by obstruction to normal CSF flow over
the cerebral convexities and delayed resorption into the venous system.
The indolent nature of the process results in enlarged lateral ventricles with relatively little increase in CSF pressure. Presumed edema,
stretching, and distortion of subfrontal white matter tracts may lead to
clinical symptoms, but the precise underlying pathophysiology remains
unclear. Some patients provide a history of conditions that produce meningeal scarring (blocking CSF resorption) such as previous
meningitis, subarachnoid hemorrhage, or head trauma. Others with
long-standing but asymptomatic congenital hydrocephalus may have
adult-onset deterioration in gait or memory that is confused with NPH.
In contrast to AD, the patient with NPH complains of an early and
prominent gait disturbance without cortical atrophy on CT or MRI.
Numerous attempts have been undertaken to improve NPH diagnosis with various special studies and to predict the success of ventricular
shunting. These tests include radionuclide cisternography (showing
a delay in CSF absorption over the convexity) and various efforts to
monitor and alter CSF flow dynamics, including a constant-pressure
infusion test. None has proven to be specific or consistently useful. A
transient improvement in gait or cognition may follow lumbar puncture (or serial punctures) with removal of 30–50 mL of CSF, but this
finding has also not proved to be consistently predictive of postshunt
improvement. Perhaps the most reliable strategy is a period of close
inpatient evaluation before, during, and after lumbar CSF drainage.
Occasionally, when a patient with AD presents with gait impairment
(at times due to comorbid subfrontal vascular injury) and absent or
only mild cortical atrophy on CT or MRI, distinguishing NPH from
AD can be challenging. Hippocampal atrophy on MRI favors AD,
whereas a characteristic “magnetic” gait with external hip rotation, low
foot clearance, and short strides, along with prominent truncal sway or
instability, favors NPH. The diagnosis of NPH should be avoided when
hydrocephalus is not detected on imaging studies, even if the symptoms otherwise fit. Of those patients identified by careful diagnosis as
having NPH, 30–50% will improve with ventricular shunting. Gait may
improve more than cognition, but many reported failures to improve
cognitively may have resulted from comorbid AD. Importantly, the
presence of positive CSF AD biomarkers or amyloid PET is associated
with lower likelihood of response to shunting. Short-lasting improvement is common. Patients should be carefully selected for shunting,
because subdural hematoma, infection, and shunt failure are known
complications and can be a cause for early nursing home placement in
an elderly patient with previously mild dementia.
Intracranial hypotension, sometimes called sagging brain syndrome,
is a disorder caused by low CSF pressure, leading to downward pressure on the subcortical structures and disruption of cerebral function.
It presents in a variable manner with headache, often exacerbated by
coughing or a Valsalva maneuver or by moving from lying to standing.
Other common symptoms include dizziness, vomiting, disruption of
sleep-wake cycles, and sometimes a progressive behavioral variant
FTD-like syndrome (Chap. 432). Although sometimes idiopathic, this
syndrome can be caused by CSF leaks secondary to lumbar puncture,
head trauma, or spinal cord arachnoid cysts. Treatment consists of
finding and patching the CSF leak.
Dementia can accompany chronic alcoholism (Chap. 453) and
may result from associated malnutrition, especially of B vitamins,
particularly thiamine. Other poorly defined aspects of chronic alcoholism may, however, also produce cerebral damage. A rare idiopathic
syndrome of dementia and seizures with degeneration of the corpus
callosum has been reported primarily in male Italian red wine drinkers
(Marchiafava-Bignami disease).
Thiamine (vitamin B1
) deficiency causes Wernicke’s encephalopathy (Chap. 307). The clinical presentation is usually a malnourished
patient (frequently but not necessarily alcoholic) with confusion,
ataxia, and diplopia resulting from inflammation and necrosis of
periventricular midline structures, including dorsomedial thalamus,
mammillary bodies, midline cerebellum, periaqueductal gray matter,
and trochlear and abducens nuclei. Damage to the dorsomedial thalamus correlates most closely with the memory loss. Prompt administration of parenteral thiamine (100 mg intravenously for 3 days followed
by daily oral dosage) may reverse the disease if given within the first
days of symptom onset. Prolonged untreated thiamine deficiency can
result in an irreversible and profound amnestic syndrome (Korsakoff ’s
syndrome) or even death.
In Korsakoff’s syndrome, the patient is unable to recall new information despite normal immediate memory, attention span, and level of
consciousness. Memory for new events is seriously impaired, whereas
knowledge acquired prior to the illness remains relatively intact.
Patients are easily confused, disoriented, and cannot store information
for more than a few minutes. Superficially, they may be conversant,
engaging, and able to perform simple tasks and follow immediate commands. Confabulation is common, although not always present. There
is no specific treatment because the previous thiamine deficiency has
produced irreversible damage to the medial thalamic nuclei and mammillary bodies. Mammillary body atrophy may be visible on MRI in the
chronic phase (see Fig. 307-6).
Vitamin B12 deficiency, as can occur in pernicious anemia, causes
a megaloblastic anemia and may also damage the nervous system
(Chaps. 99 and 442). Neurologically, it most commonly produces a
spinal cord syndrome (myelopathy) affecting the posterior columns
(loss of vibration and position sense) and corticospinal tracts (hyperactive tendon reflexes with Babinski signs); it also damages peripheral
nerves (neuropathy), resulting in sensory loss with depressed tendon
reflexes. Damage to myelinated axons may also cause dementia. The
mechanism of neurologic damage is unclear but may be related to
a deficiency of S-adenosyl methionine (required for methylation of
myelin phospholipids) due to reduced methionine synthase activity
or accumulation of methylmalonate, homocysteine, and propionate,
providing abnormal substrates for fatty acid synthesis in myelin.
Use of histamine blockers or metformin, vegan diets, autoimmunity
against gastric parietal cells, and various causes of malabsorption are
the typical causes for vitamin B12 deficiency. The neurologic sequelae
of vitamin B12 deficiency may occur in the absence of hematologic
manifestations, making it critical to avoid using the complete blood
count (CBC) and blood smear as a substitute for measuring B12 blood
levels. Treatment with parenteral vitamin B12 (1000 μg intramuscularly
daily for a week, weekly for a month, and monthly for life for pernicious anemia) stops progression of the disease if instituted promptly,
but complete reversal of advanced nervous system damage will not
occur.
Deficiency of nicotinic acid (pellagra) is associated with skin rash
over sun-exposed areas, glossitis, and angular stomatitis (Chap. 333).
Severe dietary deficiency of nicotinic acid along with other B vitamins
such as pyridoxine may result in spastic paraparesis, peripheral neuropathy, fatigue, irritability, and dementia. This syndrome has been seen
in prisoners of war and in concentration camps but should be considered in any malnourished individual. Low serum folate levels appear to
be a rough index of malnutrition, but isolated folate deficiency has not
been proved as a specific cause of dementia.
CNS infections usually cause delirium and other acute neurologic
syndromes. However, some chronic CNS infections, particularly
those associated with chronic meningitis (Chap. 139), may produce
a dementing illness. The possibility of chronic infectious meningitis should be suspected in patients presenting with a dementia or
behavioral syndrome, who also have headache, meningismus, cranial
neuropathy, and/or radiculopathy. Between 20–30% of patients in
the advanced stages of HIV infection become demented (Chap. 202).
Cardinal features include psychomotor retardation, apathy, and
impaired memory. This syndrome may result from secondary opportunistic infections but can also be caused by direct infection of CNS
neurons with HIV. Neurosyphilis (Chap. 182) was a common cause of
dementia in the preantibiotic era; it is now uncommon but can still be
encountered in patients with multiple sex partners, particularly among
patients with HIV. Characteristic CSF changes consist of pleocytosis,
increased protein, and a positive Venereal Disease Research Laboratory
(VDRL) test.
3377Alzheimer’s Disease CHAPTER 431
Primary and metastatic neoplasms of the CNS (Chap. 90) usually
produce focal neurologic findings and seizures rather than dementia,
but if tumor growth begins in the frontal or temporal lobes, the initial
manifestations may be memory loss or behavioral changes. An autoimmune, sometimes paraneoplastic, syndrome of dementia associated
with occult carcinoma (often small-cell lung cancer) is termed limbic
encephalitis. In this syndrome, confusion, agitation, seizures, poor
memory, emotional changes, and frank dementia may occur. Paraneoplastic encephalitis associated with N-methyl-d-aspartate (NMDA)
receptor antibodies presents as a progressive psychiatric disorder with
memory loss and seizures; affected patients are often young women
with ovarian teratoma. Autoimmune etiologies also include antibodies targeting leucine-rich glioma-inactivated 1 (LGI1; faciobrachial
dystonic seizures); contactin-associated protein-like 2 (Caspr2; insomnia, ataxia, myotonia); and α-amino-3-hydroxy-5-methylisoxazole-4-
propionic acid (AMPA)-receptor (limbic encephalitis with relapses),
among others (Chap. 94).
A nonconvulsive seizure disorder (Chap. 425) may underlie a syndrome of confusion, clouding of consciousness, and garbled speech.
Often, psychiatric disease is suspected, but an EEG demonstrates the
epileptic nature of the illness. If recurrent or persistent, the condition
may be termed complex partial status epilepticus. The cognitive disturbance often responds to anticonvulsant therapy. The etiology may
be previous small strokes or head trauma; some cases are idiopathic.
Nonconvulsive temporal lobe seizures can also emerge early in the
course of AD.
It is important to recognize systemic diseases that indirectly affect
the brain and produce chronic confusion or dementia. Such conditions
include hypothyroidism; vasculitis; and hepatic, renal, or pulmonary
disease. Hepatic encephalopathy may begin with irritability and confusion and slowly progress to agitation, lethargy, and coma.
Isolated vasculitis of the CNS (CNS granulomatous angiitis)
(Chaps. 363 and 427) occasionally causes a chronic encephalopathy
associated with confusion, disorientation, and clouding of consciousness. Headache is common, and strokes and cranial neuropathies
may occur. Brain imaging studies may be normal or nonspecifically
abnormal. CSF analysis reveals a mild pleocytosis or protein elevation.
Cerebral angiography can show multifocal stenoses involving mediumcaliber vessels, but some patients have only small-vessel disease that
is not revealed on angiography. The angiographic appearance is not
specific and may be mimicked by atherosclerosis, infection, or other
causes of vascular disease. Brain or meningeal biopsy demonstrates
endothelial cell proliferation and mononuclear infiltrates within blood
vessel walls. The prognosis is often poor, although the disorder may
remit spontaneously. Some patients respond to glucocorticoids or
chemotherapy.
Chronic metal exposure represents a rare cause of dementia. The
key to diagnosis is to elicit a history of exposure at work or home.
Chronic lead poisoning from inadequately fire-glazed pottery has been
reported. Fatigue, depression, and confusion may be associated with
episodic abdominal pain and peripheral neuropathy. Gray lead lines
appear in the gums, usually accompanied by an anemia with basophilic
stippling of red blood cells. The clinical presentation can resemble that
of acute intermittent porphyria (Chap. 416), including elevated levels
of urine porphyrins as a result of the inhibition of δ-aminolevulinic
acid dehydrase. The treatment is chelation therapy with agents such as
ethylenediamine tetraacetic acid (EDTA). Chronic mercury poisoning
produces dementia, peripheral neuropathy, ataxia, and tremulousness
that may progress to a cerebellar intention tremor or choreoathetosis.
The confusion and memory loss of chronic arsenic intoxication is also
associated with nausea, weight loss, peripheral neuropathy, pigmentation and scaling of the skin, and transverse white lines of the fingernails
(Mees’ lines). Treatment is chelation therapy with dimercaprol (British
anti-Lewisite, BAL). Aluminum poisoning is rare but was documented
with the dialysis dementia syndrome, in which water used during renal
dialysis was contaminated with excessive amounts of aluminum. This
poisoning resulted in a progressive encephalopathy associated with
confusion, nonfluent aphasia, memory loss, agitation, and, later, lethargy and stupor. Speech arrest and myoclonic jerks were common and
associated with severe and generalized EEG changes. The condition has
been eliminated by the use of deionized water for dialysis.
Recurrent head trauma in professional athletes may lead to a
dementia previously referred to as “punch-drunk” syndrome or
dementia pugilistica but now known as chronic traumatic encephalopathy (CTE) to signify its relevance to contact sport athletes other
than boxers (Chap. 443). The symptoms can be progressive, beginning
late in an athlete’s career or, more often, after retirement. Early in the
course, a personality change occurs, associated with social instability,
explosive rage, and sometimes paranoia and delusions. Later, memory
loss progresses to full-blown dementia, often associated with parkinsonian signs and ataxia or intention tremor. At autopsy, the cerebral
cortex shows tau-immunoreactive NFTs that are more prominent than
amyloid plaques (which are usually diffuse or absent rather than neuritic). NFTs and tau-positive reactive astrocytes are often clustered in
the depths of cortical sulci and in a perivascular distribution. TDP-43
inclusions (Chap. 432) have also been reported, highlighting the overlap with the FTD spectrum. Loss of neurons in the substantia nigra is a
variable feature, and some with TDP-43 inclusions also develop motor
neuron disease (MND) (Chap. 437).
Chronic subdural hematoma (Chap. 443) is also occasionally
associated with dementia, often in the context of underlying cortical
atrophy from conditions such as AD or HD.
Transient global amnesia (TGA) is characterized by the sudden onset
of a severe episodic memory deficit, usually occurring in persons aged
>50 years. Often the amnesia occurs in the setting of an emotional
stimulus or physical exertion. During the attack, the individual is alert
and communicative, general cognition seems intact, and there are no
other neurologic signs or symptoms. The patient may seem confused
and repeatedly ask about his or her location in place and time. The
ability to form new memories returns after a period of hours, and
the individual returns to normal with no recall for the period of the
attack. Frequently no cause is determined, but cerebrovascular disease,
epilepsy (7% in one study), migraine, or cardiac arrhythmias have all
been implicated. Approximately one-quarter of patients experience
recurrent attacks. Rare instances of permanent memory loss have been
reported in patients with TGA-like spells, usually representing ischemic infarction of the hippocampus or dorsomedial thalamic nucleus
bilaterally. Seizure activity due to AD should always be suspected in
this syndrome.
The ALS/parkinsonian/dementia complex of Guam is a rare degenerative disease that has occurred in the Chamorro natives on the island
of Guam. Individuals may have any combination of parkinsonian features, dementia, and MND. The most characteristic pathologic features
are the presence of NFTs in degenerating neurons of the cortex and
substantia nigra and loss of motor neurons in the spinal cord, although
recent reanalysis has shown that some patients with this illness also
show coexisting TDP-43 pathology. Epidemiologic evidence supports
a possible environmental cause, such as exposure to a neurotoxin or
an infectious agent with a long latency period. One interesting but
unproven candidate neurotoxin is the seed of the false palm tree, which
Guamanians traditionally used to make flour. The amyotrophic lateral
sclerosis (ALS) syndrome is no longer present in Guam, but a dementing illness with rigidity continues to be seen.
Rarely, adult-onset leukodystrophies, lysosomal-storage diseases,
and other genetic disorders can present as a dementia in middle to late
life. Metachromatic leukodystrophy (MLD) causes a progressive psychiatric or dementia syndrome associated with an extensive, confluent
frontal white matter abnormality. MLD is diagnosed by measuring
reduced arylsulfatase A enzyme activity in peripheral white blood
cells. Adult-onset presentations of adrenoleukodystrophy have been
reported in female carriers, and these patients often feature spinal
cord and posterior white matter involvement. Adrenoleukodystrophy
is diagnosed by demonstrating increased levels of plasma very-longchain fatty acids. CADASIL is another genetic syndrome associated
with white matter disease, often frontally and temporally predominant.
Diagnosis is made with skin biopsy, which shows osmophilic granules
in arterioles, or increasingly through genetic testing for mutations
in Notch 3. The neuronal ceroid lipofuscinoses are a genetically
3378 PART 13 Neurologic Disorders
Frontotemporal dementia (FTD) refers to a group of clinical syndromes
united by their links to underlying frontotemporal lobar degeneration
(FTLD) pathology. FTD, like the other major neurodegenerative diseases, is considered a disease of abnormal protein aggregation, with
either tau or transactive response DNA-binding protein of 43 kDa
(TDP-43) implicated in most cases. FTD most often begins in the fifth
to seventh decades of life and is nearly as prevalent as Alzheimer’s
disease (AD) in this age group. Early studies suggested that FTD may
be more common in men than women; however, more recent reports
cast doubt on this finding. Although a family history of dementia is
common, autosomal dominant inheritance is seen in only 10–20% of
all FTD cases.
■ CLINICAL MANIFESTATIONS
Familial and sporadic forms of FTLD present with remarkable clinical
heterogeneity. Three core clinical syndromes have been described
(Fig. 432-1). In the behavioral variant (bvFTD), the most common
FTD syndrome, social and emotional dysfunction manifests as apathy,
disinhibition, compulsivity, loss of empathy, and overeating, often but
not always accompanied by deficits in executive control. Two forms
of primary progressive aphasia (PPA), the semantic and nonfluent/
agrammatic variants, are commonly due to FTLD and are included
under the FTD umbrella. In the semantic variant, patients slowly lose
the ability to decode word, object, person-specific, and emotion meaning, whereas patients with the nonfluent/agrammatic variant develop
profound inability to produce words, often with prominent motor
speech impairment. Any of these three clinical syndromes, but most
often bvFTD, may be accompanied by motor neuron disease (MND)
(Chap. 437), in which case the term FTD-MND is applied. In addition, the corticobasal syndrome (CBS) and progressive supranuclear
palsy–Richardson syndrome (PSP-RS) can be considered part of the
FTLD clinical spectrum. Furthermore, patients may evolve from any
of the major syndromes described above to have prominent features of
another syndrome.
Findings at the bedside are dictated by the anatomic localization
of the disorder. Degeneration with atrophy occurs in the medial and
orbital frontal and anterior insula in bvFTD; the anterior temporal
region in semantic variant PPA; and the lateral frontal and precentral
gyrus of the dominant hemisphere in nonfluent/agrammatic PPA.
Typically, parietal functions such as visuospatial processing and arithmetic calculations are unaffected even late in the FTD syndromes.
Many patients with nonfluent aphasia or bvFTD later develop aspects
of PSP-RS as disease spreads into subcortical or brainstem structures, or CBS-like features appear as disease moves into perirolandic
cortices.
432 Frontotemporal Dementia
William W. Seeley, Bruce L. Miller
heterogeneous group of disorders associated with myoclonus, seizures,
vision loss, and progressive dementia. Diagnosis is made by finding
eosinophilic curvilinear inclusions within white blood cells or neuronal tissue.
Psychogenic amnesia for personally important memories can be
seen. Whether this results from deliberate avoidance of unpleasant
memories, outright malingering, or unconscious repression remains
unknown and probably depends on the patient. Event-specific amnesia
is more likely to occur after violent crimes such as homicide of a close
relative or friend or sexual abuse. It may develop in association with
severe drug or alcohol intoxication and sometimes with schizophrenia.
More prolonged psychogenic amnesia occurs in fugue states that also
commonly follow severe emotional stress. The patient with a fugue
state suffers from a sudden loss of personal identity and may be found
wandering far from home. In contrast to neurologic amnesia, fugue
states are associated with amnesia for personal identity and events closely
associated with the personal past. At the same time, memory for other
recent events and the ability to learn and use new information are preserved. The episodes usually last hours or days and occasionally weeks
or months while the patient takes on a new identity. On recovery, there
is a residual amnesia gap for the period of the fugue. Very rarely does
selective loss of autobiographic information reflect a focal injury to the
brain areas involved with these functions.
Psychiatric diseases may mimic dementia. Severely depressed or
anxious individuals may appear demented, a phenomenon sometimes
called pseudodementia. Memory and language are usually intact when
carefully tested, and a significant memory disturbance usually suggests
an underlying dementia, even if the patient is depressed. Patients in
this condition may feel confused and unable to accomplish routine
tasks. Vegetative symptoms, such as insomnia, lack of energy, poor
appetite, and concern with bowel function, are common. Onset is
often more abrupt, and the psychosocial milieu may suggest prominent reasons for depression. Such patients respond to treatment of the
underlying psychiatric illness. Schizophrenia is usually not difficult
to distinguish from dementia, but occasionally the distinction can be
problematic. Schizophrenia generally has a much earlier age of onset
(second and third decades of life) than most dementing illnesses and
is associated with intact memory. The delusions and hallucinations
of schizophrenia are usually more complex, bizarre, and threatening
than those of dementia. Some chronic schizophrenics develop an
unexplained progressive dementia late in life that is not related to AD.
Conversely, FTD, HD, vascular dementia, DLB, AD, or leukoencephalopathy can begin with schizophrenia-like features, leading to the
misdiagnosis of a psychiatric condition. Later age of onset, significant
deficits on cognitive testing, or the presence of abnormal neuroimaging suggest a degenerative condition. Memory loss may also be part of
a conversion disorder. In this situation, patients commonly complain
bitterly of memory loss, but careful cognitive testing either does not
confirm the deficits or demonstrates inconsistent or unusual patterns
of cognitive problems. The patient’s behavior and “wrong” answers to
questions often indicate that he or she understands the question and
knows the correct answer.
Clouding of cognition by chronic drug or medication use, often prescribed by physicians, is an important cause of dementia. Sedatives,
tranquilizers, and analgesics used to treat insomnia, pain, anxiety, or
agitation may cause confusion, memory loss, and lethargy, especially
in the elderly. Discontinuation of the offending medication often
improves mentation.
■ FURTHER READING
Andrews SJ et al: Interpretation of risk loci from genome-wide
association studies of ‘Alzheimer’s disease. Lancet Neurol 19:326,
2020.
Belloy ME et al: A quarter century of APOE and Alzheimer’s disease:
Progress to date and the path forward. Neuron 101:820, 2019.
Braak H, Del Tredici K: Where, when, and in what form does sporadic Alzheimer’s disease begin? Curr Opin Neurol 25:708, 2012.
Jack CR et al: NIA-AA Research Framework: Toward a biological
definition of Alzheimer’s disease. Alzheimers Dement 14:535, 2018.
Lesman-Segev OH et al: Diagnostic accuracy of amyloid versus 18F-Fluorodeoxyglucose positron emission tomography in autopsyconfirmed dementia. Ann Neurol 89:389, 2021.
Long JM, Holtzman DM: Alzheimer disease: An update on pathobiology and treatment strategies. Cell 179:312, 2019.
Rabinovici GD et al: Association of amyloid positron emission
tomography with subsequent change in clinical management among
Medicare beneficiaries with mild cognitive impairment or dementia.
JAMA 321:1286, 2019.
Rabinovici GD: Late-onset Alzheimer Disease. Continuum 25:14,
2019.
Selkoe DJ, Hardy J: The amyloid hypothesis of Alzheimer’s disease at
25 years. EMBO Mol Med 8:595, 2016.
No comments:
Post a Comment
اكتب تعليق حول الموضوع