Sie sind auf Seite 1von 13

Expert Review of Neurotherapeutics

ISSN: 1473-7175 (Print) 1744-8360 (Online) Journal homepage: http://www.tandfonline.com/loi/iern20

Diagnostic challenges in rapidly progressive


dementia

Inga Zerr & Peter Hermann

To cite this article: Inga Zerr & Peter Hermann (2018): Diagnostic challenges in rapidly
progressive dementia, Expert Review of Neurotherapeutics, DOI: 10.1080/14737175.2018.1519397

To link to this article: https://doi.org/10.1080/14737175.2018.1519397

Accepted author version posted online: 03


Sep 2018.
Published online: 17 Sep 2018.

Submit your article to this journal

Article views: 28

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iern20
EXPERT REVIEW OF NEUROTHERAPEUTICS
https://doi.org/10.1080/14737175.2018.1519397

REVIEW

Diagnostic challenges in rapidly progressive dementia


Inga Zerr and Peter Hermann
Clinical Dementia Center and National TSE Reference Center, Department of Neurology, Goettingen University Medical Center, Goettingen,
Germany

ABSTRACT ARTICLE HISTORY


Introduction: Rapidly progressive dementia is a syndrome caused by numerous disease entities. Accurate Received 19 July 2018
diagnosis is crucial as substantial proportion of these diseases is highly treatable. Others might implicate Accepted 30 August 2018
specific hygienic problems. Still, differential diagnosis remains challenging because of a huge overlap of clinical KEYWORDS
presentations. Dementia; rapidly
Areas covered: The paper reviews PubMed-listed research articles with a focus on diagnosis and progressive dementia; prion
treatment of diseases showing rapid cognitive decline such as inflammatory diseases, rapidly progres- diseases; neurodegenerative
sive neurodegenerative diseases, toxic-metabolic encephalopathies and prion diseases. The literature diseases; encephalitis;
was interpreted in the light of experience in clinically differentiating rapid progressing dementia in the differential diagnosis
framework of Creutzfeldt-Jakob-Disease (CJD) surveillance activities. An overview of relevant differential
diagnoses and diagnostic pitfalls as well as therapeutic protocols is presented.
Expert commentary: Over the last years, more and more neurologic disorders causing cognitive
symptoms, in particular various types of immune-mediated diseases have been discovered. To identify
treatable conditions and to enhance knowledge of differential diagnosis and epidemiology, we suggest
an extended diagnostic work up in cases with rapidly progressing dementia. Besides standard methods,
this should include the search for neoplasia as well as atypical encephalitis. High-dose steroid therapy
should be considered in certain clinical situations even when no evidence for inflammation is present.

1. Introduction Biochemical studies of amyloid-β, α-synuclein, tau, and


other proteins involved in the pathophysiology of neurode-
The precise definition of rapid progression in dementia (RPD) is
generative diseases revealed that they share key characteris-
lacking. Although cognitive decline may progress over a very short
tics with prions [6]. To characterize these proteins and their
period of time (days to weeks) in Creutzfeldt–Jakob disease (CJD)
associated diseases, various terms like prion, prion-like, or prio-
or encephalitis, one of the most common definitions implicates a
noid have been suggested but none of them has been widely
progression from the onset of the first symptom to dementia in
accepted, yet. With respect to this, we use the term neurode-
less than 2 years [1]. This is in line with the definition of rapid
generative disease throughout this article.
cognitive decline (RCD) used in clinical criteria for CJD [2]. On the
other hand, definitions using cognitive scores have been proposed
for specific diseases. Rapidly progressive Alzheimer’s disease 2. Epidemiology of RPD
(rpAD) can be defined by a loss of ≥6 MMSE points per year in
Because of different definitions, valid data on the prevalence of
patients with probable Alzheimer’s disease (AD) [3,4]. Rapid pro-
RPD are not available. Former observations from prion disease
gressive forms in other neurodegenerative diseases are increas-
reference centers report that sporadic CJD is the most common
ingly recognized, e.g. they have been reported for dementia with
cause for RPD followed by AD [7,8] but these figures are highly
Lewy bodies (DLB), as well [5]. Although CJD and neurodegenera-
biased for obvious reasons. Some studies performed by CJD
tive diseases like AD show very limited responses to therapeutic
reference centers focused on differential diagnoses and reported
interventions, a clinical overlap with a huge number of highly
that neurodegenerative diseases caused 52% [9], respectively
treatable disorders and conditions underlines the need for thor-
80% [10] of non-CJD cases of RPD. Other diseases like encepha-
ough diagnosis. By reviewing the current literature and consider-
litis, ischemia, or metabolic disorders were reported to be less
ing data from more than 20 years of systematic evaluation within
frequent. Most of these studies were based on neuropathological
the National Reference Center for Prion Diseases in Germany, this
examinations. This introduced another bias because autopsy is
article will point out crucial clinical features and biomarkers (fluid
rather performed in cases of irreversible disorders. Data from the
and imaging) of a wide spectrum of disorders causing RPD. In
University of California, San Francisco [11] and the German CJD
addition, we provide an overview on the most important thera-
Surveillance (Figure 1) [12] have shown that inflammatory dis-
peutic options and focus on those disorders which have been
eases come into focus when clinical data are analyzed.
reported as the most common differential diagnoses.

CONTACT Inga Zerr ingazerr@med.uni-goettingen.de Clinical Dementia Center and National TSE Reference Center, Department of Neurology, Georg-August
University Goettingen, Robert-Koch-Str. 40, Goettingen 37075, Germany
© 2018 Informa UK Limited, trading as Taylor & Francis Group
2 I. ZERR AND P. HERMANN

Figure 1. Data from the national reference center for transmissible spongiform encephalopathies.
Diagnoses of non-CJD cases which had been presented to the NRZ-TSE as suspected prion disease. (n = 118, Hermann et al. 2018).

Based on data from the National Reference Center, the aware- diagnoses highly depends on the nature of the reporting
ness for prion diseases is very high among physicians. European center and on its geographical localization. Nevertheless,
centers reported that the number of probable or definite CJD cases potentially treatable disorders like immune-mediated or infec-
accounted for only a small part of all suspected cases [12,13]. tious encephalitis are important and are observed in a sub-
To estimate the unbiased distribution of differential diag- stantial number of cases in all studies reported.
noses and the rate of treatable conditions, studies from ter-
tiary care centers are needed. Only few of these have been
performed. An overview is given in Table 1. A recent study
3. Etiologies, diagnostic procedures, and
from Brazil [14] reported that 3.7% (n = 61) of all patients
therapeutic options
admitted to a neurological department (2012–2015; n = 1648)
showed RPD. Another study from India [15] reported that 27% 3.1. Neurodegenerative diseases
(n = 187) of all patients with dementia (2008–2016; n = 693)
3.1.1. Sporadic Creutzfeldt–Jakob disease and genetic
showed RPD. These patients were rather young, mean age was
prion diseases
48 years (Brazil), respectively 50 years (India). In all of these
Human prion diseases are caused by misfolded and potentially
cohorts, immune-mediated or infectious diseases of the CNS
transmissible proteins [16]. Given mortality and incidence rates
were the most common causes for RPD. The high rate of
differ from 1.7 (multinational investigation 1993–2002) [17] to
infectious diseases in the Indian cohort was caused by high
2.2 (Germany 2016) [12] per million/year. The most common
rates of subacute sclerosing panencephalitis and
human prion disease is sporadic CJD (90% of cases). It is
neurosyphilis.
characterized by RCD and early occurrence of additional neu-
To sum up, there is evidence that RPD is not a very rare
rological symptoms such as ataxia, cortical visual impairment,
condition. The administered distribution of differential
pyramidal or extrapyramidal signs, and myoclonus. The mean
age at onset is around 65 years and the mean disease duration
Table 1. Differential diagnoses of RPD reported by tertiary referral centers. only about 5–6 months [18]. Other prion diseases have a
Athens, Zhejiang, Sao Paulo, Chandigarh, genetic origin or are caused by transmission (iatrogenic CJD
Greece [61] China [106] Brazil [14] India [15] and variant CJD). Although being rare, CJD is considered as a
(n = 68*) (n = 310**) (n = 61) (n = 187) model disease for RPD.
Infectious 5.9* 21.9 19.7 20.6 For a definite diagnosis, neuropathological investigation is
encephalitis (%)
Immune-mediated 8.8 9.0 45.9 18.2 needed. The clinical criteria for sporadic CJD (WHO criteria)
disease (%) comprise RPD + at least two other neurological signs (cerebellar
Creutzfeldt–Jakob 13.2 7.1 11.5 7.5 or visual, pyramidal or extrapyramidal, myoclonus, akinetic mut-
disease (%)
Neurodegenerative 47.0 24.8 8.2 14.4 ism) [2]. Besides a typical clinical syndrome, at least one technical
diseases (%) investigation (MRI, CSF 14-3-3 or EEG) has to show CJD-typical
Alzheimer’s disease 17.6 14.5 n.a. n.a. results. The EEG may show a specific pattern of periodic sharp-
(%)
Others (%) 29.4 10.3 n.a. n.a. wave complexes [18]. However, this pattern has a poor sensitiv-
Vascular dementia 13.2 ** n.a. 9.6 ity, especially in early disease stages. Elevated proteins 14-3-3 in
(%) CSF are highly sensitive and specific in the differentiation of
Toxic + metabolic * 10.3 n.a. 16.0
(%) sporadic CJD from other neurodegenerative diseases. But as a
Others (%) 11.8 26.9 14.7 13.4 marker of neuronal damage, it has also been reported to be
*Acute infectious diseases and toxic–metabolic disorders had been excluded; elevated in encephalitis, ischemia, or other encephalopathies
**cerebrovascular diseases had been excluded. [9] (Table 2). The CSF real-time quaking-induced conversion
EXPERT REVIEW OF NEUROTHERAPEUTICS 3

(RT-QuIC) [19,20] is able to detect pathological prion protein respective ≥3 per 6 months [3,4]. Other authors proposed a defini-
(PrPsc) with a good sensitivity around 90% and a specificity of tion indicating rpAD when the survival time is shorter than 4 years
nearly 100% [21] and has recently been suggested to be inte- [31]. In a multicenter study, the mean survival time of rpAD
grated in amended diagnostic criteria [13,22]. To date, only few patients was only 10 months [32]. It is still a subject of discussion
false-positive cases have been reported [21,23]. Nevertheless, the whether rapid progression in AD is a matter of co-pathology (e.g.
method is expensive and not available in many regions. CJD- cerebrovascular disease, Lewy bodies) but recent studies have
typical findings on MRI include strictly cortical hyperintensities shown that intrinsic factors like a high prevalence of amyloidangio-
(‘ribboning’) and/or hyperintensities in the basal ganglia pathy [33,34] and the characteristics of amyloid β and its deposi-
(Table 3). These patterns show very good diagnostic accuracies tions differentiate rpAD from AD [35–37]. Only few data on the
[2,24]. For a deeper view on neuroimaging in CJD and its differ- incidence of rpAD in relation to AD are available but a study using
ential diagnosis, we recommend reading specific reviews, e.g. data from the Alzheimer’s Disease Neuroimaging Initiative data-
Fragoso et al. [25] or Gaudino et al. [26]. base reported that 18% of patients with ‘mild’ AD (MMSE 20–26
Genetic prion diseases can show deviant syndromes and results points) showed rapid progression in the course of the disease. In
of technical investigations. They are caused by numerous muta- this study, the diagnosis of rpAD is based on a MMSE score loss of
tions of the prion gene. Some defined syndromes are Gerstmann– ≥4 points within 6 months [38].
Sträussler–Scheinker, familial CJD, and fatal familial insomnia. The The clinical diagnosis of AD is based on the presence of
sensitivity of MRI and CSF markers is lower than in sporadic CJD. dementia (impairment of memory function and at least one
Similar to genetic CJD, variant CJD occurs in younger patients that additional cognitive domain + impaired function of activities
often present with psychiatric symptoms and dysesthesia. On MRI, of daily living) and the exclusion of other causes. Since 2011,
isolated hyperintensities of the pulvinar are frequent. For a definite the NIA-AA criteria [39] were extended and include now CSF-
diagnosis, a tonsillar biopsy can be performed. Current diagnostic and imaging biomarker. The updated criteria from 2018 [40]
criteria have been published by Heath et al. in 2010 [27]. Iatrogenic allow the AD diagnosis which is based on biomarkers alone. In
CJD can be taken into account, when a patient with CJD has this context, AD has been defined as a disorder defined by
corresponding risk factors [28]. neuropathological changes and not by a clinical syndrome.
Therapeutic options are limited to a palliative concept. Only According to various publications and recent diagnostic cri-
for doxycycline, (limited) data have shown a benefit in terms teria [40], most AD patients show a specific biomarker signa-
of a prolonged disease progression in certain subtypes when ture. This includes CSF, MRI, and PET (see Tables 2 and 3). In
treated in early stages [29]. patients with rpAD, the same signature could be observed
[30,38] but CSF markers of neuronal damage like tau (respec-
3.1.2. (rp)AD tive tau in relation to p-tau) and proteins 14-3-3 [30] showed
AD is the most common cause for dementia. Cerebral depositions higher levels than in classical AD forms. In addition, a pro-
of amyloid β and hyperphosphorylated tau protein (p-tau) play a nounced hypometabolism displayed by FDG-PET in left angu-
central role in disease pathogenesis. Clinically, most patients show lar and left temporal cortices was predictive for rpAD [38].
a slow decline of cognitive functions over many years. Within the Like in all neurodegenerative diseases, no causal therapy
last 10 years, a distinct subtype of the disease called rpAD has been has been established, yet. Nonetheless, a huge amount of
described. Some clinical features have been observed which differ- different drugs and treatment strategies is currently under
entiate typical AD from rpAD. Despite RCD, rpAD patients often investigation (e.g. γ-secretase inhibitors, beta-site amyloid per-
show early occurrence of myoclonus, disturbed gait, and rigidity as cursor protein cleaving enzyme [BACE] inhibitors, and immu-
well as aphasia and hallucinations [30]. This results in a relevant notherapies) [41]. Current treatment guidelines recommend a
clinical overlap between rpAD, CJD, and other RPD. In this context, symptomatic medication with acetylcholinesterase inhibitors
RCD has been defined as a loss of ≥6 MMSE points per year, or memantine or a combination of both [42]. As long as no

Table 2. CSF abnormalities in different causes of RPD.


Oligoclonal CSF markers of neuronal damage or
Etiology Cell count bands other surrogates Specific CSF markers
Prion diseases ↔ Not present 14-3-3: ↑ – ↑↑↑ RT-QuIC: positive
Tau: ↑ – ↑↑↑
(rp)AD ↔ Not present 14-3-3: ↔ – ↑ p-Tau: ↔ – ↑
Tau: ↔ – ↑↑ aβ 1–42: ↔ – ↓
aβ ratio: ↔ – ↓
Other neurodegenerative ↔ Not present 14-3-3: ↔ – (↑) Not available
diseases Tau: ↔ – (↑) Candidate: α-synuclein↓ in synucleinopathies
Cerebrovascular disease ↔ Not present 14-3-3: ↔ – (↑↑)* Not available
(except for (except for Tau: ↔ – (↑)* Candidates: albumin ratio, neurofilament light chain,
vasculitis) vasculitis) matrix metalloproteinases
Immune-mediated ↔ – ↑↑ Present in most 14-3-3: ↔ – ↑↑↑ Various known antibodies
encephalitis cases Tau: ↔ – ↑↑ (see Table 4)
Infectious encephalitis ↑ – ↑↑↑ Present 14-3-3: ↔ – ↑↑↑ Specific PCR
Tau: ↔ – ↑↑ Specific antibodies
CSF culture
*14-3-3 and tau can be elevated after an acute ischemic event.
4 I. ZERR AND P. HERMANN

data on a specific treatment for rpAD become available, same memantine has a positive effect on clinical global impression
treatment strategies as for classical forms should be applied. [53]. Parkinsonism in DLB can be treated with dopamine ago-
nists but a special concern is the occurrence of hallucinations
and other psychotic syndrome. Typical neuroleptic drugs can
3.1.3. DLB lead to severe akinetic and vigilance crises [54]. If necessary,
Still being a subject of controversial discussion, there is some the use of safer and still effective neuroleptic drugs like cloza-
evidence from autopsy-based studies that DLB might be the sec- pine and quetiapine [55] is highly recommended.
ond most common cause for dementia (26% [43]) after AD.
According to recent criteria [44], clinical core features include
dementia, extrapyramidal signs, rapid eye movement (REM) sleep 3.1.4. Other neurodegenerative diseases
behavior disorder, visual hallucination, and fluctuating decline of DLB, Parkinson’s disease dementia (PDD), and multiple system
cognition. These fluctuations can mimic rapid disease progression atrophy (MSA) including its subtypes are the three main types of
and the presence of neurological signs implicates a huge overlap α-synucleinopathy associated with dementia [56]. PDD is usually
of up to 40% with clinical WHO criteria for possible CJD [45]. A slowly progressive and shares many characteristics with DLB
study including data from 807 DLB patients observed a mean despite the retarded occurrence of dementia in the clinical
disease duration of 8.8 years [46]. However, a subset of DLB course [44]. Because of its rather fast progressing neurological
patients shows primary RPD [5] (rpDLB). Despite some marked multisystemic symptoms, MSA is a relevant differential diagnosis
DLB features, no specific neuropathological changes could be for CJD and other encephalopathies. A nationwide study from
identified in rpDLB [47]. Iceland reported an incidence of 7 per million person-years, a
Specific findings in EEG and neuroimaging are included in the mean onset of 65 years, and a rather fast progression displayed
diagnostic criteria for DLB [44] (Table 3). Fluid markers are not by a mean survival time of 5.7 years [57]. Although dementia is a
part of the diagnostic routine (Table 2) but a decreased concen- feature that makes MSA unlikely [58], recent studies have shown
tration of α-synuclein in CSF has been reported [48] and there are that subcortical cognitive deficits are frequent [59]. Diagnostic
ongoing efforts to develop a method to detect α-synuclein mis- criteria comprise specific clinical and imaging features [58]
folded aggregates in patients with Parkinsonism [49,50]. (Table 3). Validated fluid markers are not available [60].
Although a Cochrane review in 2012 could not find a clear Tauopathies have been reported to account for up to 16.2% of
benefit from therapies with acetylcholinesterase inhibitors RPD cases [61] in single centers. Various clinical syndromes have
[51], donepezil has shown a positive effect on cognition and been described including behavioral variant frontotemporal
behavioral symptoms [52]. Some data indicate that also dementia (bvFTD), semantic dementia, primary progressive

Table 3. Imaging abnormalities in different causes of RPD.


Imaging
Etiology technique Findings
CJD MRI: HI in gray matter areas (cortical and/or nucleus caudatus and putamen ± HI of the pulvinar and dorsomedial
DWI > FLAIR/ thalamus); corresponding low signal on ADC maps; no white matter involvement and no enhancement of
T2 gadolinium
FDG-PET Hypometabolism in various regions, in some prion diseases (fatal familial insomnia) dominantly in the thalamus
AD MRI: T1 Atrophy of the medial temporal lobe
FDG-PET Hypometabolism of temporal and parietal lobes
Amyloid-PET Amyloid depositions
Other MRI Synucleinopathies: Unspecific atrophy (DLB); atrophy of putamen, middle cerebellar peduncle, pons or cerebellum
neurodegenerative (MSA)
diseases Tauopathies: Atrophy in anterior cingulate, frontoinsula, striatum and amygdala (bvFTD), predominant midbrain
atrophy (PSP); atrophy of dorsal prefrontal and perirolandic cortex, striatum, and brainstem (CBD)
FDG-PET Synucleinopathies: Occipital hypometabolism ± cingulate island sign (DLB), hypometabolism in putamen, brainstem,
or cerebellum (MSA)
Tauopathies: Hypometabolism in frontal lobes, anterior temporal cortex, and anterior cingulate cortex (FTD);
predominant midbrain hypometabolism (PSP)
PET/SPECT DLB: Reduced presynaptic dopamine transporter uptake in basal ganglia
PSP: Postsynaptic striatal dopaminergic degeneration
Myocardial DLB: Low 123iodine MBIG uptake (also reported in MSA)
scintigraphy
Cerebrovascular MRI VD: Chronic territorial infarctions, subcortical HI (FLAIR or T2), lacunes, microbleeds
diseases Acute event (infarction): DWI-HI and low ADC signal of the gray and the white matter corresponding to vessel
territories
CNS vasculitis: Infarction, subcortical HI (FLAIR or T2), leptomeningeal or parenchymal gadolinium enhancement,
microbleeds, mass bleeding
Angiography CNS vasculitis: Typical arterial segmentations
Encephalitis MRI Immune-mediated: Edema resp. T2-HI of cortical regions or basal ganglia (e.g. paraneoplastic limbic encephalitis) or of
the white matter (e.g. SREAT); sometimes gadolinium enhancement of lesions
Infectious: Edema resp. T2-HI of cortical regions (e.g. temporal in HSV-encephalitis) or basal ganglia (e.g. HIV-
encephalopathy) or of the white matter (e.g. PML); abundant gadolinium enhancement of lesions
DWI: Diffusion weighted Image; FLAIR: fluid-attenuated inversion recovery; HI: hyperintensities; FDG: fluorodeoxy-glucose; SPECT: single-photon emission computed
tomography; MBIG: metaiodobenzylguanidine; CJD: Creutzfeldt–Jakob disease; AD: Alzheimer’s disease; DLB: dementia with Lewy bodies; MSA: multisystem atrophy;
FTD: frontotemporal dementia; PSP: progressive supranuclear palsy; CBD: corticobasal degeneration; VD: vascular dementia; bvFTD: behavioral variant frontotem-
poral dementia; SREAT: steroid-responsive encephalopathy associated with autoimmune thyroiditis; PML: progressive multifocal leukoencephalopathy.
EXPERT REVIEW OF NEUROTHERAPEUTICS 5

Table 4. Inflammatory (infectious and immune-mediated) disorders causing RPD.


Immune-mediated encephalitis*
Paraneoplastic encephalopathies
Antigen Syndrome Associated neoplasia
Hu Diverse SCLC
Yo PCD Breast, gynecologic cancer
Ri Diverse Breast cancer
Ma1 Diverse Diverse
Ma2 Diverse Germ cell tumors
Tr PCD Hodgkin’s lymphoma
Zinc finger protein 4 PCD SCLC
P/Q-type VGCC LEMS, cerebellitis SCLC
Amphiphysin Encephalitis, stiff-person syndromeSCLC, breast cancer
Antibody-associated encephalopathies, possibly associated with neoplasia
Antigen Syndrome Associated neoplasia
NMDAR Encephalitis Ovarian teratoma
LGI 1 Limbic encephalitis
CASPR2 Limbic encephalitis Thymoma (rare)
AMPAR Limbic encephalitis SCLC, thymoma, breast cancer
GABAAR Encephalitis, stiff-person syndrome Thymoma
GABABR Limbic encephalitis SCLC
DPPX Encephalitis, stiff-person syndrome B-cell neoplasia
mGluR5 Encephalitis Hodgkin’s lymphoma
D2R Basal ganglia encephalitis
GlyR Encephalitis, stiff-person syndrome Thymoma, lymphoma (rare)
GAD Stiff-person syndrome, cerebellitis Neuroendocrine (rare)
Others
SREAT
Gluten encephalopathy (celiac disease)
Multiple sclerosis and ADEM
Neurosarcoidosis
Lupus cerebritis
NBD
Infectious agents (other than prions) possibly causing RPD**
Viral Bacterial
HIV Treponema pallidum (Lues)
JC-virus (progressive multifocal leukoencephalopathy) Tropheryma whipplei (Whipple’s disease)
Herpes simplex virus 1, 2 (6, 7) Mycobacterium tuberculosis
Varicella zoster virus Listeria monocytogenes
Measles (sclerosing subacute panencephalitis) Borrelia burgdorferi
Mumps Parasitic/Fungal
Epstein–Barr virus Toxoplasma gondii
Influenza virus Balmuthia
Cytomegalovirus Cryptococcus neoformans
Japanese encephalitis virus Aspergillosis
West Nile virus
Rabies
Parvovirus B19
Hepatitis C
*Based on Lancaster and Dalmau [88], Dalmau and Graus [89], and clinical experience of the German CJD Surveillance group.
**Based on Granerod et al. [87] and clinical experience of the German CJD Surveillance group.
VGCC: Voltage-gated calcium channel; SREAT: steroid-responsive encephalopathy associated with autoimmune thyroiditis; ADEM: acute disseminated encephalo-
myelitis; NBD: neuro-Behcet’s disease; PCD: paraneoplastic cerebellar degeneration; SCLC: small cell lung cancer; NMDAR: n-methyl-D-aspartate receptor; LGI
1: leucine-rich glioma inactivated 1; CASPR2: contactin-associated protein 2; AMPAR: alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor;
GABA: gamma-aminobutyric acid; DPPX: dipeptidyl-peptidase-like protein 6; mGluR5: metabotropic glutamate receptor 5; D2R: dopamine receptor D2;
GlyR: glycin receptor; GAD: glutamic acid decarboxylase.

aphasia (PPA), corticobasal syndrome, and progressive supranuc- often suffer from severe cardiac disorders or seizures which
lear palsy (PSP). Many of these syndromes can be caused by can contribute to RPD. Cerebrovascular disease is also known
different types of tauopathy and some of them are hereditary. as a frequent cofactor in neurodegenerative dementias like AD
They can also be classified by biochemical features which show a and DLB. In a community-based clinical–pathologic cohort
huge clinical overlap [62–64]. Some entities show distinct radiolo- study, 38% all AD patients had infarcts [67]. Criteria combining
gical features [65,66] (Table 3) but fluid markers have not been clinical and radiological features of VD and AD (respectively
established. AD and DLB) have been published in 2014 [68]. The term VD
comprises several conditions of different etiology [69], com-
monly used NINDS-AIREN criteria [70] are outdated, a neuro-
3.2. Cerebrovascular disease pathological gold standard is missing [71], and accurate fluid
3.2.1. Vascular dementia markers are still not available. A consensus study is currently
Vascular dementia (VD) is considered as a slowly or stepwise proposing a new classification, which involves the differentia-
progressing disorder. On the other hand, patients with VD tion of mild and major forms of vascular cognitive impairment
6 I. ZERR AND P. HERMANN

and endorses the Neurological Disorders – Canadian Stroke syndromes are associated with classic paraneoplastic syndromes
Network neuropsychological assessment protocols and recom- or with antibodies against neuronal cell-surface or synaptic pro-
mendations for imaging [72]. teins [88,89]. Frequent tumors are lung cancer (50% of patients
To date, the diagnosis of VD relies on the presence of dementia with paraneoplastic limbic encephalitis), gynecological tumors,
and evidence for cerebrovascular disease on MRI or CT (Table 3). breast cancer, and lymphoma [90]. The relevant antibodies are
Some studies have reported an elevated albumin CSF/serum ratio listed in Table 4. Nonetheless, in 7% of 163 cases with limbic
in VD compared to other dementias [73–75]. Neurofilament light encephalitis that were analyzed in 2018 in a specialized center,
[76,77] and matrix metalloproteinases [78] have also been identi- no antibodies could be found [91]. Some diseases are not asso-
fied as biomarker candidates. However, none of these markers has ciated with neoplasia or known anti-neuronal antibodies, e.g.
been validated sufficiently [79]. Therapeutic options in VD are still steroid-responsive encephalopathy associated with autoimmune
limited to the treatment of risk factors and symptoms like depres- thyroiditis (SREAT, formerly named Hashimoto’s encephalopathy)
sion and seizures. However, a meta-analysis from 2007 has shown which can present with chronic dementia [92]. MRI alterations
that donepezil had a positive effect on cognition scales and activ- (Table 3) and clinical presentations are heterogeneous. Cognitive
ities of daily living (less than in AD) [80]. disturbance, psychiatric symptoms, seizures, and ataxia are com-
mon symptoms indicating a huge overlap with other RPD.
3.2.2. Acute vascular events CSF analytics usually show slightly or moderately elevated
Data from the German CJD surveillance center indicate that more cell count (lymphocytes) or oligoclonal bands or both
than 20% of non-CJD cases had an acute cerebrovascular vascular (Table 2). These are clearly pathologic findings that are not
event (Figure 1) [12]. Especially recurring cardiac embolism or associated with CJD or other neurodegenerative diseases.
secondary seizures and venous thromboses can mimic RPD and Unfortunately, these routine markers remain normal in many
markers of neuronal destruction like CSF tau and 14-3-3 can be patients. For example, 20% of 100 patients in a study on anti-
elevated (Table 2). Despite the aforementioned disorders, the n-methyl D-aspartate receptor (NMDAR) encephalitis did not
posterior reversible encephalopathy syndrome is an important show elevated leucocytes or oligoclonal bands [93]. Therefore,
differential diagnosis. An altered mental status is one of key clinical an extended search for specific antibodies (Table 4) in the CSF
features [81] and may mimic RPD. Some other clinical symptoms of patients with RPD and no other suggestive diagnosis is
like cortical visual disturbance, seizures, and even MRI abnormal- crucial. The presence of antibodies in blood is less sensitive
ities can be misdiagnosed as CJD, encephalitis, or stroke [82]. For a [94]. A search for neoplasia and even a probative therapy with
diagnosis of acute vascular diseases, the clinical history and MRI (at steroids or intravenous immunoglobulin is a therapeutic
best) or CT (sufficient only for detection of hemorrhage) including option. Plasmapheresis and other immune-modulatory or
angiography are crucial (Table 3). immune-suppressive therapeutics can also be effective. In
case of a paraneoplastic syndrome, the identification and
3.2.3. CNS vasculitis treatment of the tumor might lead to the improvement of
Numerous disorders are able to cause secondary CNS vasculitis. then neurological syndrome.
These include infections of all kind and other systemic inflam- In the following, we describe anti-NMDAR encephalitis as a
matory diseases (e.g. Wegener’s granulomatosis, Behçet’s dis- prototype of the autoimmune mediated encephalitis. After a
ease, giant-cell arteritis, etc.) [83]. Although many case reports prodromal phase with influenza-like symptoms, patients show
on primary CNS vasculitis can be found, systematic observa- psychotic symptoms, impaired episodic memory, and seizures
tions are rare. A study from the Mayo Clinic including 163 cases of temporal origin [95]. The disease is associated with neopla-
over a 29-year period estimated an incidence of 2.4 per million sia in up to 80% (teratoma in up to 60%) [96]. CSF analyses are
person-years [83,84]. According to a review in 2012 [85], up to obligate as NMDA receptor antibodies often cannot be found
50% of patients with CNS vasculitis showed cognitive altera- in peripheral fluids [94]. Elevation of CSF leucocytes at early
tions. Other frequent symptoms were headaches and various stages and decrease in the further course as well as evidence
neurological signs. Diagnostic criteria have been proposed in for intrathecal IgG synthesis are characteristics [97]. FLAIR- or
1988 [86] defining meningeal or brain biopsy as gold standard. T2-weighted MRI may show abnormal signal hyperintensities
Advances in MRI technology and angiography allow early diag- in the temporal cortex in 50% of the cases [95]. Besides tumor
nosis (see Table 3). Although being considered as a clinical gold treatment, the therapy should consist of short-term high-dose
standard, the cerebral angiography shows varying sensitivities steroid application and at least one additional immunologic
between 40% and 90% [83]. No specific CSF markers were therapy, e.g. plasmapheresis or intravenous immunglobulin
available but 80–90% of the cases showed mildly increased (IVIG) [97]. A good outcome has been reported in up to 75%
leucocyte count or total protein concentration [83]. A treatment sufficiently treated patients [95]. Patients without an asso-
with steroids alone or in combination with cyclophosphamide ciated tumor are less likely to respond to therapy. In these
showed a response rate of 80–85% [84]. cases, a second-line therapy (rituximab or cyclophosphamide)
can be effective in up to 65% [98].
3.3. Inflammatory CNS diseases
3.3.2. Infectious encephalitis
3.3.1. Immune-mediated encephalitis Encephalitis can be associated with or directly caused by
According to a population-based study, 21% of patients with numerous viral, bacterial, fungal, and parasitic organisms.
encephalitis had acute immune-mediated encephalitis, 37% had The most common causes are herpes simplex virus and herpes
unknown causes [87]. Most immune-mediated encephalopathic zoster virus [87]. Usually, infectious CNS diseases present with
EXPERT REVIEW OF NEUROTHERAPEUTICS 7

an acute onset but a detailed preclinical history is not always suggest that a search for gynecological, abdominal, and thoracic
available. Besides, slowly progressing encephalitis with cogni- tumors should be performed in all cases of unclarified RPD.
tive symptoms can present as a RPD. In many cases, patients
with infectious encephalitis show systemic signs of inflamma- 3.4.2. Mitochondrial disorders and other genetic causes of
tion (e.g. fever, altered peripheral markers of inflammation). RPD
An elevated cell count in the CSF is suggestive for encephalitis Most mitochondrial disorders have an onset in childhood but
(Table 2). MRI findings are heterogeneous depending on the adult forms have to be taken into account when patients
underlying infectious agent but MRI lesions which enhance present with RPD, especially in cases of unexplained encepha-
gadolinium should always throw suspicion on infectious lopathies. The prevalence of pathogenic mutations seems to
causes (Table 3). In recent years, some atypical (non-herpes) be rather high (up to 11/100,000) [105]. Especially in diseases
viral encephalopathies like HIV-associated cognitive impair- like mitochondrial encephalomyopathy with lactic acidosis
ment have become more and more important [99]. Although and stroke-like episodes, but also in several forms of myoclo-
being rare, physicians should also be aware of CNS Whipple’s nus epilepsy, dementia can be one of the primary clinical
disease which is highly treatable. Neurologic symptoms and features [105]. Mitochondrial disorders, storage diseases, and
MRI lesions are various and may mimic other forms of RPD but other rare genetic causes for RPD in adults are listed in
CSF PCR is a specific diagnostic tool [100]. Table 4 gives a Table 5.
summary of common and unusual infectious agents that are
able to cause RPD. 3.4.3. Common clinical conditions mimicking RPD
Some conditions like electrolyte disturbances (e.g. hypona-
tremia), endocrine disorders (e.g. hypothyroidism), intoxica-
3.4. Other etiologies of RPD and conditions mimicking tions, or psychiatric disorders are very frequent but can
RPD cause or mimic encephalopathy and dementia (see
Table 5). Those conditions should always be considered in
Regarding experience from the National Reference Center for
a standard diagnostic workup.
Prion Diseases in Germany and the literature [12,101–103],
Table 5 gives an overview of numerous diseases and condi-
tions that may show or might mimic RPD.
3.4.4. Concomitant pathologies and comorbidities
The concept mixed dementia is well known, especially in
AD and VD [67,68], and concomitant pathologies (mostly
3.4.1. CNS neoplasia AD) have been found in 73% of autopsy cases with RPD
Neoplasm of the CNS has been reported to be an important [10]. A synergistic effect of different pathologies in one
differential diagnosis for sporadic CJD[11,12]. Especially, the pri- individual has to be considered in cases of PRD, but in
mary lymphoma of the CNS is known for causing cognitive symp- general, it remains uncertain in which way and to what
toms [104] and its ability to mimic encephalitis or several other extend these pathologies contribute to the speed of cog-
causes of RPD. But also solid or diffuse glioma or leptomeningeal nitive decline. Furthermore, physicians should keep in
spread of peripheral tumors can cause cortical symptoms like RPD. mind that comorbidities like infections, cardiac or meta-
For a diagnosis, MRI (including gadolinium application) and lumbar bolic diseases as well as secondary symptoms, and com-
puncture (to search for atypical cells) are crucial. Nevertheless, we plications of initially slowly progressive dementia (e.g.

Table 5. Summary of various other diseases and conditions that may lead to or mimic RPD.
Other diseases and conditions that may lead to or mimic RPD
CNS neoplasia Storage diseases and other genetic causes for RPD Toxic
1. CNS lymphoma 1. Wilson’s disease 1. Alcohol-related dementia
2. Solid neoplasia (primary in the CNS or metastatic) 2. NBIA 2. Benzodiazepine-related dementia
3. Diffuse gliomatosis 3. Lysosomal storage diseases (selected): 3. Methyl intoxication
4. Leptomeningeal spread of other tumors mucopolysaccharidoses, sphingolipidoses, and neuronal 4. Metal intoxications (e.g. lithium,
Metabolic ceroid lipofuscinoses mercury)
1. Hyponatremia and other electrolyte disturbances 4. Huntington’s disease 5. Side effects of chemotherapeutics or
2. Wernicke’s encephalopathy (thiamine) and other 5. CADASIL resp. CARASIL neuroleptic drugs
vitamin deficiency syndromes (e.g. B12, folate, 6. Mitochondrial diseases (selected): MELAS, Leigh Psychiatric
niacin, biotin) disease 1. Severe depression
3. Hepatic encephalopathy 7. PME: Lafora disease, ULD 2. Schizophrenic disorders
4. Uremic encephalopathy 8. Porphyria 3. Hypochondriac delusions
5. Hypothyroidism 9. Methylmalonic academia Others and secondary conditions
6. Hypoparathyroidism 10. DRPLA 1. Hypoxic brain damage
7. Porphyria 2. Epilepsy and secondary seizures
8. Adrenal insufficiency 3. Systemic inflammation (e.g. systemic
9. Extrapontine myelinolysis immune-mediated diseases, septic
encephalopathy)
4. NPH
NBIA: Neurodegeneration with brain iron accumulation; CADASIL: cerebral autosomal dominant arteriopathy with subcortical Infarcts and leukoencephalopathy;
MELAS: mitochondrial encephalopathy, lactic acidosis and stroke-like episodes; PME: progressive myoclonic epilepsies; ULD: Unverricht–Lundborg disease;
DRPLA: dentatorubral-pallidolysial atrophy; NPH: normal pressure hydrocephalus.
8 I. ZERR AND P. HERMANN

seizures, infections) might also accelerate disease progres- to rely on criteria with limited diagnostic accuracies. Another
sions and shorten life expectancies. problem emerges from the mistaken idea that dementia is almost
always caused by neurodegenerative disorders. This may easily
lead to a restriction of diagnostic and therapeutic measurements in
4. Conclusion the clinical case management.
Dementia is a syndrome of major importance for public health For an improvement of the current situation, three major
care and RPD is not a rare phenomenon. Although CJD sur- goals have to be focused:
veillance centers have reported that neurodegenerative dis-
eases are the most frequent differential diagnoses, data from ● The awareness of different etiologies of RPD has to be
tertiary care centers show that highly treatable disorders like raised among clinicians.
encephalitis are the most common causes for RPD. Effective ● More reliable noninvasive or minimal-invasive clinical
antiviral and antibacterial therapeutics for many infectious markers have to be identified and validated.
CNS diseases and immunosuppressive medications for the ● New therapeutic strategies for neurodegenerative dis-
treatment of immune-mediated encephalitis are available. eases have to be developed and established.
Still, the differential diagnosis of RPD is not trivial which
indicates the need for a thorough diagnostic workup. This Many causes of RPD are highly treatable and we identified some
should include MRI and CSF analyses. If these standard proce- studies providing evidence that these conditions are very frequent
dures show no suggestive or inconclusive diagnostic results, among cases with RPD [14,15,106]. An effective treatment of
specific technical measurements or genetic analyses will be potentially reversible RPD (e.g. infectious and immune-mediated
indicated. Even probationary therapeutic attempts can be encephalitis) has to start as early as possible in the course of the
considered in some cases. Figure 2 provides an overview. disease. Therefore, it is crucial to identify those cases at an early
stage. When effective drugs for neurodegenerative diseases
become available, the starting point of the therapy may be even
5. Expert commentary more important. Recent experimental therapeutic strategies in
RPD can be caused by numerous pathologies which require dis- Alzheimer’s disease, α-synucleinopathies, and Creutzfeldt–Jakob
similar therapeutic strategies. Unfortunately, clinicians often have disease have been targeting certain proteins in order to avoid

Figure 2. Synopsis of diagnostic measurements in cases with RPD.


FLAIR: Fluid-Attenuated Inversion Recovery, DWI: Diffusion Weighted Image, SWI: Susceptibility Weighted Image, FDG: Fluorodeoxy-glucose, PET: Positron Emission Tomography, SPECT:
Single-Photon Emission Computed Tomography, CJD: Creutzfeldt-Jakob Diseasae, AD: Alzheimer’s Disease
EXPERT REVIEW OF NEUROTHERAPEUTICS 9

conformation changes and aggregation or to remove existing Key issues


aggregations. In this context, one has to keep in mind that patho-
● Dementia is a syndrome of major importance for public
logical changes in the nervous system precede the point of the
health care and RPD is not a rare phenomenon in patients
clinical onset of these diseases. It is obvious that a substantial
referred to neurological units. Prion reference centers have
clinical benefit can only be achieved when the destruction of the
reported high rates of CJD and other neurodegenerative
neuronal integrity is not too progressed.
diseases in patients with RPD. In contrast, tertiary care
In our opinion, a faster and more accurate clinical diagnosis
centers have reported that highly treatable disorders like
will achieve more than an improved treatment of RPD. It may
encephalitis are the most common causes for RPD.
even be a presupposition for a progress in understanding
● The clinical differential diagnosis of RPD is not trivial and
underlying pathologies and developing new therapeutics in
the number of possible causes is extremely high including
the whole field of neurodegenerative disorders. Without
all kinds of neurodegenerative diseases, immune-mediated
doubt, enhanced diagnostics are necessary to evaluate the
and infectious diseases, metabolic and toxic disorders, and
effectivity and safety of new drugs in clinical trials.
genetic diseases.
Concerning clinical diagnostics, it will be important to identify
● This indicates the need for a thorough clinical diagnostic work
disease-specific markers that are not only surrogates representing
up. This has to include a detailed assessment of the clinical
general neuronal damage. In Alzheimer’s disease, the amyloid-PET
presentation and the patient’s history but also neuroimaging,
can already be performed in many centers and updated research
fluid markers (CSF and blood) and sometimes genetic analyses.
criteria [40] are based on specific biomarkers which allow diagnosis
● Because of the high prevalence of immune-mediated ence-
in early or even preclinical stages in some cases. In Creutzfeldt–
phalitis among RPD, a search for neoplasia as well as ence-
Jakob disease, the development and validation of the CSF RT-QuIC
phalitis-related antibodies should be performed. In
method has been a major improvement. For the first time, patho-
suggestive cases without evidence, a probationary treat-
logic prions (PrP scrapie) can be detected without biopsy or neu-
ment with steroids can be useful.
ropathological examination. Ongoing efforts will most likely lead
to tests that can be applied to other biomaterials (e.g. blood or
mucosa [22]). Several proteins behaving like prions have been Funding
identified in recent years and the development of aggregation
This paper was supported by the German Federal Ministry of Health [Grant
assays for such proteins in tauopathies and α-synucleinopathies no: 1369-341] via the Robert-Koch Institute.
is one of the most interesting goals in current biomarker research.
Furthermore, the number of identified antibodies that are asso-
ciated with paraneoplastic or non-paraneoplastic immune- Declaration of interest
mediated encephalitis is still increasing.
I Zerr and P Hermann are employed at the German National Reference Center
In general, the aforementioned methods have to be vali- for TSE which receives grants from the Robert-Koch Institute to maintain CJD
dated in large cohorts. The technical procedures have to be surveillance and research. The authors have no other relevant affiliations or
improved and standardized to become accessible and afford- financial involvement with any organization or entity with a financial interest in
able for small centers all over the world. or financial conflict with the subject matter or materials discussed in the
manuscript apart from those disclosed.

6. Five-year view Reviewer disclosures


Hopefully, effective treatment options for AD and maybe other Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
neurodegenerative diseases will be available in 5 years.
Especially in rapidly progressive forms of these diseases, a
fast and accurate differential diagnoses will be crucial for the References
appropriate application of new therapeutics. We expect a
constant progress in the field of biomarker-based differential Papers of special note have been highlighted as either of interest (•) or of
diagnosis. Research criteria for a biological definition of AD considerable interest (••) to readers.
1. Geschwind MD, Haman A, Miller BL. Rapidly progressive dementia.
have already been published allowing a diagnosis in early or Neurol Clin. 2007;25:783–807.
even preclinical stages [40]. Reliable biomarkers for neurode- •• Commonly used definition of rapidly progressive dementia.
generative diseases like DLB and CJD are available and will be 2. Zerr I, Kallenberg K, Summers DM, et al. Updated clinical diagnostic
developed for VD, soon. Furthermore, new antibodies related criteria for sporadic Creutzfeldt-Jakob disease. Brain.
2009;132:2659–2668.
to certain immune-mediated diseases and new mutations
•• Clinical criteria for the diagnosis of Creutzfeldt–Jakob disease.
related to dementia syndromes will be discovered. Panel diag- 3. Soto ME, Andrieu S, Arbus C, et al. Rapid cognitive decline in
nostic for genetic analyses and the search for antibodies will Alzheimer’s disease. Consensus paper. J Nutr Health Aging.
become more accessible and achievable. If the awareness for 2008;12(10):703–713.
different types of RPD rises among physicians, these tools will • Definition and description of rapidly progressive Alzheimer’s
disease.
set the stage for the improvement of differential diagnosis,
4. Schmidt C, Wolff M, Weitz M, et al. Rapidly progressive Alzheimer
therapy, and trial monitoring. Furthermore, it is an essential disease. Arch Neurol. 2011;68(9):1124–1130.
step to gain more insight into pathophysiological 5. Gaig C, Valldeoriola F, Gelpi E, et al. Rapidly progressive diffuse
mechanisms. Lewy body disease. Mov Disord. 2011;26(7):1316–1323.
10 I. ZERR AND P. HERMANN

• Description of rapidly progressive Lewy body disease. 27. Heath CA, Cooper SA, Murray K, et al. Validation of diagnostic
6. Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative criteria for variant Creutzfeldt-Jakob disease. Ann Neurol.
diseases. Nat Reviews Neurosci. 2010;11(3):155–159. 2010;67:761–770.
7. Poser S, Mollenhauer B, Kraubeta A, et al. How to improve the 28. WHO. Global Surveillance, diagnosis, and Therapy of Human
clinical diagnosis of Creutzfeldt-Jakob disease. Brain. 1999;122 Transmissible spongiform Encephalopathies: Report of WHO con-
(12):2345–2351. sultation, February 9–11, 1998. Geneva, Switzerland.
8. Geschwind MD, Shu H, Haman A, et al. Rapidly progressive demen- 29. Varges D, Manthey H, Heinemann U, et al. Doxycycline in early CJD:
tia. Ann Neurol. 2008;64(1):97–108. a double-blinded randomised phase II and observational study. J
9. Chitravas N, Jung RS, Kofskey DM, et al. Treatable neurological Neurol Neurosurg Psychiatry. 2017;88(2):119–125.
disorders misdiagnosed as Creutzfeldt-Jakob disease. Ann Neurol. 30. Schmidt C, Redyk K, Meissner B, et al. Clinical features of rapidly
2011;70(3):437–444. progressive Alzheimer’s disease. Dement Geriatr Cogn Disord.
10. Grau-Rivera O, Gelpi E, Nos C, et al. Clinicopathological correlations 2010;29:371–378.
and concomitant pathologies in rapidly progressive dementia: a 31. Josephs KA, Ahlskog JE, Parisi JE, et al. Rapidly progressive neuro-
brain bank series. Neurodegener Dis. 2015;15(6):350–360. degenerative dementias. Arch Neurol. 2009;66:201–207.
11. Geschwind MD. Rapidly progressive dementia. Continuum 32. Schmidt C, Haïk S, Satoh K, et al. Rapidly progressive Alzheimer’s
(Minneap Minn). 2016;22(2):510–537. disease: a multicenter update. J Alzheimers Dis. 2012;30(4):751–
12. Hermann P, Laux M, Glatzel M, et al. Validation and utilization of 756.
amended diagnostic criteria in Creutzfeldt-Jakob disease surveil- 33. Abu-Rumeileh S, Capellari S, Parchi P. Rapidly progressive
lance. Neurology. 2018;91(4):e331–e338. Alzheimer’s disease: contributions to clinical-pathological definition
13. Stoeck K, Sanchez-Juan P, Gawinecka J, et al. Cerebrospinal fluid and diagnosis. J Alzheimers Dis. 2018;63(3):887–897.
biomarker supported diagnosis of Creutzfeldt-Jakob disease and 34. Hecht M, Krämer LM, von Arnim CAF, et al. Capillary cerebral
rapid dementias: a longitudinal multicentre study over 10 years. amyloid angiopathy in Alzheimer’s disease: association with allo-
Brain. 2012;135(10):3051–3061. cortical/hippocampal microinfarcts and cognitive decline. Acta
• Recent data on epidemiology and differential diagnoses of Neuropathol. 2018;135(5):681–694.
Creutzfeldt–Jakob disease. 35. Cohen ML, Kim C, Haldiman T, et al. Rapidly progressive
14. Studart Neto A, Soares Neto HR, Simabukuro MM, et al. Rapidly Alzheimer’s disease features distinct structures of amyloid-β.
progressive dementia: prevalence and causes in a neurologic unit Brain. 2015;138(4):1009–1022.
of a tertiary hospital in Brazil. Alzheimer Dis Assoc Disord. 2017;31 36. Qiang W, Yau WM, Lu JX, et al. Structural variation in amyloid-β
(3):239–243. fibrils from Alzheimer’s disease clinical subtypes. Nature. 2017;541
• One of four reports on rapidly progressive dementia from (7636):217–221.
tertiary hospitals. 37. Drummond E, Nayak S, Faustin A, et al. Proteomic differences in
15. Anuja P, Venugopalan V, Darakhshan N, et al. Rapidly progressive amyloid plaques in rapidly progressive and sporadic Alzheimer’s
dementia: an eight year (2008–2016) retrospective study. PLoS disease. Acta Neuropathol. 2017;133(6):933–954.
One. 2018 cited 2018 Jul 5;13. DOI:10.1371/journal.pone.0189832. 38. Ba M, Li X, Ng KP, et al. The prevalence and biomarkers’ character-
• One of four reports on rapidly progressive dementia from istic of rapidly progressive Alzheimer’s disease from the Alzheimer’s
tertiary hospitals. disease neuroimaging initiative database. Alzheimers Dement.
16. Prusiner SB. Novel proteinaceous infectious particles cause scrapie. 2017;3(1):107–113.
Science. 1982;216(4542):136–144. 39. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of
17. Ladogana A, Puopolo M, Croes EA, et al. Mortality from Creutzfeldt- dementia due to Alzheimer’s disease: recommendations from the
Jakob disease and related disorders in Europe, Australia, and national institute on aging-Alzheimer’s association workgroups on
Canada. Neurology. 2005;64:1586–1591. diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement.
18. Collins SJ, Sanchez-Juan P, Masters CL, et al. Determinants of 2011;7:263–269.
diagnostic investigation sensitivities across the clinical spectrum 40. Jack CR Jr, Bennett DA, Blennow K, et al. NIA-AA research frame-
of sporadic Creutzfeldt-Jakob-disease. Brain. 2006;129:2278–2287. work: toward a biological definition of Alzheimer’s disease.
19. Atarashi R, Satoh K, Sano K, et al. Ultrasensitive human prion Alzheimers Dement. 2018 Apr;14(4):535–562.
detection in cerebrospinal fluid by real-time quaking-induced con- •• Underlining the importance of biomarkers in the diagnosis and
version. Nat Med. 2011;17:175–178. the differential diagnosis of Alzheimer’s disease.
20. McGuire LI, Peden AH, Orru CD, et al. Real time quaking-induced 41. Graham WV, Bonito-Oliva A, Sakmar TP. Update on Alzheimer’s
conversion analysis of cerebrospinal fluid in sporadic Creutzfeldt- disease therapy and prevention strategies. Annu Rev Med.
Jakob disease. Ann Neurol. 2012;72:278–285. 2017;68:413–430.
21. Cramm M, Schmitz M, Karch A, et al. Stability and reproducibility 42. National Institute for Clinical Excellence (NICE). Donepezil,
underscore utility of RT-QuIC for diagnosis of Creutzfeldt-Jakob Galantamine, Rivastigmine and Memantine for the Treatment of
disease. Mol Neurobiol. 2016;53(3):1896–1904. Alzheimer’s Disease. NICE Technology Appraisal Guidance 217
22. Bongianni M, Orrù C, Groveman BR, et al. Diagnosis of human prion (Review of NICE Technology Appraisal Guidance 111). National
disease using real-time quaking-induced conversion testing of Institute for Clinical Excellence. 2011.
olfactory mucosa and cerebrospinal fluid samples. JAMA Neurol. 43. Barker WW, Luis CA, Kashuba A, et al. Relative frequencies of
2017;74(2):155–162. Alzheimer disease, Lewy body, vascular and frontotemporal
23. Hayashi Y, Iwasaki Y, Yoshikura N, et al. An autopsy-verified case of dementia, and hippocampal sclerosis in the state of Florida Brain
steroid-responsive encephalopathy with convulsion and a false- Bank. Alzheimer Dis Assoc Disord. 2002;16(4):203–212.
positive result from the real-time quaking-induced conversion 44. McKeith IG, Boeve BF, Dickson DW, et al. Diagnosis and manage-
assay. Prion. 2017;11:284–292. ment of dementia with Lewy bodies: fourth consensus report of
24. Vitali P, Maccagnano E, Caverzasi E, et al. Diffusion-weighted MRI the DLB Consortium. Neurol. 2017;89(1):88–100.
hyperintensity patterns differentiate CJD from other rapid demen- 45. Tartaglia MC, Johnson DY, Thai JN, et al. Clinical overlap between
tias. Neurology. 2011;76:1711–1719. Jakob-Creutzfeldt disease and Lewy body disease. Can J Neurol Sci.
25. Fragoso DC, Gonçalves Filho AL, Pacheco FT, et al. Imaging of 2012;39:304–310.
Creutzfeldt-Jakob disease: imaging patterns and their differential 46. Graff-Radford J, Aakre J, Savica R, et al. Duration and pathologic
diagnosis. Radiographics. 2017;37(1):234–257. correlates of Lewy body disease. JAMA Neurol. 2017;74(3):310–315.
26. Gaudino S, Gangemi E, Colantonio R, et al. Neuroradiology of 47. Garcia-Esparcia P, López-González I, Grau-Rivera O, et al. Dementia
human prion diseases, diagnosis and differential diagnosis. Radiol with Lewy Bodies: Molecular Pathology in the Frontal Cortex in
Med. 2017;122(5):369–385. Typical and Rapidly Progressive Forms. Front Neurol. 2017;8:89.
EXPERT REVIEW OF NEUROTHERAPEUTICS 11

48. Llorens F, Schmitz M, Varges D, et al. Cerebrospinal α-synuclein in 69. O’Brian JT, Thomas A. Vascular dementia. Lancet. 2015;386
α-synuclein aggregation disorders: tau/α-synuclein ratio as poten- (10004):1698–1706.
tial biomarker for dementia with Lewy bodies. J Neurol. 2016;263 70. Román GC, Tatemichi TK, Erkinjuntti T, et al. Vascular dementia:
(11):2271–2277. diagnostic criteria for research studies. Report of the NINDS-AIREN
49. Fairfoul G, McGuire LI, Pal S, et al. Alpha-synuclein RT-QuIC in the international workshop. Neurology. 1993;43:250–260.
CSF of patients with alpha-synucleinopathies. Ann Clin Transl 71. Jellinger KA. Pathology and pathogenesis of vascular cognitive
Neurol. 2016;3(10):812–818. impairment-a critical update. Front Aging Neurosci. 2013;5:17.
50. Shahnawaz M, Tokuda T, Waragai M, et al. Development of a 72. Skrobot OA, Black SE, Chen C, et al. Progress toward standardized
biochemical diagnosis of Parkinson disease by detection of α-synu- diagnosis of vascular cognitive impairment: guidelines from the
clein misfolded aggregates in cerebrospinal fluid. JAMA Neurol. vascular impairment of cognition classification consensus study.
2017;74(2):163–172. Alzheimers Dement. 2018;14(3):280–292.
51. Rolinski M, Fox C, Maidment I, et al. Cholinesterase inhibitors for 73. Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvas-
dementia with Lewy bodies, Parkinson’s disease dementia and cular disease - systematic review and meta-analysis. Neurobiol
cognitive impairment in Parkinson’s disease. Cochrane Database Aging. 2009;30(3):337–352.
Syst Rev. 2012;3:CD006504. 74. Hermann P, Romero C, Schmidt C, et al. CSF biomarkers and neuropsy-
52. Mori E, Ikeda M, Kosaka K, et al. Donepezil for dementia with Lewy chological profiles in patients with cerebral small-vessel disease. PLoS
bodies: a randomized, placebo-controlled trial. Ann Neurol. One. 2014 [cited 2018 Jul 5];7. DOI:10.1371/journal.pone.0105000
2012;72:41–52. 75. Skillbäck T, Delsing L, Synnergren J, et al. CSF/serum albumin ratio
53. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients with in dementias: a cross-sectional study on 1861 patients. Neurobiol
Parkinson’s disease dementia or dementia with Lewy bodies: a Aging. 2017;59:1–9.
randomised, double-blind, placebo controlled trial. Lancet Neurol. 76. Sjögren M, Blomberg M, Jonsson M, et al. Neurofilament protein in
2010;9:969–977. cerebrospinal fluid: a marker of white matter changes. J Neurosci
54. Weintraub D, Hurtig HI. Presentation and management of psycho- Res. 2001;66(3):510–516.
sis in Parkinson’s disease and dementia with Lewy bodies. Am J 77. Zerr I, Schmitz M, Karch A, et al. Cerebrospinal fluid neurofilament
Psychiatry. 2007;164:1491–1498. light levels in neurodegenerative dementia: diagnostic accuracy in
55. Morgante L, Epifanio A, Spina E, et al. Quetiapine and clozapine in the differential diagnosis of prion diseases. Alzheimers Dement.
Parkinsonian patients with dopaminergic psychosis. Clin 2018;14(6):751–763.
Neuropharmacol. 2004;27:153–156. 78. Bjerke M, Zetterberg H, Edman A, et al. Cerebrospinal fluid matrix
56. McCann H, Stevens CH, Cartwright H, et al. α-Synucleinopathy metalloproteinases and tissue inhibitor of metalloproteinases in com-
phenotypes. Parkinsonism Relat Disord. 2014;20(Suppl 1):S62–7. bination with subcortical and cortical biomarkers in vascular demen-
57. Bjornsdottir A, Gudmundsson G, Blondal H, et al. Incidence and pre- tia and Alzheimer’s disease. J Alzheimers Dis. 2011;27(3):665–676.
valence of multiple system atrophy: a nationwide study in Iceland. J
79. Wallin A, Kapaki E, Boban M, et al. Biochemical markers in vascular
Neurol Neurosurg Psychiatry. 2013;84(2):136–140.
cognitive impairment associated with subcortical small vessel dis-
58. Gilman S, Wenning GK, Low PA, et al. Second consensus statement
ease - A consensus report. BMC Neurol. 2017;17(1):102.
on the diagnosis of multiple system atrophy. Neurology. 2008;71
80. Kavirajan H, Schneider LS. Efficacy and adverse effects of cholines-
(9):670–676.
terase inhibitors and memantine in vascular dementia: a meta-
59. Stankovic I, Krismer F, Jesic A, et al. Cognitive impairment in multi-
analysis of randomised controlled trials. Lancet Neurol.
ple system atrophy: a position statement by the neuropsychology
2007;6:782–792.
task force of the MDS multiple system atrophy (MODIMSA) study
group. Mov Disord. 2014;29:857–867. 81. Gao B, Lyu C, Lerner A, et al. Controversy of posterior reversible
60. Laurens B, Constantinescu R, Freeman R, et al. Fluid biomarkers in encephalopathy syndrome: What have we learnt in the last 20
multiple system atrophy: a review of the MSA biomarker initiative. years? J Neurol Neurosurg Psychiatry. 2018;89(1):14–20.
Neurobiol Dis. 2015;80:29–41. 82. Vilela P. Acute stroke differential diagnosis: stroke mimics. Eur J
61. Papageorgiou SG, Kontaxis T, Bonakis A, et al. Rapidly progressive Radiol. 2017;96:133–144.
83. Salvarani C, Brown RD Jr, Calamia KT, et al. Primary central nervous
dementia: causes found in a Greek tertiary referral center in Athens.
system vasculitis: analysis of 101 patients. Ann Neurol.
Alzheimer Dis Assoc Disord. 2009;23:337–346.
2007;262:442–451.
• One of four reports on rapidly progressive dementia from
84. Salvarani C, Brown RD Jr, Christianson TJ, et al. Adult primary
tertiary hospitals.
central nervous system vasculitis treatment and course: analysis
62. Taniguchi-Watanabe S, Arai T, Kametani F, et al. Biochemical classi-
of one hundred sixty-three patients. Arthritis Rheumatol. 2015;67
fication of tauopathies by immunoblot, protein sequence and mass
(6):1637–1645.
spectrometric analyses of sarkosyl-insoluble and trypsin-resistant
85. Salvarani C, Brown RD Jr, Hunder GG. Adult primary central nervous
tau. Acta Neuropathol. 2016;131(2):267–280.
system vasculitis. Lancet. 2012;9843:767–777.
63. Perry DC, Brown JA, Possin KL, et al. Clinicopathological correla-
tions in behavioural variant frontotemporal dementia. Brain. 86. Calabrese LH, Mallek JA. Primary angiitis of the central nervous
2017;140(12):3329–3345. system. Report of 8 new cases, review of the literature, and propo-
64. Foster NL, Heidebrink JL, Clark CM, et al. FDG-PET improves accu- sal for diagnostic criteria. Medicine. 1988;67:20–39.
racy in distinguishing frontotemporal dementia and Alzheimer’s 87. Granerod J, Ambrose HE, Davies NW, et al. Causes of encephalitis
disease. Brain. 2007;130:2616–2635. and differences in their clinical presentations in England: a multi-
65. Lee SE, Rabinovici GD, Mayo MC, et al. Clinicopathological correlations centre, population-based prospective study. Lancet Infect Dis.
in corticobasal degeneration. Ann Neurol. 2011;70(2):327–340. 2010;10:835–844.
66. Höglinger GU, Respondek G, Stamelou M, et al. Clinical diagnosis of • Population-based data on different causes of inflammatory
progressive supranuclear palsy: the movement disorder society brain diseases.
criteria. Mov Disord. 2017;32(6):853–864. 88. Lancaster E, Dalmau J. Neuronal autoantigens—pathogenesis,
67. Schneider JA, Arvanitakis Z, Bang W, et al. Mixed brain pathologies associated disorders and antibody testing. Nat Rev Neurol. 2012;8
account for most dementia cases in community-dwelling older (7):380–390.
persons. Neurology. 2007;69(24):2197–2204. 89. Dalmau J, Graus F. Antibody-mediated encephalitis. N Engl J Med.
68. Dubois B, Feldman HH, Jacova C, et al. Advancing research diag- 2018;378(9):840–851.
nostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet •• A comprehensive and up-to-date guide to diagnosis and ther-
Neurol. 2014;13:614–629. apy of immune-mediated encephalitis.
12 I. ZERR AND P. HERMANN

90. Gozzard P, Woodhall M, Chapman C, et al. Paraneoplastic neurologic 99. Alford K, Vera JH. Cognitive impairment in people living with HIV in
disorders in small cell lung carcinoma: a prospective study. Neurology. the ART era: a review. Br Med Bull. 2018 cited 2018 Jul 5;14.
2015;85:235–239. DOI:10.1093/bmb/ldy019.
91. Graus F, Escudero D, Oleaga L, et al. Syndrome and outcome of 100. Compain C, Sacre K, Puéchal X, et al. Central nervous systeminvol-
antibody-negative limbic encephalitis. Eur J Neurol. 2018 [cited vement in Whipple disease: clinical study of 18patients and long-
2018 Jul 5];6. DOI:10.1111/ene.13661 term follow-up. Medicine. 2013;92(6):324–330.
92. Afshari M, Afshari ZS, Schuele SU. Pearls & oysters: hashimoto 101. Rosenbloom MH, Atri A. The evaluation of rapidly progressive
encephalopathy. Neurology. 2012;29;78(22):134–137. dementia. Neurologist. 2011;17(2):67–74.
93. Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor
102. Appleby BS, Lyketsos CG. Rapidly progressive dementias and the
encephalitis: case series and analysis of the effects of antibodies.
treatment of human prion diseases. Expert Opin Pharmacother.
Lancet Neurol. 2008;7(12):1091–1098.
2011;12:1–12.
94. Prüss H, Dalmau J, Harms L, et al. Retrospective analysis of NMDA
receptor antibodies in encephalitis of unknown origin. Neurology. 103. Mahajan S, Appleby BS. Comprehensive and methodical: diagnostic
2010;75:1735–1739. and management approaches to rapidly progressive dementia.
Curr Treat Options Neurol. 2017;19(11):40.
95. Dalmau J, Lancaster E, Martinez-Hernandez E, et al. Clinical experi-
104. Van der Meulen M, Dirven L, Habets EJJ, et al. Cognitive function-
ence and laboratory investigations in patients with anti-NMDAR
ing and health-related quality of life in patients with newly diag-
encephalitis. Lancet Neurol. 2011;10:63–74.
nosed primary CNS lymphoma: a systematic review. Lancet Oncol.
96. Tüzün E, Zhou L, Baehring JM, et al. Evidence for antibody- 2018;19(8):e407–e418.
mediated pathogenesis in anti-NMDAR encephalitis associated
105. Chinnery PF. Mitochondrial disorders overview. In: Adam MP,
with ovarian teratoma. Acta Neuropathol. 2009;118:737–743.
97. Irani SR, Bera K, Waters P, et al. N-methyl-D-aspartate antibody ence- ®
Ardinger HH, Pagon RA, et al. editors. GeneReviews [Internet].
phalitis: temporal progression of clinical and paraclinical observations in [Updated 2000 Jun 8; Updated 2014 Aug 14]. Seattle (WA):
a predominantly non-paraneoplastic disorder of both sexes. Brain. University of Washington, Seattle; 1993–2018.
2010;133:1655–1667. 106. Zhang Y, Gao T, Tao QQ. Spectrum of noncerebrovascular rapidly
progressive cognitive deterioration: a 2-year retrospective study.
98. Titulaer MJ, McCracken L, Gabilondo I, et al. Treatment and prog-
Clin Interv Aging. 2017;12:1655–1659.
nostic factors for long-term outcome in patients with anti-NMDA
• One of four reports on rapidly progressive dementia from
receptor encephalitis: an observational cohort study. Lancet
tertiary hospitals.
Neurol. 2013;12(2):157–165.

Das könnte Ihnen auch gefallen