Dementias

Published on 12/04/2015 by admin

Filed under Neurology

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 2103 times

Chapter 66 Dementias

Dementia Syndrome

The term dementia is of Latin origin and means “devoid of the mind.” It is used to describe a persistent state of serious cognitive, functional, and emotional deterioration from a previously higher level of functioning and should not be confused with mental retardation, which denotes deficient cognitive development. Dementia can result from sudden significant brain insults such as strokes or trauma, infectious or metabolic disorders resulting in irreversible brain parenchymal changes, or most commonly from insidious progressive neurodegenerative processes. Regardless of the cause, all dementias have a significant impact on health and an individual’s ability to take care of him- or herself. The burden of dementia on patients’ quality of life, their relatives, and friends should not be underestimated.

Epidemiology

The exact incidence and prevalence of dementia from any cause remains unknown. The most common etiologies for the dementia syndrome are neurodegenerative disorders. Neurodegenerative dementias typically occur in late life with age; heretability; preexisting conditions, such as Parkinson disease; and vascular risk factors being the most important contributors. As the proportion of elderly individuals is rapidly increasing worldwide, dementia is becoming a highly significant global healthcare problem. Between 1997 and 2025, the elderly population—defined as persons 65 years of age and older—is projected to increase from 62.7 to 136.9 million in the Americas, from 17.7 to 37.9 million in Africa, from 112.5 to 169.8 million in Europe, from 60.5 to 166.7 million in Southeast Asia, and from 110.7 to 267.7 million in the Western Pacific Region of Asia (World Health Organization, 1998).

Recently the National Institutes of Health–funded Aging, Demographics, and Memory Study (ADAMS) published representative data of dementia prevalence in the United States (Langa et al., 2005; Plassman et al., 2007). One of ADAMS enrollment criteria was age of 70 years or older, and approximately a third of participants were older than 85 years. The study included a detailed cognitive battery (Langa et al., 2005). Diagnosis of dementia was based on Diagnostic and Statistical Manual of Mental Disorders (DSM)-III-R and DSM-IV criteria. Dementia prevalence increased from 5% among 71- to 79-year-olds to 37.4% in those 90 years and older (Plassman et al., 2007). In addition to older age, the authors reported that African American race and lower educational achievement independently contributed to increased odds of dementia syndrome of any etiology.

Diagnostic Criteria

In 2001, the Practice Parameters Subcommittee of the American Academy of Neurology (AAN) published a summary of evidence-based guidelines for early detection, diagnosis, and management of dementia (Knopman et al., 2001). These guidelines offered basic recommendations for diagnosis and both pharmacological and nonpharmacological management of dementia. The experts recommend the use of the American Psychiatric Association’s DSM criteria (the most recent being the DSM-IV) for establishing a diagnosis of dementia syndrome (Box 66.1). It should be noted, however, that despite the requirement for memory impairment in some forms of dementia, memory impairment is not an early symptom.

Increased focus on the earliest stages of dementia has led to the recognition of a state called mild cognitive impairment (MCI). MCI was initially considered a transition state between normal aging and dementia of the Alzheimer type, but the concept quickly evolved to denote an intermediate state between normal cognitive aging and any type of dementia. Practice parameters on MCI from the Quality Standards Subcommittee of the AAN have been published (Petersen et al., 2001b). MCI is an increasingly important focus of research and clinical attention. Identifying early disease—especially Alzheimer disease (AD) and vascular cognitive impairment—and initiating therapeutic interventions to prevent or slow decline are the goals. However, the concept of MCI as a clinical entity has undergone refinement and multiple modifications (Dubois and Albert, 2004; Petersen et al., 2001a, 2006; Petersen and Morris, 2005; Winblad et al., 2004) and has most recently been challenged as an artificial construct (Dubois et al., 2007). MCI is now recognized as a nonspecific state that may progress to AD or a non-AD dementia, may remain stable, or may revert to normal cognition. Many studies now attempt to define MCI of the AD type as the harbinger of AD. Because the onset of functional decline is typically late in the course of the disease, the combination of characteristic cognitive features in the presence of positive disease biomarkers has been proposed as sufficient for the diagnosis of the disease in the early predementia stages. These new criteria were proposed for use in AD (Dubois et al., 2007), but it is very likely this idea will be adopted for other dementias in the near future.

General Approach to Dementia Diagnosis

The essential feature of dementia is the acquired and persistent compromise in multiple cognitive domains that is severe enough to interfere with everyday functioning (see Box 66.1). This definition stands in contrast to delirium or acute confusional states (ACSs), which are distinguished primarily by prominent deficits or fluctuations in attentional processing. Although dementia syndromes tend to be chronic, progressive, and irreversible, and ACSs tend to be acute to subacute, fluctuating, and reversible, these distinctions are more relative than absolute. Toxic, metabolic, or other systemic physiological disturbances are more likely to be reversible which is not the case for neurodegenerative or cerebrovascular dementia syndromes. On the other hand, dementia renders the patient more vulnerable to delirium, highlighting the need for a comprehensive evaluation of potentially reversible etiological disorders in the context of even well-established dementia syndrome. Careful evaluation of persons referred for dementia evaluation can identify treatable or reversible disorders in up to 20% of cases (Hejl et al., 2002).

Distinguishing cognitive impairment associated with an underlying disorder from potentially reversible cognitive symptoms (e.g., depression, metabolic encephalopathy) often demands ongoing evaluation, including appropriate diagnostic tests and sometimes a therapeutic challenge with an antidepressant drug. Abrupt onset of thinking changes in temporal relation to a psychological stressor, poor effort on cognitive testing (particularly with demanding tasks), and prominent neurovegetative signs such as insomnia and anorexia are characteristic of depression-associated cognitive impairment. Drug-induced cognitive impairment occurring as a result of anticholinergic agents, sedative-hypnotic drugs (e.g., benzodiazepines), or opiate analgesics commonly occurs in the elderly and may cause or contribute to cognitive symptomatology. Any temporal association of cognitive decline with initiation or titration of certain drugs should prompt taper and withdrawal of the offending agent. Multifactorial etiology is common in the elderly, and the search for potentially reversible or modifiable conditions should be done with vigor and vigilance.

History

A detailed comprehensive history of the presenting illness is the single most important part of the evaluation of cognitively impaired subjects. In general, the history should be obtained from both the patient and a reliable and knowledgeable informant, because cognitive deficits, lack of insight, and lapses in judgment actively interfere with accurate self-reporting. A comprehensive history should determine the initial manifestations, mode of onset, and course over time (Box 66.2).

Box 66.2 Historical Components of the Dementia Evaluation

Insidious onset and gradual progression over time are characteristic for degenerative disorders as the cause of dementia, whereas abrupt or discrete temporal onset raises suspicion for an associated neurological condition such as a stroke or a tumor, extenuating life circumstances, or medication change. Age, cultural background, educational level, and prevailing social and occupational demands modulate the clinical expression of dementia and its clinical significance. Recognizing changes from a previous level of functioning or altered behavioral patterns may help discern subtle diagnostically relevant clinical deficits. AD typically manifests with recent memory impairment combined with variable degrees of language, visuospatial, and executive dysfunction. Initial isolated disturbances in any of the latter cognitive domains, especially in the settings of preserved episodic memory or prominent initial changes in behavior and composure, warrant considering alternative diagnoses, such as cerebrovascular disease, mass lesion, or a focal neurodegenerative condition. Early parkinsonian features prompt an evaluation for dementia with Lewy bodies (DLB), Parkinson disease dementia (PDD) or another parkinsonian dementing disorder, vascular dementia, and normal-pressure hydrocephalus. Concurrent medical conditions, history of neurological insults, and primary disturbances of sleep are other factors that have to be accounted for as possible causes of dementia or concomitant factors that exacerbate its expression.

Cognitive Assessment

Standardized assessments such as the Mini-Mental State Examination (MMSE) (Folstein et al., 1975) provide a brief focused survey of cognitive domains most often affected in AD. The MMSE is a crude assessment that offers very limited assessment of memory and executive functions, however. The Montreal Cognitive Assessment (MoCA) is another short focused test that can be easily adopted in the busy private neurologist’s practice (Nasreddine et al., 2005). In addition to the cognitive domains tapped into by the MMSE, the MoCA also includes drawing a cube, the clock-drawing test, and a shortened version of Trails B, a test of working memory, set shifting, and complex attention, as well as a more detailed verbal memory test. MoCA may be better suited for detection of cognitive changes in the predementia stage of AD and other types of dementia (Gagnon et al., 2010; Hoops et al., 2009; Nazem et al., 2009; Smith et al., 2007; Videnovic et al., 2010). Additional tests for attention, language, praxis, executive and visuomotor functioning, and abstract thinking should be used selectively at the bedside or during an office visit to augment the initial crude cognitive screen (Table 66.1). Finally, for more refined cognitive assessment, patients should be referred for an evaluation by a trained neuropsychologist. Assessment by the latter is especially informative in borderline cases when the physician is uncertain of the presence of cognitive decline beyond what is expected by aging alone or under special circumstances such as limited education.

Table 66.1 Clinical Cognitive Assessment* (Normal Ranges)

Cognitive Feature Means of Assessment
Working memory Digit span forward (7 ± 2)
Complex attention (require manipulation of items in working memory) Digit span backward (6 ± 2)Months in reverse order (15-20 seconds)
Orientation Time, location, autobiographical data
Language Confrontational naming (high- and low-frequency items)
  Verbal fluency (e.g., animals, grocery items) (18 ± 6/1 minute)
  Repetition (sentences of varying length)
  Comprehension (yes-or-no questions; performing multistep tasks)
  Reading aloud/comprehension
  Sentence writing (spontaneous, to dictation)
Visuospatial Figure copying (two- and three-dimensional figures)
  Visual scene analysis (describe whole and individual parts)
  Line bisection (place “X” in center of horizontal line)
  Clock drawing (draw clock face with hands set to “10 after 11”)
Verbal recent memory Word list (3-5 words): immediate/delayed/recognition recall
  Paragraph recall
Nonverbal recent memory Figure copy recall
Remote memory Historical events (family milestones, recent presidents)
Abstract conceptualization Similarities (rose/tulip, poem/statue); proverb interpretation
Praxis Have patient demonstrate saluting a flag, hammering a nail
Sequencing Graphomotor sequencing (have patient draw a simple alternating pattern)
  Luria gestures (sequentially alternating fist, side, palm)

* Further details regarding administration and interpretation can be found in Hodges, J.R., 1994. Cognitive Assessment for Clinicians. Oxford University Press, Oxford.

Neuropsychiatric Assessment

Neuropsychiatric symptoms are exceedingly common among the cognitively impaired. Mood state (depressed, euphoric), vegetative status (eating, sleeping), changes in personality (apathetic, disinhibited), and alterations in perception (hallucinations) or thought (delusions) are major areas to be probed in the course of diagnostic assessment. The presence or absence of certain neuropsychiatric symptoms may guide the physician in the differential diagnosis. For instance, while apathy is exceedingly common among all neurodegenerative dementias, euphoria is only rarely seen in AD or DLB but common in frontal-variant frontotemporal dementia (fvFTD). Early hallucinations, delusions, and fluctuations, which can also predate the onset of cognitive decline, point to DLB. Attention to these and other neuropsychiatric concomitants of dementia is important because they represent treatable components of excess morbidity and often have a profound impact on caregiver burden and the need for institutionalization (Kaufer et al., 1998).

Differentiating between primary neurodegenerative disorder with concomitant depressive symptoms and primary depressive disorder that can result in pseudodementia can be challenging. Good history taking is essential and sometimes a trial of antidepressants is warranted. It is also very important to distinguish apathy (loss of interest or motivation) from primary depression on the basis of neutral affect and the absence of vegetative signs.

Although structured interviews for neuropsychiatric assessment in dementia have been developed, their use is restricted primarily to research settings. Recently a brief questionnaire version of the Neuropsychiatric Inventory (NPI) (Cummings, 1997) was designed for the practice setting. The Neuropsychiatric Inventory Questionnaire (NPIQ) (Kaufer et al., 2000) is relatively easy to administer and can provide a clinical screening examination for common neuropsychiatric manifestations of dementia and their associated impact on caregivers.

Neuroimaging

Based on evidence-based methodology, the AAN guidelines for diagnostic evaluation of dementia (Knopman et al., 2001) recommended the use of neuroimaging to screen patients with cognitive impairment. These recommendations resulted from the findings of one class II study showing that 5% of all patients with cognitive complaints harbored a causative nondegenerative lesion such as a slow-growing brain neoplasm (most commonly of the frontal lobes), subdural hematoma (SDH), or normal-pressure hydrocephalus (NPH) (Chui and Zhang, 1997). Inasmuch as the vast majority of MCI patients harbor neurodegenerative pathology, it is sensible to extend the guidelines for diagnostic imaging to that patient cohort as well. Justification for a structural imaging evaluation of the cognitively impaired individual is twofold: it can help detect a potentially treatable disorder in need of urgent targeted intervention, and it can identify vascular ischemic comorbidity that could either be the source of or a contributing factor to cognitive decline. Magnetic resonance imaging (MRI) is preferred, but in those instances where MRI technology is not available or when an MRI is contraindicated (for instance in patients in pacemakers), computed tomography (CT) should be used.

Although several other neuroimaging techniques and analytic approaches are vigorously being developed as candidate diagnostic and prognostic biomarkers, none of them are yet ready for routine clinical use. The scientific advances based on these technologies will be discussed in detail in the specific disorders sections that follow.

Neurodegenerative Dementias

Alzheimer Disease

AD was originally described in 1907 by the German psychiatrist and neuropathologist, Alois Alzheimer. The index case was a 51-year-old woman with paranoid delusions, progressive memory impairment, and subsequent progressive aphasia. At autopsy, Alzheimer noted brain atrophy, and as he applied the newly available silver stains, he uncovered the senile plaques (now termed neuritic or amyloid plaques) consisting of dystrophic neurites clustered around what subsequently was revealed to be a central amyloid core. He also described a second deposition, the neurofibrillary tangle, consisting of intraneuronal staining in a fibrillar pattern. In honor of Alois Alzheimer, Kraepelin subsequently named the condition Alzheimer’s disease. What initially was considered a presenile condition was later appreciated to be far more common in the elderly after 65 years of age, and with the age distinction removed, AD became the most common neurodegenerative disorder and one of the most common diseases of the aging population. It is now listed as the fifth most common cause of death (Alzheimer’s Association, 2010).

Advancing knowledge of the clinical onset and progression, neuroimaging, genetics, neurochemistry and neuropathological cascades, and molecular biology of neuritic plaques and neurofibrillary tangles has set the stage for current and future therapeutic interventions. Autosomal dominant AD has been traced to three genetic mutations in the amyloid precursor protein (APP) gene and the presenilin-1 and presenilin-2 (PS1, PS2) genes. These cases typically affect the middle-aged population. The roles of genetic risk factors for the more common sporadic late-onset disease are the subject of intense research. Inheritance of the E4 allele of the apolipoprotein E (ApoE) gene remains the most established genetic risk factor for AD. Many other candidates are under study, consonant with advances in collecting and analyzing population samples and in human genome screening and gene identification. Improved understanding of the pathological mechanisms will be the basis for future therapies.

Diagnostic Criteria

The most commonly used and widely accepted criteria for dementia of the Alzheimer type are the National Institute of Aging and Stroke–Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA) criteria (McKhann et al., 1984). While these criteria are very useful and well operationalized for the dementia state of AD, the accumulating evidence of a long prodromal (or latent) AD state has challenged our former understanding of AD and resulted in incredibly insightful research into the MCI state and more recently into prodromal AD. The AD field is now rapidly moving toward early and presymptomatic AD diagnosis that will rely heavily on disease biomarkers as outlined in the Dubois criteria for prodromal AD (Dubois et al., 2007) (Box 66.3). As such, AD has moved from being a “diagnosis of exclusion,” post documenting normal findings on a battery of laboratory studies, to a specific diagnosis based on clinical course and a characteristic pattern (or evolving pattern) of neuropsychological deficits in the presence of disease-associated biomarkers.

Box 66.3 Dubois Diagnostic Criteria for Alzheimer Disease

Epidemiology

AD is the most common cause of dementia worldwide. ADAMS recently published nationally representative data of dementia prevalence in the United States (Langa et al., 2005; Plassman et al., 2007). Diagnosis of dementia of the Alzheimer type was based on the NINCDS-ADRDA criteria. Overall dementia of the Alzheimer type accounted for 70% of dementia cases, ranging from 47% among those aged 71 to 75 years to 80% in the age 90+ group. The ADAMS data revealed that as of 2002, there were 3.4 million dementia cases in the United States (95% confidence interval [CI], 2.8-4.0), of whom 2.4 million (95% CI, 1.9-2.9) were due to AD. In addition to older age, the authors reported that African American race, lower educational achievement, and ApoE E4 genotype independently contributed to increased odds of dementia syndrome of any etiology (Plassman et al., 2007). In a following publication (Plassman et al., 2008) the ADAMS investigators reported that an estimated 22.2% (or 5.4 million Americans) 71 years or older have cognitive impairment in the absence of overt dementia. Of these, 2 million subjects were reported to have prodromal AD, defined as cognitive impairment without dementia but with a pattern of clinical symptoms or performance on neuropsychological testing suggestive of prodromal AD and no other medical or neuropsychiatric condition present to preclude an eventual diagnosis of AD. The annualized rate of progression to dementia of the Alzheimer type in prodromal AD subjects was substantially higher than the rate of progression to dementia among all cognitively impaired (17%-20% per year in prodromal AD versus 12% in cognitive impairment but no dementia from any cause). Among those who progressed to dementia, 83% were diagnosed with dementia of the Alzheimer type, 16.7% were diagnosed with vascular dementia, and 0.4% with dementia of undetermined cause (Plassman et al., 2008).

Based on our discussion thus far, it should not be surprising that advancing age is the most powerful risk factor for the development of AD. Other risk factors include the presence of one or more of the following: ApoE E4 allele in a given individual, lower education level, family history of AD, and cardiovascular risk factors (Bendlin et al., 2010; Rocchi et al., 2009; Steptoe et al., 2011). Prevalence but not incidence appears to be higher in women than in men, suggesting that susceptibility is similar, but duration of survival is greater in women. Midlife hypertension, elevated homocysteine, and elevated fat in the diet (the latter perhaps in combination with presence of an ApoE E4 allele) also are reported risk factors (Bendlin et al., 2010).

Clinical Presentation

AD is a progressive disorder of recent episodic memory, language, visuospatial function, and executive function associated with high frequency of neurobehavioral abnormalities at some point in the course. Onset of AD usually is in late life. If onset age is younger than 45 years, an autosomal dominant pedigree usually is involved. Most commonly, a PS1 mutation is the cause. In elderly individuals presenting with mild memory loss, the diagnosis of prodromal AD could be somewhat challenging owing to the presence of comorbid conditions that may alter cognition (e.g., congestive heart failure, cerebrovascular disease) or if medications for such conditions have anticholinergic side effects. Serial follow-up evaluations may be necessary to establish decline and a characteristic pattern of dementia. Nonetheless, if memory is not a prominent early complaint, other potential causes of dementia should be considered. In unselected series, diagnostic accuracy has been relatively low when tested against autopsy diagnosis. Using established diagnostic criteria for AD such as those of the NINCDS/ADRDA in conjunction with a standardized evaluation, clinical diagnostic accuracy of about 90% can be achieved (Knopman et al., 2001), accuracy approaching 90% can be anticipated, and few if any “treatable causes” are missed.

Memory Loss

Memory impairment is a sine qua non in the diagnosis of AD. Although AD can manifest with neuropsychological deficits other than memory (e.g., aphasia, abnormal executive function, apathy or other personality change), memory tasks in general are the earliest deficits noted. Memory is a complex cognitive function involving several anatomical systems underlying different types of memory. Terminology for memory is varied, and definitions are not always interchangeable.

AD is classically associated with episodic memory impairment. Episodic memory is the memory store for personal experiences that occur in a particular spatial and temporal context. Unlike working memory (see later discussion), episodic memory does not have to be actively maintained (or “rehearsed”) for successful retrieval. Episodic memory is dependent on a number of neocortical structures, but the medial temporal lobes, particularly the hippocampus, play a central role. AD even in its earliest stages prominently impairs episodic memory, particularly for recent events. Recent episodic memory is assessed by delayed recall (i.e., remembering material over intervals longer than 30 seconds to a few minutes, during which an intervening distracting task is given to prevent active rehearsal). Other tests of episodic memory include information about orientation (e.g., date and location) and current events. Deficits in episodic memory can only be definitively established in the context of sufficiently preserved attention and concentration. For instance, distractibility and inattention can prevent the delirious patient from successfully encoding any incoming information, and he or she will fail a later recall test. Such deficits should not be attributed to AD-type pathology until the patient is retested in a more lucid state.

Early in the disease course of AD, distant episodic memories (i.e., memories before disease onset) tend to be spared. This somewhat paradoxical phenomenon may be accounted for by neocortical consolidation of episodic memories over time, which renders old memories less dependent on hippocampal function. Although detailed examination of remote memory may reveal some impairment, especially later in the disease course, the brunt of the early symptomatology lies in recent episodic memory.

Working memory is a limited-capacity storage system in which internal representations are actively held “on line.” Working memory can be divided into two systems: one responsible for maintenance or storage of information, and a second central executive system responsible for coordinating information processing and manipulation. Tasks that involve simple rehearsal of items, such as keeping a phone number “on line” (or forward digit span) and word and block span, appear to require both inferior prefrontal cortex for maintenance and more posterior cortical regions for storage. Working memory tasks that require manipulation of items, also known as complex attention tasks (e.g., digit span backwards, letter/number sequencing), require a more diffuse network including the dorsolateral prefrontal cortex. Mild impairment in these complex attention tasks may be noted relatively early in the course of AD, whereas the more simple working memory tasks tend not to be impaired until later in the disease course.

A third type of memory, semantic memory (Tulving, 1987, 1992), is defined as our knowledge of facts about the world and is not associated with a spatiotemporal context of the learning event. For example, while we know that a tiger has stripes and that Washington DC is the capital of the United States, we do not recall the context in which we learned these pieces of information. Although episodic memory impairment is more prominent in AD, semantic memory is also impaired relatively early in the disease course. Some of the language impairment seen in AD relates to disintegration of semantic memory. A common test of semantic memory is the category fluency test, when the patient is asked to name as many items as possible from a given category such as animals or vegetables. Semantic memory can be reduced in the early stages of AD.

Finally, procedural memory, which depends on the implicit learning of tasks and actions, is usually preserved in AD. AD patients are capable of procedural or implicit learning in the absence of any apparent parallel declarative learning.

Aphasia, Apraxia, and Visuospatial Impairment

Language disturbance, especially verbal fluency and word finding, generally is an early feature of AD. In some patients, aphasia may be a prominent early feature, with more widespread cognitive disturbance occurring later. A helpful diagnostic aid in AD is that naming in semantic categories (e.g., animals) is more impaired than orthographically constrained tasks (e.g., words starting with a particular letter). This finding has been attributed to a breakdown in semantic memory. In practice, this means that patients with AD will produce more words beginning with a given letter (phonemic fluency task) than animals (semantic fluency task). This may be particularly useful in differentiating early AD from the effects of depression, in which the opposite pattern is seen. Of note, a reduction in category fluency also occurs with aging, so that in the “oldest old” (>80 years), it may be less informative in relation to the presence or absence of AD (Ravdin et al., 2003).

Difficulties with praxis usually occur later in the course after memory and language disturbances are evident, although in some cases, apraxia may be a prominent early feature.

Decline in visuospatial skills is a common symptom. Perceptual deficits can be prominent early in the course—even prior to diagnostic ascertainment.

Subtle executive deficits usually are found even in the predementia stages if carefully sought, but more overt frontal lobe deficits such as intrusiveness, difficulty shifting attention or maintaining set, as well as lack of self-care usually occur later. This is in contrast with fvFTD, where these phenomena occur early and often are the presenting features.

As noted earlier, anosognosia, or unawareness of the cognitive deficit, can be an early feature of AD. This denial of any problems may present a difficult management problem and place stress on caregivers.

Atypical Alzheimer Disease Variants

In addition to the classic AD presentation, several relatively rare variants of AD should be recognized.

The frontal variant of AD presents with prominent behavioral and/or personality changes in addition to short-term memory loss. These patients often are impatient, irritable, impulsive, and disinhibited. They show impairments on tests of frontal executive performance such as categorical verbal fluency and Trail-Making A (Chen et al., 1998; Johnson et al., 1999), as well as on response inhibition and set shifting (Chen et al., 1998) and show greater pathological involvement of the frontal neocortex (Johnson et al., 1999).

Posterior cortical atrophy (PCA) is a relatively rare AD variant. PCA patients present with prominent visuospatial dysfunction such as partial or full Balint syndrome (simultanagnosia, ocular apraxia and ocular ataxia), partial or full Gerstmann syndrome (acalculia, agraphia, right/left disorientation, finger agnosia), apperceptive visual agnosia, and environmental disorientation. In addition they frequently show visual-field deficits or constructional, dressing, and ideomotor apraxia. Relatively preserved memory and insight until later in the disease course is the norm (Mendez et al., 2002; Renner et al., 2004; Tang-Wai et al., 2004). The visual processing deficits can be related to both dorsal (“where”) and ventral (“what”) visual-stream impairment but tend to involve the former more prominently, consistent with the topography of the pathology. This form of AD is associated with profound parieto-occipital atrophy and higher AD pathology burden in the primary visual and secondary visual-association cortices (Renner et al., 2004; Tang-Wai et al., 2004). Other pathological conditions with this syndrome have been described, including the Heidenhain variant of Creutzfeldt-Jakob disease (CJD), DLB, corticobasal degeneration, and dementia lacking distinctive histological features (Renner et al., 2004; Tang-Wai et al., 2004).

Finally, the occasional AD patient may also present with early progressive language involvement (Kramer and Miller, 2000) or significant parkinsonian signs and symptoms (Cummings, 2000; Kurlan et al., 2000).

Neuropsychiatric Features

While dementia is classically defined by cognitive impairment, almost all AD patients exhibit a wide range of neuropsychiatric symptoms. Mood state (depressed, euphoric), vegetative status (eating, sleeping), changes in personality (apathetic, disinhibited), and alterations in perception (hallucinations) or thought (delusions) are the major areas to be probed in the course of diagnostic assessment. Although structured interviews for neuropsychiatric assessment in dementia have been developed, their use is restricted primarily to research settings. As noted earlier, a brief questionnaire version of the Neuropsychiatric Inventory (NPI) designed for practice settings—the Neuropsychiatric Inventory Questionnaire (NPIQ)—has been developed to provide a clinical screening examination for common neuropsychiatric manifestations of dementia and their associated impact on caregivers (Kaufer et al., 2000).

Neuropsychiatric assessment plays an important role in differential diagnosis and sometimes reveals the most pressing therapeutic needs. For instance, in the moderate to advanced stages of AD, patients may present with irritability and agitation to the point of violent verbal and physical outbursts, sundowning, hallucinations, and paranoid delusions may create a strain on the spouse, family, and caregivers. When prominent, these symptoms may have to be addressed first, even prior to initiation of anticholinergic medications or memantine. Behavioral symptoms once manifest tend to worsen over the course of the disease; however, for the individual patient, symptoms may fluctuate and may not be present at each clinical evaluation (Cummings, 2000).

Apathy can be found in 42% of those with mild, 80% of those with moderate, and 92% of those with advanced AD (Mega et al., 1996). It presents with loss of interest in previously enjoyed activities (e.g., hobbies, social outings, spending time with beloved relatives), aloofness, diminished spontaneity and emotional behavior, and reduced motivation. It is thought to reflect disruption of the connections within the frontosubcortical–anterior cingulate circuitry and their connections with other cortical regions. Apathy and depression commonly co-occur, but they are not synonymous with each other (Cummings, 2003). Apathy can be distinguished from primary depression on the basis of its neutral affect and the absence of vegetative signs.

Depression is very common in AD, occurring in 10% of mild, 40% to 60% of moderate, and 60% or more of severe AD patients (Mega et al., 1996). The symptoms are rarely severe enough to merit diagnosis of major depressive disorder; more often they represent minor depression/dysphoria. Risk factors for developing depression are familial or personal history of depressive disorder, female gender, and younger age (Lyketsos and Olin, 2002).

Anxiety is another early feature of AD. In the early stages, anxiety may be a manifestation of the patient’s subjective awareness of his/her cognitive decline, his or her increased dependency on others, and fear of the disease and its progression. In the moderate stages, anxiety over abandonment and fear of being left alone are common. Changes in the daily routine and the environment can trigger anxiety in the demented patient and could easily escalate to agitation and aggression.

Agitation and irritability frequently co-occur. Agitation is more common in males, those with later onset of dementia, and those of more advanced age. It encompasses disruptive, aggressive, and/or resistive behaviors and is related to changes in frontal cortex on functional imaging studies and postmortem examination (Cummings, 2003). Common sources of resentment and irritability are the patient’s inability to successfully accomplish tasks that were accomplished with ease in the past, or a feeling of being mistreated or ignored.

AD patients may show a whole host of psychotic features such as hallucinations, delusions, or delusional misidentifications. These typically occur in the moderate to severe stages in AD, whereas in DLB they can occur early on and even be the first manifestation of the disorder. Most hallucinations are in the visual modality. Delusions also tend to favor the later stages of the disorder and occur in 30% to 50% of patients. Most common are delusions of infidelity, theft, and paranoia. Delusions often co-occur with aggression, anxiety, and aberrant motor behavior (Cummings, 2003).

Attention to the neuropsychiatric features of dementia is important because they represent treatable components of excess morbidity and often have a profound impact on caregiver burden and prompt institutionalization (Kaufer et al., 1998).

Laboratory Studies

The evidence-based guidelines of the AAN for diagnostic evaluation of dementia (Knopman et al., 2001) recommend routine screening for vitamin B12 deficiency and hypothyroidism. Other blood tests, such as screening for syphilis, are also justifiable if a clinical suspicion for neurosyphilis is present, either because of high-risk behavior or because of location in an endemic region.

Genetic testing for the ApoE genotype is not recommended on a routine basis. A large multicenter study demonstrated that the presence of the ApoE E4 allele increased the positive predictive value of diagnosing AD by only 4% over diagnoses made on clinical grounds alone (90% versus 94%) (Mayeux et al., 1998).

The AAN guidelines noted that CSF tests for β-amyloid, tau, and neuropil thread protein (AD7C-NTP) gave insufficient data, demonstrating values above and beyond the relatively high sensitivity and specificity of the clinical diagnosis of AD (Knopman et al., 2001). New studies assessing the diagnostic or predictive capabilities of β-amyloid and tau or phosphorylated tau (phosphotau) in the CSF, however, suggest the possibility that they may have a role to play in difficult cases, confirmation of diagnosis, or prediction of development of AD in patients in the predementia stages (Mattsson et al., 2009). Low CSF β-amyloid (Aβ1-42) coupled with elevated tau or phosphotau increases the likelihood that the patient has AD-type pathology (Mattsson et al., 2009; Sunderland et al., 2003). However, it should also be kept in mind that some pathologically confirmed AD patients have shown normal CSF Aβ1-42 and tau levels prior to death (Brunnstrom et al., 2010). In the predementia stages, a CSF pattern suggestive of AD relates to a very high probability of developing AD in the next 5 years (Hansson et al., 2006).

Concentrations of CSF neurotransmitters, neuropeptides, amino acids, and trace elements are of no diagnostic value. Ubiquitin levels in CSF are increased in AD, but the levels are similar to those found in other neurodegenerative disorders. In blood, acute-phase proteins may be elevated but are not helpful in specific diagnosis.

Genetics

A family history of AD is a major risk factor. Familial AD (FAD) has two forms: early-onset autosomal dominant and late-onset familial AD. The latter seems to show a complex polygenic pattern but has increased frequency in families. The former is associated with mutations in one of three genes: the amyloid precursor protein (APP), and the presenilin 1 (PSEN1) and 2 (PSEN2) genes. Early-onset autosomal dominant families in the world have been identified with PS1, accounting for the vast majority of cases. PS1 likely encodes a protein from the γ-secretase complex, an enzyme complex responsible for the cleavage of the APP molecule to β-amyloid. Consideration of genetic testing for mutations requires appropriate pretest advice and counseling, because positive results have deterministic implications for the family. All three genetic mutations—in APP, PSEN1, and PSEN2—increase brain and blood levels of β-amyloid.

Some but not all late-onset FAD pedigrees are associated with inheritance of the E4 allele of ApoE. In addition to the late-onset familial cases, ApoE also contributes to sporadic disease. Increased amyloid load and earlier age at onset are related to ApoE4 gene dosage (Mayeux et al., 1998). The apoE4 genotype is, however, only a risk factor, being neither sufficient nor necessary for disease development, and this is why it is not used routinely in clinical evaluations.

The search in the human genome for other gene variants that may affect risk of AD is ongoing. Genetic variations (polymorphisms) generally have been sought in proteins or lipoproteins related to the pathogenesis of AD (e.g., amyloid metabolism, inflammation, oxidative stress). Numerous genetic risk factors have been proposed, including allelic variants in sortilin-related receptor L (SORL1), clusterin (CLU, also known as ApoJ), phosphatidylinositol binding clathrin assembly protein (PICALM), and many others. To date, only the ApoE4 gene has been consistently confirmed as a “risk gene.” New searches for risk genes in AD use genome scans to look for candidate regions, and loci on several chromosomes are under investigation.

Neuroimaging

The AAN dementia practice parameter guidelines state that at least one unenhanced CT or MRI scan should be performed in patients with cognitive decline to rule out unexpected structural lesions and also to provide information about potential silent vascular injury (Knopman et al., 2001). MRI, with its improved resolution, allows better quantification of cerebral structures and better discrimination of normal from mildly affected patients with AD than is possible with CT. Noninvasive neuroimaging has greatly aided the accurate diagnosis of AD; structural lesions such as tumors, hydrocephalus, subdural hemorrhage, and strokes are identified easily. MRI and to a lesser extent CT aid in identification of vascular lesions that may be primary causes of dementia or contributory to cognitive decline in cases of mixed AD/vascular dementia.

Mesial temporal atrophy including the entorhinal cortex, hippocampus, and amygdala are considered typical for the prodromal AD stages (Apostolova et al., 2006, 2009; Jack et al., 2004) (Fig. 66.1). In the dementia stage, global brain atrophy—more striking in the temporoparietal than in the frontal regions—and ventricular enlargement are also pronounced (Apostolova et al., 2007; Thompson et al., 2003) (Fig. 66.2). A gradient-echo sequence on MRI could reveal cortico-subcortical microhemorrhages suggestive of the presence of vascular amyloidosis.

New and advanced methodologies provide unique opportunities to study the earliest changes in the hippocampal structure. Some studies have documented subtle atrophy present as early as 3 years prior to MCI and 6 years prior to the dementia stages of AD (Apostolova et al., 2010b) (Fig. 66.3). Imaging biomarkers are presently being developed as diagnostic and prognostic biomarkers as well as surrogate biomarkers for clinical trials, with hippocampal atrophy, the most validated structural biomarker, already being accepted as a biomarker criterion for AD presence in the prodromal AD stages (Dubois et al., 2007).

Functional brain imaging with single-photon emission computed tomography (SPECT) and positron emission tomography (PET) can identify disease-specific patterns such as temporoparietal abnormalities in AD, frontal or anterior temporal abnormalities in FTD, and temporo-parieto-occipital abnormalities in DLB (O’Brien, 2007) (Fig. 66.4). SPECT findings of blood-flow abnormalities in a temporoparietal distribution may aid in confirmation of AD, especially in cases without significant atrophy. PET using [18F]fluorodeoxyglucose (FDG), a measure of energy utilization in the brain that predominantly marks synaptic activity, also shows temporoparietal deficits, is more sensitive than SPECT, and can confirm the diagnosis (see Fig. 66.4). Although validation studies of these methods generally have shown high sensitivity, relatively lower specificity poses the risk of false-positive diagnoses. To date, the sensitivity and specificity of PET have not been assessed outside of research clinics (Silverman, 2004; Silverman et al., 2003). It is nevertheless intriguing that similar although less marked deficits in FDG-PET in the cingulate, temporal, and parietal cortices have been demonstrated in presymptomatic persons homozygous for ApoE E4 (Reiman et al., 1996). Further work to more rigorously validate these methods is necessary before they can become more widely applicable (Cummings et al., 2007; Thal et al., 2006).

The recent development of PET ligands for imaging proteins contained in amyloid plaques—Pittsburgh Compound B (PIB) (Klunk et al., 2004; Mathis et al., 2007)—or concomitantly labeling amyloid plaques and neurofibrillary tangles—[18F]-FDDNP (Small et al., 2006)—offers the prospect of a preclinical or early diagnostic biomarker for AD (Mintun et al., 2006). PIB uptake is seen in the same cortical regions that show diminished FDG-PET activity (Fig. 66.5), but PIB imaging offers the advantage of greater spatial resolution and greater effect sizes relative to conventional FDG-PET imaging (Ziolko et al., 2006).

Amyloid PET imaging shows a bimodal uptake distribution in MCI, with some subjects showing AD-like and others normal control (NC)-like retention patterns (Kemppainen et al., 2007; Pike et al., 2007). Longitudinal amyloid imaging studies in the MCI and NC states has revealed that high PIB retention conveys substantially higher risk for progression from MCI to AD (Okello et al., 2009) and for cognitive decline in NC (Morris et al., 2009). PIB retention in amyloid-rich regions has also been documented in postmortem specimens (Ikonomovic et al., 2008; Thompson et al., 2009). Although PIB binding shows the expected correlation with cognitive function cross-sectionally (Jack et al., 2008, 2009; Mormino et al., 2009; Pike et al., 2007; Tolboom et al., 2009), longitudinal PIB studies have surprisingly uncovered that PIB retention levels off in the symptomatic stages, suggesting its greater usefulness as a preclinical rather than an overt clinical biomarker (Engler et al., 2006).

Pathology

Brain atrophy with regional neuronal and synaptic loss and the presence of amyloid and neuritic plaques, and neurofibrillary tangles (NFTs) are the primary pathological features of AD. Deposition of amyloid in the cerebrovascular wall is always present but to a variable degree.

Amyloid protein isolated from plaque cores is predominantly a 40- to 42-amino-acid peptide (Aβ1-42 or Aβ1-40) derived from its larger precursor protein, APP. APP is a transmembrane protein expressed in both neural and non-neural tissue. The APP gene located on the long arm of chromosome 21 has been linked to autosomal dominant early-onset AD. Elevation of Aβ1-42 concentrations with increased synaptic activity suggests it plays a role in synaptic function (Cirrito et al., 2005). A number of metabolic pathways for the processing of APP are recognized, and a shift in metabolism to the β-peptide is thought to be fundamental to amyloid deposition (Selkoe and Schenk, 2003). In the primary metabolic pathway, α-secretase cleaves APP just above the surface of the membrane, resulting in a large-fragment, soluble APP (sAPPα). This cleavage occurs within the β-amyloid domain, precluding Aβ production (Fig. 66.6). The β-amyloid fragment, either Aβ1-42 or Aβ1-40, is cleaved by two different proteases, the β- and γ-secretases at the N- and C-termini, respectively. Monomers aggregate into an oligomeric structure that is neurotoxic. Fibrillization leads to insoluble amyloid that is less neurotoxic. Aβ1-42 has a greater propensity to fibril formation and is the predominant form in early plaques. Vascular amyloid is predominantly Aβ1-40. Posttranslational modification of the fragments may play a role in transition to neuritic plaques. In AD, an apparent partial shift to the amyloidogenic pathway may occur, or there may be impaired clearance, resulting in greater β-amyloid production and subsequent deposition of diffuse in the brain tissue. Basic research studies have shown that the monomeric Aβ1-42 can lower the threshold for neuron death in cell culture and induce oxidative stress. Thus, β- or γ-secretase inhibition is a proposed therapeutic strategy, as are immunotherapies or other strategies to decrease amyloid content in the brain (Fig. 66.7). Markers of inflammation such as α1-antichymotrypsin and other proteases and peptidases are found in neuritic plaques (NP), as is ApoE. ApoE is thought to play a role in amyloid removal. The ApoE E4 allele appears to be less effective in aiding the removal of β-amyloid from the brain, and this may be the reason that patients with AD who are ApoE4 carriers have more amyloid deposition in their brains and earlier onset of disease than non-ApoE E4 carriers.

NP, also called senile plaques, are found predominantly in the cerebral cortex, especially in the association areas and the hippocampus (Fig. 66.8). Amyloid deposition does not follow the topographic sequence for the development of NFTs; it is seen initially in the cortex, then in the hippocampus, and then in other regions (Arnold et al., 1991). NPs range in diameter between 25 and 200 mm and consist of abnormal nerve processes often referred to as dystrophic neuritis, processes of activated microglia and astrocytes, and a central core of β-amyloid (see Fig. 66.8, A). β-Amyloid is a 40- or 42-amino-acid peptide that is cleaved from the large APP molecule. Diffuse or immature plaques are not associated with dystrophic neurites or with fibrillar amyloid; immunostaining reveals the presence of the β-amyloid protein, predominantly Aβ-1-42. It is believed that diffuse plaques precede classical neuritic plaques and that progressive fibril formation by the monomeric Aβ-1-42 into oligomers leads to initiation of an inflammatory process that results in protease and peptidase deposition and neuropil destruction. Some authors refer to “burnt-out plaques” in which a prominent amyloid core is surrounded by a very thin rim of dystrophic neurites.

The primary component of NFTs is the microtubule-associated protein, tau. Tau protein is a cytoskeletal protein found predominantly in axons. In AD, tau is abnormally hyperphosphorylated and extensively cross-linked, forming an insoluble intracellular deposit (see Fig. 66.8, B). This altered form has reduced binding to microtubules, thereby disrupting the cytoskeleton and leading to interference with neuronal metabolism and subsequently to neuron death. Tau has six isoforms. These isofoms result from alternative splicing of the gene; three of the isoforms have three microtubule binding domain repeats (3R tau), andthree of the isoforms have four repeated sequences (4R). Variations seen in Western blots of tau isolated from brain in AD and Pick disease, corticobasal degeneration, and hereditary tauopathy indicate differential deposition of isoforms (Wszolek et al., 2006). Ubiquitin is frequently found in association with tangles, probably representing an attempt by the neuron to degrade the abnormal protein. Deposition of NFT appears to begin in the transentorhinal and entorhinal cortex, spreading from there to the hippocampus followed by the temporal neocortex and beyond (Delacourte et al., 1999). There is early laminar predilection is initially for layer 2 and then layer 5 of the entorhinal cortex followed by the CA1 and subicular region of the hippocampus, and the pyramidal cells of neocortical layers 3 and 5. Ultrastructurally, NFTs consist of paired helical filaments with an individual filament diameter of 10 nm wound in a double helix, with a total diameter of 200 nm and a periodicity of 160 nm. NFTs are not exclusive to AD and can be found in other conditions such as dementia pugilistica, prion disease, and Kufs disease. Neuropil threads (curly fibers) are also found and represent paired helical filament–containing neurites. Their number usually parallels the severity of neurofibrillary tangle formation.

NP and NFT burden correlate with disease severity, with the correlation being stronger with NFTs. Still, cases with high neuropathological burden can be seen in the absence of dementia. Synaptic and neuronal loss shows the highest correlations with global cognitive impairment. However, to date, neuropathological criteria for AD continue to be based on plaque and tangle numbers.

Other histological AD features include granulovacuolar degeneration (see Fig. 66.8, C) and Hirano bodies (see Fig. 66.8, D). Granulovacuolar degeneration is typically seen in hippocampal and consists of clear, round, intracytoplasmic basophilic vesicles 4 to 5 mm in diameter that immunolabel for tubulin, ubiquitin, and neurofilament. Hirano bodies are eosinophilic rodlike structures located almost exclusively in the Sommer sector of the hippocampal pyramidal layer, adjacent to and occasionally in neuronal bodies. Hirano bodies contain actin and actin-associated proteins, tau, and a C-terminal fragment of APP. Mild cortical spongiosis that is distinct from the spongiform changes of CJD can be found in the association cortices, sparing the primary sensory and motor cortex in some cases.

Cell loss preferentially involves the large neurons of the deeper layers of the cortex. Dysfunction of the cholinergic system in early stages (before loss of the synthetic enzyme ChAT) is suggested by the loss of basal forebrain neurons and receptors for the cholinergic neurotrophin nerve growth factor, as well as by the sensitivity of patients with AD to anticholinergics. Studies of patients with end-stage AD in the 1970s and 1980s uncovered extensive neuron loss in the nucleus basalis of Meynert and other cholinergic basal forebrain nuclei. These structures are the origin of the cholinergic projection to cerebral cortex and other forebrain regions. This loss was found to associate with loss of choline acetyltransferase (ChAT) in the projection fields of these nuclei. More recent work, however, revealed that (1) the loss of ChAT does not occur until much later in AD’s course (DeKosky et al., 2002), and (2) that cholinergic cells in the cholinergic basal forebrain neurons are preserved until very late in the course (Mufson et al., 2002). Neuron loss occurs to a lesser extent within the locus ceruleus and the nucleus raphe. As these studies were performed in late-stage cases, it is possible that these neurons, too, are preserved longer than was earlier thought.

The reason for the selective damage to the cholinergic system, with loss of ChAT and acetylcholinesterase, is not known. A number of other subcortical ascending neurotransmitter projections are involved, in particular the noradrenergic and serotonergic systems. Changes in the glutamatergic system also occur. Glutamatergic receptors decline is especially prominent in the pyramidal neurons (in cortex and in entorhinal cortex), which bear the brunt of cell loss and NFT deposition. The discovery of the cholinergic deficit has led to the first rational treatment for AD. The report of glutamatergic dysfunction led to the other currently approved therapeutic strategy.

Treatment

Acetylcholinesterase Inhibitors

Acetylcholinesterase inhibitors (AChEIs) are the first line of pharmacological treatment in AD. Treatment of AD with AChEIs was judged to be an accepted standard of care in the AAN practice guidelines for dementia (Doody et al., 2001). The following three agents are now available in the United States, Europe, and many other parts of the world: donepezil (Aricept), rivastigmine (Exelon), and galantamine (Reminyl). All three medications are effective and as shown in double-blind placebo-controlled trials can potentially stimulate or stabilize performance. Initial studies were 3 to 6 months in duration; double-blind placebo-controlled trials of up to 1 year (donepezil) show maintenance of medication effects, and open-label extension studies suggest continued efficacy for several years. Sudden improvement in cognitive function occurs in a small percentage of subjects. Slowing of decline or stabilization over time, difficult to determine in a brief exposure to the drug, also occurs with these medications. Gastrointestinal cholinergic-mediated side effects are commonly seen with AChEIs. Side effects are more common during the dose escalation phase of treatment; emphasizing this to patients and caregivers will enhance compliance. Gastrointestinal effects can be reduced by giving these drugs after meals or slowing the dose escalation rate. Beneficial effects of these agents also have been shown on neurobehavioral symptoms, notably apathy and visual hallucinations, and on stabilizing activities of daily living functions or slowing their decline. If therapy with one agent appears ineffective or causes intolerable side effects, another AChEI should be tried. Because of the nature of the neurochemical action of these medications, withdrawal effects are possible on switching drugs, so prolonged washout periods should be avoided.

Glutamate Receptor Modulators

The abnormalities in glutamate metabolism and receptors noted postmortem in brains of patients who had AD led to hypotheses that glutamate metabolic dysregulation leads to excitotoxicity and neuronal death in AD (Rogawski and Wenk, 2003). Memantine, an N-methyl-d-aspartate (NMDA) antagonist that had been available in Germany since the 1980s, was assessed as solitary therapy in two pivotal trials in moderate to severe AD (MMSE score of 5 to 15) in the United States (Reisberg et al., 2003) or as an add-on to ongoing AChEI therapy (Tariot et al., 2004). Both studies showed a significant benefit at the end of the trial in cognition and function, and memantine was approved for use in moderate to severe AD. No specific side effects were identified in the pivotal trials; side effects were significantly more frequent than in the placebo groups.

Other Cognitive-Enhancing or Disease-Modifying Drugs

With the vast increase in knowledge of the disease mechanisms operative in AD, many medications aimed at slowing the disease process and ameliorating cognitive decline are under development or evaluation. These include drugs currently approved for other indications, new medications, and complementary or alternative medications. Most proposed or unregulated preparations have been suggested by virtue of their potential ability to interfere with the known pathobiological cascades in AD, notably oxidative stress and inflammation. Vitamin E, an antioxidant, in a dose of 1000 IU twice daily had some effect on delaying time to nursing home placement, decline from moderate to severe AD, or death (Sano et al., 1997). However, recent data suggesting that high doses of vitamin E may cause cardiac toxicity, coupled with the failure of high-dose vitamin E to slow conversion of MCI to AD (Petersen et al., 2005) have resulted in abandonment of vitamin E as a treatment. Other free radical inhibitors and antioxidants are used, but without support from prospective data.

Estrogen or estrogen-progesterone replacement therapy was shown in several retrospective studies to delay or prevent emergence of AD, but two studies, one large and one small, showed that they have no effect on disease progression (Henderson et al., 2000; Mulnard et al., 2000). The Women’s Health Initiative Memory Study (WHIMS) showed no effect of these agents on incidence of either MCI or AD; in fact, a slight increase in dementia incidence was noted (Shumaker et al., 2003).

Anti-inflammatory agents also have been tried in symptomatic AD; neither low-dose prednisone nor nonsteroidal antiinflammatory drugs (NSAIDs) have been shown to slow progression of symptoms.

The use of Ginkgo biloba as a treatment for AD has now been credibly disputed by two separate studies. A carefully performed double-blind placebo-controlled 6-month-long study of ginkgo showed no effect at 6 months (Schneider et al., 2000). Another randomized double-blind placebo-controlled 7-year-long clinical trial with 3069 community-dwelling participants aged 72 to 96 demonstrated no benefit of gingko on cognitive performance in normal cognition or MCI (Snitz et al., 2009).

Latrepirdine (Dimebon) attracted interest after showing potential beneficial effects in AD animal models, a phase II human trial, and a phase III double-blind trial with mild- to moderate-stage patients (Doody et al., 2008). A second phase III monotherapy trial showed no benefit. An ongoing study is assessing the utility of latrepirdine in patients receiving donepezil.

Medications for Behavioral and Neuropsychiatric Symptoms

The behavioral and neuropsychiatric symptoms of AD are treated in several ways. First, if possible, are nonpharmacological techniques. Disorientation may be reduced by providing a quiet, familiar environment with clear labels on doors and other objects throughout the house. Efficient lighting to the bathroom or other amenities is important to reduce confusion at night.

Acute confusional states also may be produced by intercurrent medical problems. Even minor urinary tract or other infections may lead to marked behavioral disruption. Any patient with AD presenting with sudden deterioration of cognition should undergo careful clinical examination and investigation for signs of medical or surgical disease; depending on dementia severity, the patient may not be able to articulate the problem.

Aggressive behavior may pose considerable management problems and should always invoke thorough clinical assessment to determine the etiology of the symptoms. Psychological intervention includes specific training for professional and family caregivers in dealing with aggression, the need for alertness to the signs of an impending crisis, and the use of positive and clear language to reassure and distract the patient.

Depressive symptoms should be treated with selective serotonin uptake inhibitors (SSRIs) because they have the least anticholinergic effects. They also may ease anxiety, irritability, or other nonspecific symptoms that may accompany depression. The SSRI, citalopram, may be useful for agitation.

Agitation or disruptive behavior may require a neuroleptic. The newer atypical antipsychotic medications (quetiapine, risperidone, olanzapine) have been recommended in low doses with careful titration, allowing time for the medications to exert their effects. The Clinical Antipsychotic Trials of Intervention Effectiveness (Schneider et al., 2006), however, indicated that within the limitations inherent in attempts to do a carefully controlled outpatient trial on agitation, side effects, and limited efficacy, the atypical antipsychotics did not show clear advantages over the older antipsychotic in the study. Traditional neuroleptics are more likely to produce extrapyramidal symptoms, which may worsen cognitive function. The newer antipsychotic agents are associated with increased mortality in the elderly and have had a “black box” warning label added by the U.S. Food and Drug Administration (FDA). The older antipsychotics, however, also carry an increased risk of death (Schneeweiss et al., 2007). Thus, judicious use of all of these medications, with frequent reassessment of the need for such a drug (at least every month or two), is appropriate.

Short-acting benzodiazepines frequently are used for sleep but should not be used indefinitely; trazodone, an antidepressant with sedative properties, frequently is used to promote sleep. The newer sleep medications have not been assessed in controlled trials for their effects and possible adverse effects in AD, so no recommendation for or against their use can be made at this time.

Future Therapies

Although efforts will continue to develop better cognitive enhancers and effective neurobehavioral medications with favorable side-effect profiles, significant attention now is directed at therapies that would directly interdict the pathological cascades in AD. Much discussion centers on whether NFTs or amyloid may be “primary” in disease causality. While both pathological cascades are the target of intense research interest and study, and it must be noted that the relationship between NPs and NFTs is unknown. Studies to disrupt NFT deposition center on preventing them, because the extensive cross-linking and hyperphosphorylation make removing them a difficult if not impossible task. Glycogen synthase kinase 3 (GSK3) is thought to be involved in the phosphorylation of tau, and targeting GSK3 or finding other ways to interfere with pathological tau phosphorylation is under study.

Interruption of the amyloid cascade also has a number of strategies, including β-secretase inhibitors, γ-secretase inhibitors or modifiers, or α-secretase enhancers (see Fig. 66.7). Compounds that would insert themselves on the polymerizing β-amyloid, the so-called plaque busters, conceivably could aid in slowing β-amyloid accumulation. Accelerated removal of β-amyloid once it is formed, either by antibody infusion (passive immunization) or active immunization to produce endogenous antibodies, is effective in transgenic mouse models. Initial active immunization trials in humans were interrupted by neuroinflammation and encephalitis in a subset of the subjects, probably due to T-cell sensitization. Autopsy studies in several immunized individuals from that study showed the expected neuroinflammatory changes (Orgogozo et al., 2003), as well as a remarkable amyloid clearance in several areas of the cortex, suggesting that—as in mice—the amyloid in humans can be removed (Masliah et al., 2005; Nicoll et al., 2003). Future studies will determine the feasibility of removing amyloid safely, whether it results in slowing of cognitive decline, and whether it has any effects on NFT formation.

Cerebral Amyloid Angiopathy

β-Amyloid deposits can be seen in the cerebral blood vessels of all patients with AD (see Fig. 66.8, E). Severity of involvement varies, ranging from the presence of small amounts of amyloid to major deposits that distort artery architecture, causing cortical microinfarcts and microaneurysm with or without cerebral hemorrhages. Familial cases of AD and cerebral amyloid angiopathy (CAA) are associated with multiple hemorrhages in the brain, spinal cord, and leptomeninges. Although these hemorrhages could be clinically silent, patients may present with progressive dementia and spastic paraparesis. MRI using a gradient-echo sequence can show multiple small hemorrhages in the brain and spinal cord. The most common forms of familial CAA without AD manifest with spontaneous lobar hemorrhages rather than dementia (i.e., Icelandic and Dutch cerebral amyloid angiopathy). Lobar hemorrhage in late life in nonfamilial cases also is suggestive of CAA (Table 66.2). This diagnosis is suspect when the hemorrhage is “lobar,” located at the cortical gray/white-matter junction, and not in the typical distribution of hypertensive hemorrhages (i.e., basal ganglia, brainstem, or cerebellum). AD can be accompanied by severe amyloid angiopathy.

Table 66.2 Boston Criteria for Diagnosis of Cerebral Amyloid Angiopathy–Related Hemorrhage

Definite CAA Full postmortem examination demonstrating CAA
  Absence of other diagnostic lesions
Probable CAA with supporting pathology in specimen Clinical data and histopathological specimen (evacuated hematoma or biopsy) demonstrating some degree of CAA
  Absence of other diagnostic lesions
Probable CAA Clinical data and MRI or CT demonstrating multiple hemorrhages restricted to lobar, cortical, or corticosubcortical regions (cerebellar hemorrhage allowed) in individuals age ≥55 years
Possible CAA Clinical data and MRI or CT demonstrating single lobar, cortical, or corticosubcortical region hemorrhage in individuals age ≥55 years

CAA, Cerebral amyloid angiopathy; CT, Computed tomography; MRI, magnetic resonance imaging.

Adapted from Knudsen, K.A., Rosand, J., Karluk, D., et al., 2001. Clinical diagnosis of cerebral amyloid angiopathy: validation of the Boston Criteria. Neurology 56, 537-539.

More recently, an inflammatory presentation of amyloid angiopathy has been described. This syndrome is associated with subacute cognitive decline, seizures, and extensive white-matter abnormalities and microhemorrhages on imaging studies. Histologically, this presentation was found to be associated with giant-cell inflammatory response. The CAA-related inflammation appears to be relatively responsive to corticosteroids and cyclophosphamide (Eng et al., 2004).

Frontotemporal Dementia Spectrum

The frontotemporal dementias (FTDs) are a group of neurodegenerative dementias of varied etiology in which the frontal or temporal lobes, or both, are affected out of proportion to the rest of the brain, with variable degrees of subcortical pathology and degeneration. The first pathologically described case of FTD consistent with Pick disease was published in 1892 by the German neurologist, Arnold Pick. The clinical syndrome of frontal lobe degeneration of non-Alzheimer type with onset typically between the ages of 50 and 60 years and featuring insidious personality change, disinhibition, and subsequent gradual loss of speech output was described by Gustafson and Brun in the early 1990s (Gustafson et al., 1990). The FTD spectrum is a clinically and pathologically inhomogeneous group. Several distinct clinical phenotypes have been described: one behavioral variant, frontal-variant FTD (fvFTD); two language variants, primary progressive aphasia (PPA) and semantic dementia (SD); and one variant with associated motor neuron disease (MND), FTD-MND. Pathologically, FTDs have been attributed to a variety of molecular defects with new ones still being described. Population estimates of the incidence of FTD are not available, but most series estimate that it makes up 10% to 15% of the neurodegenerative dementias in clinical autopsy series.

Diagnostic Criteria

The most recently proposed criteria (McKhann et al., 2001) developed for the FTD spectrum in general are presented in Box 66.4. These are relatively well suited for clinical practice. In the research environment, however, the most widely accepted criteria for the FTD spectrum disorders are the Neary criteria (Neary et al., 1998). These are presented in Boxes 66.5, 66.6, and 66.7 (also see Clinical Presentation section).

Clinical Presentation

Consideration of a diagnosis of FTD follows elimination of possible systemic causes, other CNS cognitive or behavioral degenerative disorders (e.g., Huntington disease, AD) and substance abuse, which because of the lack of social concern in the patient, frequently is entertained as a cause by family, friends, or physicians (see Box 66.4). Absence of widespread knowledge of features and symptoms of FTD probably keeps it from being diagnosed more frequently.

The classification of FTDs is exceedingly controversial owing to large gaps in our understanding of the cellular and molecular pathophysiology and their associations with distinct clinical phenotypes (Geser et al., 2010). In this chapter, we will first describe the clinical phenotypes, and later on (see Genetics and Pathology sections) we will talk about the distinct molecular and neuropathological subtypes.

In general, two patterns of presentation and progression of FTD are seen: progressive behavioral disturbance and progressive language disturbance. In the first, personality changes are prominent, executive function is impaired, and reasoning is altered. The progressive aphasias are a group of neurodegenerative syndromes with a hallmark of language dissolution as the primary presenting symptom. They generally are grouped with the FTDs on the basis of their histopathological features.

Frontal-Variant Frontotemporal Dementia

Frontal-variant FTD affects males and females equally. The mean duration of illness is approximately 8 years, although the range is from 2 to 15 years. Family history of a similar disorder in a first-degree relative is present in approximately half of cases, and some families demonstrate an autosomal dominant mode of inheritance. The presence or absence of a family history does not determine the behavioral pattern; familial cases occur in the disinhibited, inert, and stereotypical forms of the disorder. No known geographical influences on prevalence exist.

The classic clinical variant of FTD is fvFTD, also known as behavioral-variant FTD, primary progressive behavioral disorder, and behavioral disorder and dysexecutive syndrome. Behavioral characteristics vary along a spectrum depending on the part of the frontal lobes affected. Some patients are disinhibited, overactive, and restless and show prominent orbitofrontal involvement. Others with more widespread frontal involvement are apathetic and lack motivation and initiative. In general, fvFTD is associated with insidious early personal conduct and personality changes. Patients are frequently socially inappropriate, tactless, and disinhibited or aloof and socially isolated. FvFTD patients show impaired judgment and readily disobey socially accepted norms and boundaries. These are frequently described as exceedingly self-centered, shallow, lacking empathy and insight, emotionally labile or withdrawn, or impulsive. They pay less attention to their work or home responsibilities. Their behavior can be purposeless and lacking in goals. Obsessions and compulsions are common and may take the form of hoarding, unusual collecting, consumption of the same food item (especially sweets), and even consumption of inedible items. Patients experience difficulty in organizing or sequencing, without evidence of agnosia or ideomotor dyspraxia. Detailed neuropsychological evaluation reveals impairment of frontal executive abilities such as abstract thinking, decision making, planning, sequencing and set shifting, and diminished free verbal recall, with characteristic sparing of visual memory (Hodges and Miller, 2001). Box 66.5 lists the Neary criteria for fvFTD.

Primary Progressive Aphasia

Nonfluent PPA (also known as progressive nonfluent aphasia or PNFA) is characterized by early prominent language impairment with dysfluent, effortful, and agrammatical language output in the context of preserved language comprehension until late in the disease course (see Box 66.6). Anomia and phonemic paraphasic errors are pronounced early on. Speech output requires manifest effort and is nonfluent and hesitant with phonemic and semantic errors. Writing often is similarly affected, although dissociations in the degree of speech versus written impairment occasionally may be noted. Word finding and repetition are impaired, and spelling is poor. Early on, comprehension is preserved, as is insight into the disorder and nonlanguage cognitive skills including memory, allowing some patients to maintain employment and productivity for years after symptom onset. Speech output becomes progressively constrained, and dyspraxia ensues, impairing communication. As the disease advances, comprehension eventually deteriorates, but patients may retain some understanding even without the ability to speak or communicate with gestures. Social skills are preserved early in the course, but frontal symptoms may occur later. Behavioral disturbances are mild, and PPA patients are typically socially competent until late in the disease.

Some investigators now also identify the fluent form of PPA with greater early impairment in comprehension and preservation of articulation and words per utterance (Mesulam, 2003). Other investigators consider any fluent form of progressive aphasia to be SD, regardless of the presence of agnosia. It is unclear whether such subdivisions have meaning with regard to etiology and histopathology. To some extent, these forms differ by the prominence of pathology along the perisylvian fissure, with changes in the nonfluent form located more anteriorly along the axis. In almost all cases, anomia is a prominent initial symptom.

Semantic Dementia

SD (also known as semantic aphasia and temporal-variant FTD) presents with characteristic language impairment resulting from semantic knowledge loss (i.e., loss of conceptual knowledge and word meaning) (see Box 66.7). As a result, the language output in SD, while being fluent, well articulated, and grammatically correct, shows impoverished content and semantic supraordinate paraphasias (e.g., naming a lion an “animal” or a flower a “plant”). Further semantic loss leads to frequent substitutions of many nouns with “it” or “thing” to the point that the message the individual is trying to convey is completely lost. Impaired comprehension for both spoken and written language is also characteristic. Fluency belies profound anomia and word comprehension deficits, most evident on neuropsychological testing, which usually is necessary to diagnose and evaluate the patient over time. SD is characterized by a unique type of dyslexia called surface dyslexia, where irregular words such as “colonel,” “pint” or “yacht” are read and written with spelling-to-sound correspondence. At the same time, ability to repeat is preserved, and reading and writing are fluent.

The loss of general knowledge, by definition, extends beyond language. Accordingly, prosopagnosia and object recognition difficulties occur despite preserved primary perception. At the same time, performance on copying tasks and perceptual matching can be normal. Usually, widespread impairments of semantic knowledge across all sensory modalities emerge with disease progression.

Unlike individuals with PPA, SD patients frequently show frontally mediated behavioral abnormalities similar to those seen in FTD (Bozeat et al., 2000; Hodges and Miller, 2001). Although the dramatic disinhibition of FTD typically is absent, other frontal behaviors such as obsessions, compulsions, preoccupations, and behavioral stereotypy may appear. Ultimately, both PPA and SD patients progress to mutism or use of only a few words.

Frontotemporal Dementia with Motor Neuron Disease

FTD-MND patients may have behavioral or aphasic FTD presentation with additional amyotrophic lateral sclerosis (ALS)-like motor neuron symptoms such as fasciculations, muscle weakness, hyperreflexia, spasticity, and upgoing plantar reflexes. Additionally, they may have parkinsonian signs. Swallowing difficulties and choking may emerge, necessitating alternative methods of feeding and airway protection. On the other hand, frontal lobe deficits frequently can be found with detailed testing in ALS. It is estimated that FTD develops in 10% of patients with ALS, suggesting that these two disorders may be along the clinicopathological spectrum of a similar underlying neurodegenerative phenomenon (Chen-Plotkin et al., 2010).

Differentiating Frontotemporal Dementia from Alzheimer Disease

Although FTD patients sometimes complain of “diminished memory function” this usually is due to poor attention or a dysexecutive syndrome. Careful testing usually reveals relatively intact memory function. Obviously, memory testing is limited in patients with significant language or semantic impairment, but notably, recognition memory is spared, in contrast with AD (Graham et al., 1997). Neuropsychological testing, especially of recent memory function, may give normal results early in the course. Executive dysfunction can manifest as problems in set shifting, sequencing, abstract thinking, fluency, or confrontation naming. In contradistinction to those with AD, patients with FTD often show greater difficulty in naming words starting with a particular letter (e.g., words beginning with f) than in identifying members of a category (e.g., animals).

Early in the course of FTD, behavioral inappropriateness becomes prominent, contrasting with AD, in which social graces are maintained until later in the course. Frontal-variant AD with early prominent frontal lobe symptoms can potentially present a diagnostic dilemma. Assessment of memory function and consideration of a functional imaging study (i.e., FDG-PET) usually will lead to the correct diagnosis. Motor abnormalities such as ptosis, difficulty swallowing, or peripheral weakness occur occasionally in FTD but are not seen in AD. If an autosomal dominant family history is present, genetic studies of the tau gene, looking for specific mutations, should be considered after careful discussions with the family members about the implications of knowing the genetic information. Occasionally, extrapyramidal symptoms will develop late in the course of FTD in patients without a family history, presumably as a result of disruption of corticobasal ganglia pathways.

Genetics

The genetic underpinnings of FTD are exceedingly complex and evolving concepts (Fig. 66.9). The discovery of genetic mutations in the tau (MAPT) gene on chromosome 17 led to increased focus on tau mutations or other metabolic abnormalities in the tau gene in FTD. A consensus conference on FTD with parkinsonism reviewed data for 13 families with linkage of their clinical symptoms to chromosomal locus 17q21-22 (Foster et al., 1996). A host of tau abnormalities have been described since the initial FTDP17 mutation was described. It has been determined that MAPT mutations cause between 5% and 15% of FTD cases, with age of onset ranging between 25 and 65 years of age. Some cases may also present as progressive supranuclear palsy and corticobasal degeneration (van Swieten and Heutink, 2008).

Tau protein, a microtubule binding protein found in neurons and glia, is needed for stabilization and polymerization of microtubules. The region of the tau molecule that binds to microtubules has repeat sequences of 31 or 32 amino acids. Six isoforms of tau in the adult human brain result from alternative splicing of the gene; three of the isoforms have three microtubule binding domain repeats (3R tau), and three of the isoforms have four repeated sequences (4R). Mutations in the MAPT gene associated with FTDP17 may occur in either exons or introns and may alter the production of either 3R or 4R tau, or alter the ability of tau to bind to microtubules. Most genotypic alterations do not correlate well with clinical phenotypes. However, overproduction of the 4R isoforms (associated with mutations that affect the splicing of exon 10) appears to associate with parkinsonian symptoms, while mutations that do not affect exon 10 splicing of exon 10 are associated more frequently with dementia-prominent forms. Ghetti and colleagues (2003) have summarized the current knowledge of the relationship of the mutations and their associated clinical syndromes in a recent review paper.

In 2006, some cases previously attributed to FTDP17, with conspicuous absence of tau pathology, were shown to be actually due to mutations in the progranulin gene (GRN) located on chromosome 17 (Chen-Plotkin et al., 2010; van Swieten and Heutink, 2008). The age at onset in these cases can be highly variable, ranging from 39 to 89 years (van Swieten and Heutink, 2008). 70% to 90% of patients with GRN mutations have a positive family history of dementia with parkinsonism. With more than 50 GRN gene mutations described to date, there is considerable phenotypical heterogeneity with GRN mutations. While described in FTD, GRN mutations have been reported to also cause AD, and corticobasal degeneration. Plasma and CSF levels of progranulin have been found to be reduced nearly fourfold in both affected and unaffected subjects with PGRN mutations Low (75% reduction) plasma progranulin levels may be used as a screening tool for PGRN mutations (Finch et al., 2009).

FTD with TAR-binding DNA protein (TDP-43) inclusions is the most common pathological variant in the FTD spectrum. TARDBP mutations have been strongly associated with familial forms of ALS (Geser et al., 2010). Yet familial FTD-MND cases without mutations in the TARDBP gene on chromosome 1 that encodes TDP-43 have been described suggesting that other genetic causes of FTD-MND exist. (Gijselinck et al., 2009). Recently one group reported a single base substitution in the TARDBP gene in an elderly woman diagnosed with the apathetic subtype of fvFTD without MND (Borroni et al., 2009). Cases with GRN mutations characteristically present with ubiquitin-positive TDP-43 positive inclusions (Chen-Plotkin et al., 2010).

Recent studies also have identified mutations in the valosin gene on chromosome 9 and the CHMP2B gene on chromosome 3 associated with FTD. The FTD cases related to valosin mutations are very rare and may include Paget disease and inclusion body myositis as part of the clinical manifestations. FTD cases with CHMP2B mutations are similarly rare and can be associated with MND (van Swieten and Heutink, 2008).

Neuroimaging

MRI (or CT) scans in fvFTD frequently show frontal and temporal cortical atrophy (Fig. 66.10), which are often times asymmetrical. Nonfluent PPA usually presents with left perisylvian atrophy, particularly in the inferior frontal cortex, while fluent PPA tends to be associated with more inferior, middle, and polar temporal lobe involvement (Mesulam, 2003). SD presents with bilateral anterior temporal lobe involvement. Greater right temporal lobe disease is associated with prosopagnosia, while progressive limb apraxia is associated with frontal premotor and parietal pathology.

Functional imaging studies with either SPECT or FDG-PET show frontotemporal patterns of hypoperfusion/hypometabolism. Occasionally when no cortical atrophy is evident on structural imaging, a functional imaging study with SPECT or PET can aid the diagnosis by revealing a frontal functional deficit. In PPA, SD, and progressive apraxia, PET or SPECT imaging reveals profound hypoperfusion/hypometabolism in the affected areas that also show atrophy on MRI.

Pathology

Histopathology

There is a growing trend to classify the FTD spectrum disorders based on their histopathological appearance. While it is true the underlying histology does not appear to relate closely to either the clinical syndrome (fvFTD versus PPA, for instance) or the presence of a positive family history, it is the histopathological defect that is most closely related to the underlying pathophysiology. Recognizing the great need for systematic nomenclature for pathological classification, an expert panel recently suggested a protein-based nomenclature as the most simple, consistent, transparent, and capable of accommodating future discoveries (Mackenzie et al., 2009) (Table 66.3).

The first histopathological class, FTLD-tau (frontotemporal lobar degeneration secondary to tauopathy), contains classic Pick disease, corticobasal degeneration, progressive supranuclear palsy, argyrophilic grain disease, multiple system atrophy with dementia, and unclassifiable forms. As in AD, detection of tau pathology in FTD requires application of silver stains or immunostaining with tau antibodies, or both.

The tau-negative FTLD spectrum consists of several histopathological subtypes. FTLD-TDP is the subtype characterized by inclusions staining for TDP-43 (Fig. 66.11, A-B). The majority of tau-negative ubiquitin-positive cases appear to be associated with TDP-43 aggregation (Chen-Plotkin et al., 2010; Geser et al., 2010). TDP-43 is a nuclear protein of uncertain function. In FTLD cases, TDP-43 is hyperphosphorylated, undergoes ubiquitination, and is cleaved to generate C-terminal fragments. TDP-43 aggregates in the cytoplasm and disappears from the nucleus. Of importance, the inclusions occur in the neurons of the affected regions that correspond well with the clinical symptoms. FTLD-TDP commonly presents as FTD-MND. Similar inclusions are also found in lower motor neurons in patients with ALS. Patients found to have TDP-43-positive inclusions can present anywhere along a clinical spectrum from pure ALS to pure FTD. The fact that ALS and FTD-MND feature TDP-43 inclusions as a primary pathological finding suggests that these two disorders share a similar neurodegenerative pathobiology and belong to the spectrum of TDP-43 proteinopathies (Chen-Plotkin et al., 2010; Geser et al., 2010). Three subtypes with distinct clinicopathological correspondence have been described: (1) cases with neuronal TDP-43 inclusions and glial inclusions but few or absent neurites and predilection for frontal cortex, usually present with FTD-MND; (2) cases with thick dystrophic neurites without a discrete cortical laminar predilection that tend to present with striking temporal atrophy and SD features; and (3) cases with superficial cortical spongiosis and predominantly neuronal pleomorphic intracytoplasmic TDP-43 and thin immunoreactive neurites in the pyramidal layer of the hippocampus inclusions, usually present with the fvFTD subtype but can also manifest as nonfluent PPA with or without MND (Dickson, 2009). Presence of neuronal intracytoplasmic inclusions correlates with short survival (Geser et al., 2010).

Cases with inclusions staining for ubiquitin but not TDP-43 are designated FTLD-UPS and typically belong to one of two types: the atypical FTLD with ubiquitinated inclusions (aFTLD-U) or FTLD linked to chromosome 3 (FTD-3).

A few of the FTDs with ubiquitin-positive inclusions have been identified as neuronal intermediate filament inclusion disease (Cairns et al., 2004). In these cases, the ubiquitin-positive inclusions immunostain for neurofilament and α-internexin. Such cases are now proposed to be classified as FTLD-intermediate filament (FTLD-IF), whereas the formerly known dementia lacking distinctive neuropathology (DLDH) cases are now classified as FTLD-no inclusions (FTLD-ni). Cases with basophilic inclusions of unidentified biochemistry are now referred to as basophilic inclusion body disease (BIBD) (Mackenzie et al., 2009).

In addition to characteristic inclusions, FTLD can also show microvacuolar or spongiform changes and gliosis with or without swollen neurons. Classic Pick disease, which was the first FTLD disorder described by Arnold Pick in 1892, is characterized histologically by severe astrocytic gliosis, ballooned cells, and intraneuronal inclusions (see Fig. 66.11, C-F). In other cases the large cortical neurons, primarily in laminae III and V, are lost, and spongiform change and microvacuolation are seen in lamina II. Occasionally the neuronal loss is accompanied by diffuse gliosis but with little or no spongiform change or microvacuolation. Swollen neurons or inclusions that are both tau- and ubiquitin-positive are present in some cases. The limbic system and striatum show more severe damage.

In contrast to AD, cholinergic deficits have not been reported in FTD. Serotonin and serotonin receptors, somatostatin, and corticotropin-releasing factor (CRF) levels are reported to be decreased. CSF norepinephrine, homovanillic acid, and 5-hydroxyindoleacetic acid are decreased. The variable severity of involvement and distribution of the neurochemical changes seen in autopsy cases has precluded full characterization of the affected systems.

Parkinsonian Dementias

Parkinsonian dementia syndromes encompass a broad range of etiological disorders that share a heterogeneous combination of cognitive and motor features (Table 66.4). In general, the disorders in this group have an insidious onset and gradual progression. A positive family history of a similar clinical syndrome implicates an autosomal dominant or familial variant. The type, timing, and topography of parkinsonian motor signs are important for differential diagnosis, as are associated cognitive, neuropsychiatric, or other specific clinical manifestations. Although most degenerative disorders in this category are more prevalent with advancing age, others, particularly the metabolic disorders, tend to occur earlier in life. The clinical and anatomical features of selected degenerative parkinsonian dementias are illustrated in Table 66.5.

Table 66.4 Classification and Salient Features of Parkinsonian Dementia Syndromes

Etiology/Syndrome Distinguishing Features
DEGENERATIVE (SPORADIC)
Parkinson disease with dementia (PDD) Parkinsonism initially, later onset of dementia
Dementia with Lewy bodies (DLB) Recurrent visual hallucinations, fluctuating cognition, variable parkinsonian signs
Progressive supranuclear palsy (PSP) Balance and bulbar dysfunction, vertical gaze palsy
Corticobasal degeneration (CBD) Asymmetric limb signs (apraxia, myoclonus)
Multisystem atrophy (MSA) “Parkinson-plus” syndromes
Cerebellar type Brainstem/cerebellar atrophy, ocular dysmotility
Parkinsonian type Motor parkinsonism, dysautonomia
DEGENERATIVE (FAMILIAL)
Huntington disease (HD) Autosomal dominant; chorea, athetosis, personality changes
Neuroacanthocytosis Autosomal dominant; HD mimic, acanthocytic red blood cells
Machado-Joseph disease Autosomal dominant; ataxia and dysarthria, cerebellar atrophy
Progressive subcortical gliosis Autosomal dominant; white matter gliosis, frontal atrophy
Familial frontotemporal dementia (FTDP-17) Autosomal dominant; chromosome 17–linked, frontal atrophy
SECONDARY PARKINSONIAN SYNDROMES
Drug-induced encephalopathy/parkinsonism Relevant drug exposure (neuroleptic, antiemetic)
Vascular parkinsonism Multiple subcortical infarcts; may mimic PSP
Normal-pressure hydrocephalus Prominent gait disturbance (magnetic), urinary incontinence, subcortical dementia
Whipple disease May mimic PSP; cerebrospinal fluid pleocytosis, gastrointestinal symptoms prominent
Dementia pugilistica Repetitive head trauma ± cavum septum pellucidum on MRI
INHERITED METABOLIC DISORDERS
Wilson disease Autosomal recessive; early onset, impaired copper clearance
Neurodegeneration with brain iron accumulation (Hallervorden-Spatz disease) Familial and sporadic disorders with variable age of onset and subcortical iron deposits
Idiopathic basal ganglia calcification Autosomal dominant and recessive; subcortical calcium deposits
Parkinson disease with dementia (PDD) Parkinsonism initially, later onset of dementia

Classification

Parkinsonian dementia syndromes encompass a broad range of etiological disorders that share a heterogeneous combination of cognitive and motor features (see Table 66.4). The two main etiological categories are primary degenerative syndromes, representing disorders that typically are either sporadic or inherited, and secondary parkinsonian syndromes, attributable to a variety of cerebral insults including drug-induced cognitive-motor syndromes. An intermediate (and less common) category of disorders that feature extrapyramidal dysfunction and associated cognitive alterations includes systemic metabolic derangements involving the excess accumulation of heavy metals or minerals in basal ganglia structures.

In addition, the primary degenerative syndromes may be parsed into two broad molecular families: synucleinopathies, consisting of dementia with Lewy bodies (DLB), Parkinson disease dementia (PDD), and multisystem atrophy (MSA); and tauopathies, consisting of corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), and familial FTD.

Synucleinopathies

Dementia with Lewy Bodies

Diagnostic Criteria

Parkinson disease (PD), PDD, and DLB are neurodegenerative disorders from the Lewy body spectrum disorders. DLB achieved consensus recognition as a distinct clinicopathological entity in 1996 (McKeith et al., 1996). Our conceptualization of it continues to evolve (McKeith et al., 1999, 2000b, 2005). From a nosological standpoint, DLB is conceived as being distinct from AD and PD, but substantial clinical and pathological overlap with both AD and PD is recognized, making the relationship among these disorders controversial. In the recently revised diagnostic criteria (McKeith et al., 2005) (Table 66.6), two additional clinical features have been designated as “suggestive”: rapid eye movement (REM) sleep behavior disorder and neuroleptic sensitivity. Either of these two features can count as a core feature if at least one core feature is present.

Table 66.6 Consensus Diagnostic Criteria for Dementia with Lewy Bodies

Symptom/Sign Cardinal Manifestations Frequency
Dementia Attentional, frontal-executive, and visuospatial deficits, often worse than in Alzheimer disease; short-term (episodic) memory relatively better than in Alzheimer disease 100%
CORE FEATURES
Fluctuating cognition Variable timing of altered level of attention or arousal; distinct from sundowning 60%-80%
Visual hallucinations Recurrent; typically involve animate subjects; variable degree of insight; reminiscent of anticholinergic delirium 50%-75%
Parkinsonian motor signs Spontaneous; rigidity and bradykinesia most common; intention tremor more common than resting tremor 80%-90%
SUGGESTIVE FEATURES
REM sleep behavior disorder Loss of atonia during REM sleep; individuals appear to act out dreams; may be combative or violent 25%-50%
Neuroleptic sensitivity Severe cognitive and motor adverse reaction to antipsychotic agents; may increase mortality 50%
Decreased tracer uptake

  SUPPORTIVE FEATURES (COMMON BUT LACKING DIAGNOSTIC SPECIFICITY)

CT, Computed tomography; EEG, electroencephalogram; MIBG, metaiodobenzylguanidine; MRI, magnetic resonance imaging; PET, positron emission tomography; REM, rapid eye movement; SPECT, single-photon emission computed tomography.

Modified from McKeith, I.G., Dickson, D.W., Lowe, J., et al., 2005. Dementia with Lewy bodies: diagnosis and management: third report of the DLB Consortium. Neurology 65, 1863-1872.

Epidemiology

DLB is found at autopsy in approximately 20% of late-onset dementias (alone or in combination with AD). In one European study, probable DLB was diagnosed in 15.8% and possible DLB in 4.1% of all dementia cases (Aarsland et al., 2008b). The estimated population-based prevalence of DLB is between 10 and 200/10,000 (Brayne et al., 2006). The mean age of onset is 75 years, with a range of 50 to 80 years. There is a slight male predominance (Geser et al., 2005). Older age at onset, fluctuating cognition and hallucinations at disease onset, and associated AD pathology herald shorter survival (Jellinger et al., 2007).

Clinical Presentation

Although the dementia syndrome of DLB is similar enough to AD that many patients will warrant both diagnoses, marked variability in the clinical presentation is characteristic. The initial manifestations of DLB may be cognitive deficits or any of the core features of DLB alone or in conjunction with a dementia syndrome (see Table 66.6).

Fluctuating cognition is observed in up to 90% (Geser et al., 2005) and should be routinely solicited from the informant. Fluctuations in attention, arousal, or cognitive performance can lead to high variability in cognitive performance on formal testing.

Psychotic symptoms (delusions and hallucinations) are present in up to 75% of DLB patients. Unlike AD, DLB patients can present with hallucinations prior to dementia onset. Visual hallucinations are the most frequent neuropsychiatric symptom in DLB, occurring in up to 63% of patients. They are typically complex, detailed, brightly colored, three-dimensional images of people and animals. Less frequent themes are hallucinations of children, objects, insects, fire, and birds. In about half of patients, they co-occur with auditory hallucinations. Hallucinations may leave the patient indifferent or elicit various emotions—fear, amusement, or anger. A diurnal pattern with more (and/or more severe) hallucinations in the evening and at night, accentuating the impact of solitude, inactivity, and poor lightning, is evident in a third of patients with DLB.

Delusions are common in DLB and can occur in up to half of patients. The most common themes are delusional misidentification, followed by paranoid beliefs (theft, conspiracy, harassment, abandonment, infidelity) and phantom boarder. Nondelusional suspiciousness and paranoia are also frequent. Delusional misidentifications can help differentiate DLB from AD. Mistaking TV personalities for home visitors, Capgras syndrome (belief that a family member has been replaced by an imposter), mistaking one’s mirror image for another person, and delusion of their residence not being their home are significantly more common in DLB relative to AD.

DLB patients frequently display REM sleep behavior disorder (RBD) with loss of physiological skeletal muscle tone during REM cycles and dream enactment. RBD can be an early prodrome of DLB symptom predating dementia or extrapyramidal feature onset by decades. The positive predictive value for DLB in the demented patient with parkinsonism and RBD is 92% (Boeve et al., 2001). RBD is also common in other α-synucleinopathies including PD, PDD, and MSA. In addition to RBD, DLB patients also suffer from decreased sleep efficiency, increased upper airway resistance, obstructive sleep apnea/hypopnea, and periodic limb movements of sleep (Boeve et al., 2003).

The typical parkinsonian features in DLB consist of symmetrical rigidity and bradykinesia. Intention tremor is more common than rest tremor. Asymmetric presentation is relatively uncommon, in contradistinction to PDD. The distinction of DLB from PDD has been arbitrarily based on the relative temporal onset of core features. DLB is defined as cognitive or neuropsychiatric features (or both) appearing within 1 year of motor signs. If motor signs appear more than 1 year before cognitive or neuropsychiatric symptoms, patients are typically diagnosed with PDD.

A caveat in diagnosing DLB involves differentiating disease manifestations from iatrogenic drug side effects. Dopaminergic agents commonly used to treat motor dysfunction may provoke or exacerbate psychosis. Antipsychotic agents may induce or worsen motor disability. Careful questioning regarding the relative temporal onset of various signs and symptoms and their possible relationship to medication adjustment is important. The complex interplay of disease effects and treatment side effects often poses an ongoing challenge in the management of DLB.

Dysautonomia is usually seen later in the disease course, although some cases with dysautonomia at initial presentation have been reported, as well as frequent falls, dysphagia, and urinary incontinence. A rapidly progressive course with aphasia, dyspraxia, and severe visuospatial disturbances can be seen. Some DLB patients progress to end stages and death within 1 to 2 years (Geser et al., 2005).

Laboratory Studies

There are no specific blood or CSF tests for DLB to date. Recently, increased levels of CSF total tau have been associated with shorter survival (Bostrom et al., 2009). Increase in the oxidized α-helical form of Aβ1-40 (Aβ1-40*) yielded a diagnostic sensitivity and specificity of 81% and 71% in differentiating DLB and PDD from AD (Bibl et al., 2006). Research efforts toward the development of reliable fluid biomarkers for DLB are underway.

Genetics

The genetics of DLB has been poorly understood. Familial DLB in a Belgian kindred has been recently mapped to chromosome 2q35-q36 (Meeus et al., 2010). DLB has been also associated with duplications in the α-synuclein (SNCA) gene on chromosome 4 (Kasuga et al., 2010) and the leucine-rich repeat kinase 2 (LRRK2) gene on chromosome 12 (Qing et al., 2009). It has been suggested that oxidative stress up-regulates both SNCA and LRRK2 expression in DLB (Qing et al., 2009). Mutations in the glucocerebrosidase (GBA) gene on chromosome 1 have been associated with both DLB and PD (Mata et al., 2008).

Neuroimaging

Structural neuroimaging (MRI or CT) can reveal relatively nonspecific generalized brain atrophy with typically modest hippocampal involvement in DLB (Fig. 66.12). Several research groups have demonstrated diffuse temporal, parietal, and frontal cortical atrophy (Ballmaier et al., 2004; Beyer et al., 2007b; Burton et al., 2002) as well as atrophic changes of the dorsal midbrain, hypothalamus, and substantia innominata (Whitwell et al., 2007).

Functional brain imaging offers the prospect of aiding the clinical diagnosis of DLB. Occipital lobe hypoperfusion on SPECT or PET (Fig. 66.13) differentiates DLB from AD with a sensitivity of 65% and specificity of 87% (Lobotesis et al., 2001). A PET study of autopsy-confirmed AD and DLB subjects also demonstrated that the presence of occipital hypometabolism differentiated DLB from AD with a sensitivity of 90% and specificity of 87% (Minoshima et al., 2001). Unfortunately occipital hypometabolism is not always present in DLB patients hence its diagnostic contribution is somewhat limited.

The most established functional neuroimaging biomarker in DLB is dopamine transporter imaging. [123I]-FP-CIT SPECT was reported to show 78% sensitivity and 90% specificity for differentiating DLB from non-DLB dementia (McKeith et al., 2007). Evidence of dopaminergic abnormalities in the basal ganglia has been accepted as a suggestive feature for DLB in the revised McKeith criteria (McKeith et al., 2005).

Myocardial iodine-131-meta-iodobenzylguanidine (MIBG) scintigraphy is another imaging modality that may help with DLB diagnosis. Recently Estorch et al. reported sensitivity of 94% and a specificity of 96% of MIBG scanning in predicting DLB diagnosis in MCI subjects (Estorch et al., 2008).

Treatment

Antipsychotic Agents

The use of any antipsychotic agent in DLB or PDD must be approached with caution. Use of typical antipsychotic agents (e.g., haloperidol) is discouraged in DLB because a syndrome of severe motor, cognitive, and autonomic dysfunction has been reported in 50% or more of persons with DLB exposed to such agents. The syndrome of heightened neuroleptic sensitivity increases the risk of death and may provoke rapid inexorable clinical decline. Although neuroleptic sensitivity has been reported to occur with atypical antipsychotic agents, and relative risk is believed to be lower, comparative studies are still lacking.

Quetiapine (Seroquel) is a relatively new atypical antipsychotic agent with potential benefit for treating psychosis in parkinsonian syndromes. A 12-week open-label trial of quetiapine in 151 persons with PD and other late-life psychotic disorders showed significant improvement in overall neuropsychiatric symptom burden and global disease severity rating at a median dose of 100 mg/day (McManus et al., 1999). Somnolence, dizziness, and postural hypotension were the most common side effects reported, with extrapyramidal side effects occurring in only 6% of subjects. The dose range of quetiapine varies widely, from 12.5 to 400 mg/day, based on individual differences in sensitivity and tolerability, with excess sedation being the most common dose-limiting side effect.

Clozapine (Clozaril) also may be an effective treatment for refractory psychosis in PD; however, the requirement for periodic blood monitoring because of the risk of agranulocytosis prohibits its use as a first-line agent.

Aripiprazole (Abilify) and ziprasidone (Geodon) are newer atypical antipsychotic agents that are associated with relatively low risk of exacerbating parkinsonian motor dysfunction. Little data are available as yet to support their use in DLB or PDD.

The increased risk of mortality in dementia associated with the use of three of the novel antipsychotics—risperidone, olanzapine, and quetiapine—to treat agitation in AD (Schneider et al., 2006) resulted in a “black box” warning for several of these agents.

Some physicians advocate use of AChEIs as first-line agents for treatment of psychosis in DLB, but supporting data are lacking, although use of AChEIs for control of the cognitive symptoms of DLB (and PD) is widespread.

Dopaminergic Therapy

Initial reports of patients with DLB being relatively unresponsive to l-dopa treatment may have resulted in part from inadequate dosing. Fear of exacerbating neuropsychiatric symptoms in PDD or DLB has made physicians cautious about using higher doses. Studies examining the response of parkinsonian motor signs in DLB to l-dopa suggest a significant proportion of responders. In view of the prevalence of DLB and associated functional motor disability, controlled studies with l-dopa for treating parkinsonian motor features of DLB are needed. In the absence of scientific data, it probably is best to initiate treatment with carbidopa plus l-dopa at doses of 12.5 mg and 50 mg, respectively, twice a day, with gradual dose titration as tolerated or needed to achieve a therapeutic clinical response. Virtually no data are available on the use of dopamine agonists such as pramipexole (Mirapex), ropinirole (Requip), bromocriptine (Parlodel), or pergolide (Permax) in DLB. Although pramipexole and ropinirole are used by many clinicians for initial therapy in PD, the side effects of somnolence and sleep attacks may complicate DLB symptoms. Selegiline (Eldepryl) carries a risk of exacerbating psychosis and usually has only a modest effect on motor dysfunction, so its use in DLB is not prudent. Entacapone (Comtan), a catechol-O-methyltransferase (COMT) inhibitor, is used exclusively in conjunction with l-dopa. Amantadine (Symmetrel), an NMDA antagonist with anticholinergic properties, should be used cautiously if at all. Anticholinergic agents such as benztropine (Cogentin) and trihexyphenidyl (Artane) should be avoided. Memantine (Namenda), a congener of amantadine, was used to treat parkinsonian motor dysfunction in Europe before its approval as an antidementia agent. Empirical data are limited regarding its use in DLB or PDD.

Two drugs commonly used to treat urinary incontinence are oxybutynin (Ditropan) and tolterodine (Detrol). Both agents are strongly anticholinergic, so their use in DLB (and other parkinsonian dementia syndromes) should be reserved for circumstances in which the benefits of treatment outweigh the burden of nontreatment and potential for adverse cognitive side effects. Newer medications for incontinence that work through different neurotransmitter systems may prove more efficacious for treatment of incontinence associated with DLB and PDD.

Parkinson Disease Dementia

Idiopathic PD, the most common neurodegenerative movement disorder, results from a defect in the metabolism of α-synuclein. In addition to the classic motor features of rigidity, bradykinesia, tremor, and postural abnormalities, individuals with PD frequently display a host of non-motor features. Progressive cognitive impairment and behavioral problems are commonly seen in PD patients even early in the disease course (Janvin et al., 2006a, 2006b; Muslimovic et al., 2005).

Epidemiology

PD affects 1 in 100 people over the age of 60 (Galvin, 2006; Poewe and Wenning, 1998). PD incidence and prevalence increase with age and are greater in males. According to a recent meta-analysis, the worldwide incidence and prevalence of PD are 1.6/1000 and 9.5/1000, respectively, among the elderly age 65 years and older (Hirtz et al., 2007).

Up to 90% of PD patients develop overt dementia (PDD) (Buter et al., 2008). Cognitive impairment in PD is perhaps one of the most understudied non-motor syndromes. The onset of cognitive decline in relation to initial PD presentation is highly variable (Aarsland et al., 2007), suggesting individually predetermined neuronal vulnerability, with the additional contribution of environmental factors and comorbidities.

Clinical Presentation

PD manifests with four cardinal symptoms: tremor, rigidity, bradykinesia, and postural instability. Progressive cognitive impairment and behavioral problems are commonly seen in PD patients even early in the disease course (Foltynie et al., 2004). Individuals with PD are at two- to sixfold increased risk for developing PDD relative to elderly normal controls (Breteler et al., 1995; Rajput et al., 1987). Both overt dementia (Aarsland et al., 2003) and a predementia state (i.e., PDMCI) (Janvin et al., 2003) have been described in PD.

PDD is characterized by pronounced slowing of cognitive and motor skills, episodic memory deficits, and executive, attentional, and visuospatial dysfunction (Aarsland et al., 2008a; Bronnick et al., 2007; Camicioli and Fisher, 2004; Galvin, 2006). Behavioral disturbances are common in PDD. Visual hallucinations and RBD are frequently seen and have been shown to correlate with executive dysfunction and attention deficits (Barnes and Boubert, 2008).

PDMCI is associated with impaired visuospatial and executive function, but recently, deficits in episodic memory have also been reported (Aarsland et al., 2009; Dubois and Pillon, 1997; Foltynie et al., 2004; Janvin et al., 2003; Muslimovic et al., 2005). PD patients who show cognitive decline consistent with PDMCI are five times more likely to receive a diagnosis of dementia over the next 4 years (Janvin et al., 2006a). There is some evidence that tests of attention and executive function may predict conversion from PDMCI to PDD (Janvin et al., 2006a; Williams-Gray et al., 2007), but these data need to be expanded upon and replicated. Other risk factors for future development of dementia include increasing age, greater motor disability, significant bradykinesia, presence of postural instability and significant gait disorder, as well as early hallucinations (Aarsland et al., 2001, 2003; Alves et al., 2006; Mayeux et al., 1992).

Genetics

In the last decade, several causative genetic mutations for both early- and late-onset PD have been identified. Mutations in the synuclein (SNCA or PARK1) gene on chromosome 4, the parkin (PARK2) gene on chromosome 12, the DJ-1 (PARK7) gene on chromosome1, or the leucine-rich repeat kinase 2 (LRRK2 or PARK8) gene on chromosome 12 can cause both early- and late-onset PD (Klein et al., 2009). Recently the glucosidase (GBA) gene on chromosome 1, coding the enzyme deficient in Gaucher disease, has gained a lot of attention. PD patients are in fact five times more likely to carry mutations in GBA relative to the normal population. GBA mutations are now received as the most common genetic cause of PD (Velayati et al., 2010).

Neuroimaging

To date, structural imaging has not been regarded as diagnostically useful in PD and PDD. The role of structural MRI in PD is confined to ruling out other etiologies such as basal ganglia stroke lesions, diffuse white-matter ischemic changes, midbrain atrophy, or strikingly unilateral frontoparietal atrophy that could suggest PSP or CBD, respectively. However, several recent studies have documented that structural differences can be seen in PD and especially in PDD. There seems to be widespread cortical atrophy of the limbic, temporal, parietal, frontal, and occipital regions in PDD relative to normal controls and nondemented PD subjects (Beyer et al., 2007a; Burton et al., 2004; Hwang et al., 2010). PDMCI subjects were also reported to show frontal and temporal atrophy (Beyer et al., 2007a; Meyer et al., 2007). Other structural changes associated with cognitive decline in PD are ventricular enlargement and caudate atrophy (Apostolova et al., 2010a; Meyer et al., 2007).

An FDG-PET pattern similar to the one seen in AD, with hypometabolism affecting the posterior cingulate, parietal, and temporal cortices, has been reported in PDD (Brooks, 2010). Striatal [123I]-FP-CIT SPECT binding is reduced to a similar degree in PD, PDD, and DLB versus AD. (O’Brien et al., 2004).

Multiple pathological substrates may contribute to the cognitive and neuropsychiatric features of PDD. These include neurotransmitter deficiencies in dopamine (substantia nigra and ventral tegmental area), norepinephrine (locus ceruleus), and serotonin (raphe nuclei), as well as Lewy bodies in cortical and subcortical regions. Bohnen et al. conducted PET imaging with a radiolabeled acetylcoline analogue and found cholinegric deficits of 70% or higher in the cerebral cortex in PDD. Nondemented patients with PD also demonstrated marked cholinergic deficits relative to both control subjects and patients with AD (Bohnen et al., 2003). Whether marked cholinergic deficit is a prognostic sign of impending dementia in PD remains to be determined.

Pathology

The pathology underlying cognitive deficits in PD is not yet completely understood but seems to include both neurotransmitter deficits (dopamine, serotonin, acetylcholine, noradrenalin), structural changes, and cortical Lewy body (LB) deposits (Braak et al., 2005; Emre, 2003). The pathological changes in PDMCI and PDD consist of progressive degeneration of the dopaminergic and other neurotransmitter systems and dysfunction of multiple neuronal circuits (Perry et al., 1991). On gross macroscopic inspection, the brain frequently shows mild to moderate atrophy and ventriculomegaly. Brainstem dissection reveals the pathognomonic macroscopic signs of PD: pallor of the substantia nigra and locus coeruleus. Histopathologically, the neurodegenerative changes of PD consist of abundant LB, Lewy neurites, and neuronal loss in the midbrain, substantia nigra, and locus coeruleus, with associated depletion of pigmented neurons (Braak et al., 2005; Emre, 2003). In addition, widespread argyrophilic α-synuclein-positive tau-negative glial inclusions can be seen diffusely in the cerebral cortex, basal ganglia, brainstem, cerebellum, and even in the spinal cord (Jellinger and Mizuno, 2003).

Postmortem investigations in PDD have demonstrated several neuropathological findings: cortical LB pathology, degenerative changes in critical cortico-subcortical circuits, and coexistent AD pathology (Jellinger and Mizuno, 2003). Individuals with PD who develop dementia late in the disease course tend to show diffuse LB but only modest AD pathology (Apaydin et al., 2002). Brains of PDD subjects show significantly higher densities of LB and Lewy neurites in the entorhinal cortex as well as the CA2-3 hippocampal subregions and the amygdala relative to brains of nondemented PD subjects (Bertrand et al., 2004).

The extent of contribution of AD pathology to PDD has proven difficult to understand. Concomitant AD pathology (i.e., neuritic plaques, NFTs) is seen in over 50% of PDD subjects on postmortem examination (Jellinger, 2001) and contributes to cognitive decline (Harding and Halliday, 2001). Nondemented PD (PDND) typically exhibits AD changes corresponding to Braak staging of IV or less (Braak and Braak, 1991), whereas PDD subjects usually harbor advanced Braak stage cortical pathology (V-VI) (Delacourte et al., 1999). AD pathology in PDD has been associated with much shorter survival (Jellinger et al., 2002).

Cortico-subcortical circuit degeneration in PDD manifests in reduced prefrontal choline acetyltransferase activity, dopaminergic system hypofunction, and D1 receptor decline in the caudate nuclei. These changes are seen even in the absence of AD pathology (Bruck et al., 2001; Mattila et al., 2001). Additionally, the involvement of the noradrenergic and serotoninergic systems has been implicated for the development of cognitive and behavioral changes in PD (Gerlach et al., 2001; Jellinger, 1999). The relationship between cholinergic depletion, PDD, and visual hallucinations is particularly intriguing. Recent research has shown that there is more severe cholinergic depletion in PDD compared to AD, which appears to be related to cognitive impairment (Bohnen et al., 2003, 2006). In addition, PD patients with hallucinations were found to respond better to cholinesterase inhibitors than those without (Burn et al., 2006).

Multisystem Atrophy

Multisystem atrophy refers to a degenerative parkinsonian disorder with variable associated features including autonomic, cerebellar, and pyramidal tract dysfunction. The prevalence of MSA has been estimated to be 4.4 cases per 100,000, making it slightly less common than PSP (Schrag et al., 1999). Three clinical variants of MSA were initially recognized: (1) striatonigral degeneration, (2) Shy-Drager syndrome, and (3) olivopontocerebellar atrophy (OPCA). All share a common pathological substrate of α-synuclein–containing glial cytoplasmic inclusions, which are distributed variably in the cortex, subcortical regions, cerebellum, spinal cord, and dorsal root ganglia. Newer consensus diagnostic criteria for MSA (Gilman et al., 1998) have suggested two main types based on the predominant clinical feature: (1) MSA-parkinsonian (MSA-P) and (2) MSA-cerebellar (MSA-C). Accordingly, previously described cases of striatonigral degeneration and Shy-Drager syndrome are now classified as MSA-P, and sporadic OPCA conforms to MSA-C. Of importance, however, most cases of MSA have some degree of parkinsonism and autonomic dysfunction, and about half will have either cerebellar or pyramidal tract dysfunction. RBD is associated with MSA, just as with PD and DLB, suggesting that this disorder may be a common manifestation of synucleinopathies.

In MSA-P, neuronal loss typically is severe in the putamen and substantia nigra compared with PD, and the disorder generally is distinguished on clinical grounds by the absence of a resting tremor, more severe autonomic dysfunction, and evidence of pyramidal tract involvement (e.g., spasticity, presence of a Babinski sign). Cognitive functioning in MSA-P is relatively preserved, although executive deficits characteristically are observed on formal neuropsychological testing. Severe dysautonomic manifestations early in the course, such as orthostatic hypotension, impotence, urinary problems, constipation, and hyperhidrosis, also are common.

MSA-C (formerly sporadic OPCA) is distinguished primarily by the presence of dysarthria and ataxia in association with marked cerebellopontine atrophy on brain MRI. Parkinsonian motor signs and autonomic dysfunction are present to a variable degree. Impaired saccadic eye movements and vertical gaze palsy also may occur. Pathologically distinct hereditary forms of OPCA (MCA-C) due to trinucleotide repeat have been described. These have been also be classified as a hereditary spinocerebellar ataxia syndrome. Distinguishing clinical signs of hereditary forms of OPCA may include concomitant optic atrophy, retinal degeneration, neuropathic signs, or spastic paraparesis.

MSA can show several characteristic features on MRI. These include atrophy and hyperintensity of the putamen, slit-like hyperintensity of the posterolateral margin of the putamen, brainstem atrophy, hyperintensity of the middle cerebellar peduncles, and cruciform hyperintensity of the pons (the so-called hot cross bun sign) (Fig. 66.15). As expected MSA-C also shows significant cerebellar atrophy.

Pathologically MSA is characterized by extensive oligodendroglial α-synuclein pathology known as glial cytoplasmic inclusions. Dystrophic neurites can be found in the putamen, inferior olive, and brainstem nuclei (Fig. 66.16).

Patients with MSA-P initially may respond to l-dopa, but the duration of benefit typically is short lived, and treatment in many cases is not well tolerated. Orthostatic hypotension may be particularly disabling and can be exacerbated by l-dopa and other dopaminergic drugs. Fludrocortisone (Florinef) and midodrine (Proamatine) often provide a measure of symptomatic relief.

Tauopathies

Corticobasal Degeneration

The disorder now commonly referred to as corticobasal degeneration (CBD) initially was described by Rebeiz and colleagues in the 1960s as “corticodentatonigral degeneration with neuronal achromasia.” CBD is a relatively rare but distinctive parkinsonian dementia syndrome characterized by unilateral or asymmetrical signs of parkinsonian rigidity, myoclonus, dystonia, and apraxia, often associated with myoclonic jerks, grasp reflex, cortical sensory signs such as hemineglect, agraphesthesia and astereognosis, and hyperreflexia. Limb apraxia occurs in the majority of affected persons, and many also exhibit “alien hand” features such as levitation of the affected limb, lacking the feeling of owning the limb, forced groping, utilization behavior, and intermanual conflict. During the course of the disease, unilateral parkinsonian features or gait disorder will develop in virtually all patients. CBD affects both genders and all races.

MRI frequently helps with the differential diagnosis in CBD. CBD patients usually show asymmetrical frontoparietal atrophy affecting the motor and sensory cortices, which is more severe on the side contralateral to the affected limb. High T1 signal intensity in the subthalamic nucleus, midbrain atrophy, and T2 striatal hypointensity have also been reported (Sitburana and Ondo, 2009; Tokumaru et al., 2009). As expected, functional imaging with PET and SPECT show strikingly asymmetrical hypoactivity contralateral to the affected side (Fig. 66.17).

The pathological hallmarks of CBD are astrocytic gliosis, most prominent in the superficial cortical layers and at the grey/white junction, and swollen achromatic neurons (neuronal achromasia) that are distributed asymmetrically in discrete (frontal or parietal) cortical areas and in subcortical regions (Fig. 66.18). Several types of intraneuronal tau-immunoreactive inclusions are seen in the cortex, thalamus, basal ganglia, substantia nigra, and locus coeruleus. These are commonly seen as densely packed, small, globose NFT-like inclusions and dispersed filamentous inclusions. Tau-immunoreactive cell processes, oligodendroglial coiled bodies, and astrocytic plaque-like lesions are characteristic for CBD.

Parkinsonian motor signs in CBD show a modest response to l-dopa, with approximately 25% of patients who received this treatment showing some benefit (Kompoliti et al., 1998). Clonazepam may be useful for treating myoclonus and dystonia in some patients. Limited clinical experience with AChEI therapy in CBD generally has been unrewarding.

Progressive Supranuclear Palsy

PSP (Steele-Richardson-Olszewski syndrome) is distinguished clinically from PD by the absence of a rest tremor, greater axial (neck and trunk) than limb rigidity, the presence of severe dysarthria and dysphagia as a result of the supranuclear palsy, a greater degree of gait and balance impairment, and limitation in voluntary downward gaze that can be overcome with the doll’s eyes head maneuver (“supranuclear” gaze palsy). Prevalence estimates of PSP based on community studies are as high as 6.4 per 100,000 (Schrag et al., 1999). Clinicopathological studies often find this entity misdiagnosed as PD. Standardized clinical criteria for diagnosing PSP, derived from symptoms in histopathologically proven cases, have been proposed (Box 66.9). High sensitivity and specificity for these criteria have been demonstrated in an independent clinical sample with autopsy-confirmed diagnosis (Lopez et al., 1999).

Early postural instability and falls, vertical supranuclear palsy with downgaze paresis, and akinetic-rigid symmetrical parkinsonism are the most common clinical features. Myoclonus, resting tremor, and pyramidal tract signs are rare. Cognitive symptoms reflecting frontal lobe dysfunction and a subcortical dementia typically appear. Most but not all patients develop clinically significant dementia. Apathy, dysphoria, and anxiety are common neuropsychiatric concomitants of PSP. Pathological laughing and crying (pseudobulbar affect) frequently are present and have to be distinguished from a primary mood disturbance.

Gross atrophy of the midbrain tegmentum may be evident on MRI (Boxer et al., 2006; Oba et al., 2005). Additional features suggestive of PSP include enlargement of the third ventricle and signal increase in the midbrain and inferior olives. Mild to moderate frontal and temporal cortical atrophy can be seen in PSP, never quite reaching the severity of cortical atrophy seen in CBD (Fig. 66.19). Recently, using susceptibility-weighted MRI, one research group reported that PSP could be distinguished from MSA and idiopathic PD based on hypointensity of the red nucleus and putamen, respectively (Gupta et al., 2010).

The histopathological hallmark of PSP are globose NFTs composed of aggregated 4R tau protein that accumulate in prefrontal cortex and a number of subcortical regions, principally the globus pallidus, substantia nigra, and subthalamic nucleus (Dickson, 2009) (Fig. 66.20). Tau-reactive tangles also occur in glial cells. Reactive gliosis and neuronal loss are variable features. The localization of tau pathology in PSP corresponds well to the clinical phenotype. Severe brainstem involvement is seen in the akinetic type, while dementia and CBD-like presentation and/or speech apraxia are commonly associated with cortical tau pathology. Characteristic features are the tufted astrocytes usually found in motor cortex and striatum, the abundance of thread-like processes, and oligodendroglial coiled bodies. This is distinct from CBD, where threads but not oligodendroglial coiled bodies are seen (Dickson, 2009).

Symptomatic pharmacological therapy of PSP has had very limited success. Although occasional modest and transient improvement in motor function has been observed with l-dopa and other dopaminergic agonists, adverse effects (i.e., orthostatic hypotension, psychosis) often outweigh the benefits. Similar observations with the AChEI, donepezil, have been reported. In a placebo-controlled double-blind study involving 21 patients with PSP, modest improvement in memory test scores was offset by deterioration in functional mobility (Litvan et al., 2001). Depression, anxiety, and pseudobulbar affect may respond to SSRI agents, which generally are well tolerated.

Metabolic Parkinsonian Disorders

Wilson Disease

Wilson disease (WD), or hepatolenticular degeneration, is a systemic disorder of copper metabolism with an autosomal recessive pattern of inheritance. Although a relatively infrequent cause of dementia and parkinsonism, it is one etiological disorder for which diagnostic peripheral biomarkers and disease-reversing treatment are available. Approximately 40% to 50% of patients with WD present with neurological signs in the second or third decade of life, with parkinsonian signs (tremor, dystonia, and rigidity) and cerebellar signs (dysarthria, clumsiness, and unsteady gait) being most common. Limb tremors of all types may be present, including a proximal irregular “wing-beating” tremor and facial dystonia, often producing a “sardonic” smile. Cognitive abnormalities and a variety of neuropsychiatric disturbances (anxiety, depression, impulsiveness, disinhibition, paranoid delusions) often accompany motor disturbances.

Kayser-Fleischer rings composed of copper-containing granules in the periphery of the cornea almost invariably are present in cases of WD with neurological signs and are visualized reliably by slit-lamp examination. MRI abnormalities typically are evident on T2-weighted images—bilateral symmetrical signal hyperintensities in the basal ganglia, midbrain, pons, and thalamus (Fig. 66.21).

The genetic defect in WD results in markedly diminished levels of ceruloplasmin, the plasma transport protein for copper. Although this protein can be measured in serum, demonstrating elevated copper excretion in a 24-hour urine specimen is a more reliable and definitive test for symptomatic WD. Standard treatments to minimize the neurotoxic effects of excessive systemic copper include dietary restriction and d-penicillamine.

Neurodegeneration with Brain Iron Accumulation

Neurodegeneration with brain iron accumulation (NBIA) was initially described in 1922 as an autosomal recessive childhood-onset parkinsonian dementia associated with rusty brown discoloration of the medial globus pallidus and substantia nigra pars reticulata at autopsy. A variety of atypical forms have been included under the rubric of NBIA and referred to as NBIA syndrome. These tend to present as later-onset dementia, dystonia, and parkinsonism and have associated basal ganglia iron deposition and neuroaxonal dystrophy. Virtually all patients with classic NBIA and approximately 33% to 50% of those with later-onset neuroaxonal dystrophy, or NBIA syndrome, have a mutation in the gene encoding pantothenate kinase 2 (PANK2) located on chromosome 20 (Hayflick, 2003). Compared with affected persons with the late-onset syndrome who test negative for PANK2 mutation, patients with late-onset syndrome and a mutation at the PANK2 locus can exhibit more severe speech and psychiatric disturbances. Persons with a PANK2 mutation, whether associated with early-onset classic NBIA or a later-onset syndrome of NBIA, can exhibit the “eye-of-the-tiger” sign (central hyperintensity surrounded by an area of hypointensity) in the globus pallidus on T2-weighted MRI. There is currently no available therapy for NBIA; management is largely symptomatic and supportive.