Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 1.
Published in final edited form as: Mov Disord. 2017 May 13;32(7):995–1005. doi: 10.1002/mds.27034

Which ante-mortem clinical features predict progressive supranuclear palsy pathology?

Gesine Respondek 1,2, Carolin Kurz 3, Thomas Arzberger 2,3,4, Yaroslau Compta 5, Elisabet Englund 6, Leslie W Ferguson 7, Ellen Gelpi 8, Armin Giese 4, David J Irwin 9, Wassilios G Meissner 10,11,12, Christer Nilsson 13, Alexander Pantelyat 14, Alex Rajput 7, John C van Swieten 15, Claire Troakes 16, Keith A Josephs 17, Anthony E Lang 18, Brit Mollenhauer 19, Ulrich Müller 20, Jennifer L Whitwell 21, Angelo Antonini 22, Kailash P Bhatia 23, Yvette Bordelon 24, Jean-Christophe Corvol 25, Carlo Colosimo 26, Richard Dodel 27, Murray Grossman 9, Jan Kassubek 28, Florian Krismer 29, Johannes Levin 30, Stefan Lorenzl 31,32,33, Huw Morris 34, Peter Nestor 35, Wolfgang H Oertel 36, Gil D Rabinovici 37, James R Rowe 38, Thilo van Eimeren 39, Gregor K Wenning 29, Adam Boxer 37, Lawrence I Golbe 40, Irene Litvan 41, Maria Stamelou 36,42,*, Günter U Höglinger 1,2,*; for the Movement Disorder Society-endorsed PSP Study Group
PMCID: PMC5543934  NIHMSID: NIHMS870529  PMID: 28500752

Abstract

Background

Progressive supranuclear palsy (PSP) is a neuropathologically defined disease presenting with a broad spectrum of clinical phenotypes.

Objective

To identify clinical features and investigations that predict or exclude PSP pathology during life, aiming at an optimization of the clinical diagnostic criteria for PSP.

Methods

We performed a systematic review of the literature published since 1996 to identify clinical features and investigations that may predict or exclude PSP pathology. We then extracted standardized data from clinical charts of patients with pathologically diagnosed PSP and relevant disease controls, and calculated sensitivity, specificity and positive predictive value of key clinical features for PSP in this cohort.

Results

Of 4166 articles identified by database inquiry, 269 met predefined standards. The literature review identified clinical features predictive of PSP, including features of the 4 functional domains, i.e., “ocular motor dysfunction”, “postural instability”, “akinesia”, and “cognitive dysfunction”. No biomarker or genetic feature was found reliably validated to predict definite PSP.

High-quality original natural history data were available from 206 patients with pathologically diagnosed PSP and from 231 pathologically diagnosed disease controls (54 corticobasal degeneration, 51 multiple system atrophy with predominant parkinsonism, 53 Parkinson's disease, 73 behavioral variant frontotemporal dementia). We identified clinical features that predicted PSP pathology, including phenotypes other than Richardson's syndrome, with varying sensitivity and specificity.

Conclusions

Our results highlight the clinical variability of PSP and the high prevalence of phenotypes other than Richardson's syndrome. Features of variant phenotypes with high specificity and sensitivity should serve to optimize clinical diagnosis of PSP.

Keywords: Progressive supranuclear palsy, clinical features, diagnosis, clinico-pathological series, systematic review

Introduction

Progressive supranuclear palsy (PSP) is a neurodegenerative disease, first described by JC Richardson and colleagues in 19641. Its prevalence was estimated from a clinical perspective to be 3-6/100.0002,3. Age at symptom onset is 65 years and disease duration to death is 6-9 years, on average2,4. Neuropathological examination provides the gold standard for diagnosis, defining the disease entity5-7. The NINDS-SPSP (National Institute of Neurological Disorders and Stroke and Society for Progressive Supranuclear Palsy) criteria for the clinical diagnosis of PSP7 are commonly applied as ante mortem diagnostic standard. A diagnosis of “probable” PSP requires presence of vertical supranuclear gaze palsy (vSNP) plus postural instability (PI) and falls within 1 year of disease. For a diagnosis of “possible” PSP, either vSNP or a combination of slow vertical saccades and PI with falls within 1 year need to be present 7. This clinical manifestation of PSP with predominant ocular motor dysfunction and PI is called Richardson's syndrome (PSP-RS). The NINDS-SPSP criteria have excellent specificity8, 9 but low sensitivity early in the clinical course9-12 because typical features of PSP-RS are either absent or become apparent only after several years in a significant proportion of PSP patients4,9,10,13-34. A recent analysis of autopsy-confirmed patients suggested that 60-75% of patients with ascertained PSP pathology have variant PSP syndromes (vPSP) other than PSP-RS4. Too often, patients with vPSP, but also with PSP-RS, are diagnosed only after several years or never during lifetime4. However, early diagnosis is urgently warranted, since disease-modifying treatments are being developed, and would ideally be initiated before relevant cognitive or motor impairment is present35,36.

Therefore, the International Parkinson and Movement Disorder Society-endorsed PSP Study Group (MDS-PSPSG) aimed to optimize clinical diagnostic criteria for PSP. To identify clinical features and investigations that reliably predict or exclude PSP pathology, we first performed a systematic literature review. We then verified the diagnostic value of the suggested features in the largest clinico-pathological cohort of PSP reported thus far, in comparison to relevant disease controls [corticobasal degeneration (CBD), multiple system atrophy with predominance of parkinsonism (MSA-P), Parkinson's disease (PD), or frontotemporal lobar degeneration (from any underlying non-PSP/CBD proteinopathy) presenting with a behavioral variant frontotemporal dementia (FTLD-bvFTD)].

Methods

Systematic literature review

The MDS-PSPSG steering committee (GUH, ALB, IL, LIG, MS) assembled working groups of key experts to conduct a systematic review of published literature on specific aspects relevant to the diagnosis of PSP. Members of the groups defined key questions (Table 1) and search terms (Supplemental Table 1). Literature was searched on PubMed, Cochrane, Medline, and PSYCInfo databases for entries from 1996 until 2015, using search terms for each key question and for PSP (“Progressive Supranuclear Palsy” OR “Progressive Supranuclear Ophthalmoplegia” OR “Steele Richardson Olszewski syndrome”). Study group members were encouraged to add relevant articles for consideration throughout the project period (end of 2016), particularly those published after 2015. All titles and abstracts of identified articles were reviewed independently by 2 investigators (GH, CK) to select research articles, systematic reviews and meta-analyses, published in English using either a postmortem diagnosis or the highly specific NINDS-SPSP criteria as diagnostic standard. Three independent members of the consortium (GUH, GR, CK) analyzed methodology (internal validity, overall assessment, description) of these articles using the checklist of the Scottish Intercollegiate Guidelines Network (SIGN, www.sign.ac.uk)37 and rated their methodological quality as “very reliable”, “OK”, or “insufficient”. “Insufficient” articles were excluded from further analysis. For each included article, 2 independent experts collated standardized information on 1) study design, 2) evidence level, 3) patient characteristics, 4) key test or feature, 5) key findings, and 6) diagnostic value. Written summaries were provided to all MDS-PSPSG members. Evidence was summarized for each key question by the working groups (Supplemental Table 1).

Table 1. Key questions of the systematic literature review.

Key Question
1. How effective are clinical diagnostic criteria to predict or exclude neuropathologically defined PSP?
2. Which signs / symptoms /syndromes predict or exclude neuropathologically defined PSP?
3. How effective is acute / chronic levodopa / apomorphine testing to predict or exclude PSP?
4. How effective is objective autonomic function testing to exclude PSP?
5. How effective is neuropsychological testing to predict or exclude PSP?
6. How effective is clinical or quantitative oculomotor analysis to predict or exclude PSP?
7. How effective is genetic testing to predict or exclude PSP?
8. Is there a biomarker for PSP?

Clinico-pathological case series

This work was approved by the ethics committee of the Technical University of Munich and the participating centers. Autopsied cases with detailed clinical information and a definite diagnosis of PSP5,6,38, CBD5,39, MSA-P40, PD41, and a clinical syndrome of bvFTD42 with FTLD from any underlying non-PSP/CBD proteinopathy (i.e. FTLD-Tau including FTDP-17 or Pick's disease, FTLD-TDP or FTLD-FUS)43 were recruited from 9 brain banks with expertise in neurodegenerative diseases (Ludwig-Maximilians-University, Munich, Germany; University Hospital of Bordeaux, France; King's College, London, UK; University of Lund, Lund, Sweden; Erasmus Medical Center, Rotterdam, Netherlands; Hospital Clinic–IDIBAPS, Barcelona, Spain; University of Saskatchewan, Canada; Johns Hopkins University, Baltimore, USA; University of Pennsylvania, Philadelphia, USA). Before death, all donors had given written informed consent according to the Declaration of Helsinki for the scientific use of their brains and medical records. While cases with minor age-related co-pathology such as amyloid-beta pathology, primary age-ralated tauopathy (PART), argyrophilic grain disease (AGD), aging-related tau-astrogliopathy (ARTAG) were included into analysis, cases with more than 1 pathological diagnosis were excluded. Clinical data were acquired as described previously4. In short, demographic data, age at disease onset and death, disease duration, initial clinical diagnosis, final clinical diagnosis, and 37 clinical features (Supplemental Table 2) including year of their onset were abstracted from clinical records using a standardized template by local physicians. Features not specifically mentioned in the records were considered absent.

Statistics

Demographic and clinical data of groups were compared with analysis of variance (ANOVA) and post hoc Fisher's least significant difference test. P<0.05 was considered statistically significant. Prevalence, positive predictive value (PPV) and specificity of clinical features was calculated for PSP versus all control cases.

Results

Systematic Literature Review

Two key questions on radiological biomarkers of PSP are reported in detail in an accompanying paper in this issue of Movement Disorders44. The literature research for the 8 non-imaging key questions (Table 1) is reported herein. From the identified 4166 abstracts, 1035 publications met the criteria for full review. The methodology of 269 articles was considered “very reliable” or “OK”. Evidence pertinent to the diagnosis of PSP for each key question is shown in detail in Supplemental Table 1 and summarized in the following paragraphs.

The NINDS-SPSP criteria were found to be very specific, but not sensitive in the early clinical course9-12,45. Alternative clinical diagnostic criteria were not better for early diagnosis9,11. Major challenges are absence of features specific for PSP-RS, particularly ocular motor dysfunction in some patients with PSP, and clinical overlap with other diseases (particularly PD, MSA-P, CBD, and FTD).

Two large clinico-pathological series of PSP patients identified 3 coherent clusters of symptoms by hypothesis-free cluster analysis4,33. These were 1) vSNP and falls, 2) parkinsonian signs/symptoms, and 3) cognitive symptoms. We considered these functional domains most relevant to the clinical manifestation of definite PSP.

The literature from the past 20 years emphasizes vPSP syndromes other than PSP-RS: definite PSP patients were reported in small series with initial predominance of ocular motor dysfunction (PSP-OM)4, PI (PSP-PI)4,46, parkinsonism resembling idiopathic Parkinson's disease (PSP-P)10,33, frontal lobe cognitive or behavioral presentations (PSP-F)18,23,24, progressive gait freezing (PSP-PGF)16,19,34,47, speech/language disorders (PSP-SL) including non-fluent/agrammatic variant of primary progressive aphasia (nfaPPA)15,22,31 and progressive apraxia of speech (AOS)22,48, corticobasal syndrome (PSP-CBS)14,27,49, primary lateral sclerosis (PSP-PLS)32, or cerebellar ataxia (PSP-C)50,51.

A broad spectrum of diseases other than PSP have been clinically reported to present similarly to PSP (PSP look-alikes) and need to be considered as differential diagnosis (Supplemental Table 1).

Available studies on neuropsychological testing are limited by lack of autopsy confirmation and inclusion of mostly PSP-RS cases. The few studies with autopsy-confirmation revealed that PSP may present with features classically attributed to FTD, such as bvFTD18,23,24 and nfaPPA31. The typical frontal syndrome of PSP appears to comprise apathy, bradyphrenia (i.e., slowness of thinking), executive dysfunction, reduced phonemic verbal fluency, impulsivity, disinhibition, and perseveration52-56.

Studies addressing ocular motor analysis in autopsy-confirmed cases reported high specificity of vSNP and reduced vertical saccade velocity for PSP57,58. Further studies reported,,eyelid-opening apraxia“59-61, frequent macro square wave jerks62, and non-specific ocular symptoms (diplopia28,60, blurred vision28,60, burning eye sensation28, photophobia28,60,63, blepharospasm59,64, and reduced blinking rate65) as characteristic for PSP.

The literature on genetics of PSP confirms that PSP is generally sporadic. However, non-Mendelian family histories for neurodegenerative diseases were found in up to 33% of PSP index patients66,67. Homozygosity for the H1 haplotype of MAPT and polymorphisms at STX6, MOBP and EIF2AK3 represent risk factors for sporadic PSP68. Mendelian inheritance of PSP-like syndromes occurs rarely due to MAPT mutations69,70 but the similarity to PSP-RS is only partial. Mutations in other genes can present as PSP-like syndromes, but they have either no or an uncertain relationship to definite PSP (Supplemental Table 1).

Studies on fluid biomarkers in PSP lack of sufficient sample numbers, homogeneity (clinical phenotype, co-morbidities, co-medication, etc.), and autopsy-confirmation, but also suffer technical shortcomings (heterogeneous sample processing, assay limitations, lack of independent confirmation). It is established, however, that CSF concentrations of total and phosphorylated tau are not increased in PSP patients, unlike in Alzheimer's disease (AD)71.

Clinico-pathological case series

Detailed clinical data were available for autopsy confirmed cases of PSP (N=206), CBD (N=54), MSA-P (N=51), PD (N=53), and FTLD-bvFTD (N=73). Characteristics of 100/206 PSP patients have been described previously4. A subset of control patients (CBD, MSA-P, PD, FTLD-bvFTD) have been published previously in disease-specific studies but not in systematic comparative evaluations of PSP features. Their demographic data, as shown in Table 2, are consistent with previously published data4,72-75. No PSP patient had a symptom onset prior to 41 years of age, nor a Mendelian inheritance pattern or a known MAPT mutation. FTLD cases had varying pathology (75% TDP-43, 21% Tau (other than PSP or CBD), 4% FUS); 34% (N=25) were hereditary, 5% (N=4) with MAPT mutations, 29% (N=21) with known mutations other than MAPT; 23% (N=17) were associated with motor neuron disease.

Table 2. Demographic data of the pathology confirmed cohort.

PSP CBD MSA-P PD FTLD-bvFTD
N 206 54 51 53 73
Age at onset 66.2 ± 0.6
[41-91]
63.3 ± 1.3*
[42-81]
59.3 ± 1.3***
[40-80]
58.8 ± 1.5***
[40-80]
57.1 ± 1.0***
[35-74]
Age at death 74.0 ± 0.6
[54-94]
69.8 ± 1.2**
[49-85]
66.8 ± 1.2***
[51-90]
73.1 ± 1.2
[56-90]
63.8 ± 1.2***
[41-84]
Disease duration 7.9 ± 0.3
[2-27]
6.8 ± 0.4
[3-12]
7.2 ± 0.4
[2-15]
14.6 ± 1.0***
[3-34]
6.7 ± 0.5
[1-20]

Demographic data of definite PSP, CBD, MSA, PD, and FTD patients. Data are mean ± SD [range]. ANOVA followed by post hoc LSD test:

*

P<0.05,

**

P<0.01,

***

P<0.001, vs. PSP.

Abbreviations: PSP, progressive supranuclear palsy; CBD, corticobasal degeneration; MSA-P, multiple system atrophy with predominant parkinsonism; PD, Parkinson's disease; FTLD, frontotemporal lobar degeneration; bvFTD, behavioral variant of frontotemporal dementia.

Table 3 lists the diagnoses made at initial and final ante mortem clinical evaluation. For 122 (59%) and 182 (88%) of 206 PSP patients, initial and final diagnoses were recorded, respectively. PSP was correctly diagnosed in 31 of 122 cases (25%) initially and in 114 of 182 patients (63%) at final visit. PD was the most common clinical misdiagnosis in PSP patients [28/122 (23%) initially, 18/182 (10%) at final visit].

Table 3. Initial and final clinical diagnosis in pathologically diagnosed patients.

PSP CBD MSA-P PD FTLD-bvFTD






Initial clinical diagnosis %
(N / N)
PSP 25.4 (31/122) 6.7 (2/30) 6.3 (2/32) 0.0 (0/34) 1.9 (1/52)
CBS 1.6 (2/122) 0.0 (0/30) 0.0 (0/32) 0.0 (0/34) 0.0 (0/52)
MSA 0.0 (0/122) 3.3 (1/30) 6.3 (2/32) 2.9 (1/34) 0.0 (0/52)
PD 23.0 (28/122) 3.3 (1/30) 71.9 (23/32) 82.4 (28/34) 0.0 (0/52)
FTD 9.8 (12/122) 33.3 (10/30) 0.0 (0/32) 0.0 (0/34) 76.9 (40/52)
FTD-MND 0.0 (0/122) 0.0 (0/30) 0.0 (0/32) 0.0 (0/34) 7.7 (4/52)
MND 0.8 (1/122) 0.0 (0/30) 0.0 (0/32) 0.0 (0/34) 1.9 (1/52)
Parkinsonism 11.5 (13/122) 13.3 (4/30) 0.0 (0/32) 2.9 (1/34) 0.0 (0/52)
LBD 1.6 (2/122) 0.0 (0/30) 0.0 (0/32) 0.0 (0/34) 0.0 (0/52)
NPH 0.0 (0/122) 0.0 (0/30) 3.1 (1/32) 0.0 (0/34) 0.0 (0/52)
AD 1.6 (2/122) 6.7 (2/30) 0.0 (0/32) 0.0 (0/34) 0.0 (0/52)
Dementia 2.5 (3/122) 6.7 (2/30) 0.0 (0/32) 0.0 (0/34) 3.8 (2/52)
Cerebral vasculopathy 2.5 (3/122) 3.3 (1/30) 0.0 (0/32) 0.0 (0/34) 0.0 (0/52)
Essential tremor 0.8 (1/122) 0.0 (0/30) 0.0 (0/32) 2.9 (1/34) 0.0 (0/52)
Depression 4.1 (5/122) 0.0 (0/30) 0.0 (0/32) 0.0 (0/34) 0.0 (0/52)
Mixed 7.4 (9/122) 10.0 (3/30) 6.3 (2/32) 5.9 (2/34) 5.8 (3/52)
Other 8.2 (10/122) 18.8 (6/30) 6.3 (2/32) 2.9 (1/34) 1.9 (1/52)
Final clinical diagnosis
% (N / N)
PSP 62.6 (114/182) 13.7 (7/51) 5.9 (3/51) 7.8 (4/51) 1.4 (1/69)
CBS 1.6 (3/182) 27.5 (14/51) 2.0 (1/51) 0.0 (0/51) 2.9 (2/69)
MSA 1.6 (3/182) 0.0 (0/51) 70.6 (36/51) 3.9 (2/51) 0.0 (0/69)
PD 9.9 (18/182) 0.0 (0/51) 11.8 (6/51) 80.4 (41/51) 0.0 (0/69)
FTD 3.3 (6/182) 29.4 (15/51) 0.0 (0/51) 0.0 (0/51) 65.2 (45/69)
FTD-MND 0.0 (0/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 10.1 (7/69)
MND 0.5 (1/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 4.3 (3/69)
Parkinsonism 2.7 (4/182) 3.9 (2/51) 2.0 (1/51) 3.9 (2/51) 0.0 (0/69)
LBD 0.5 (1/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
NPH 0.0 (0/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
AD 3.8 (7/182) 3.9 (2/51) 0.0 (0/51) 0.0 (0/51) 5.8 (4/69)
Dementia 2.2 (4/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
Cerebral vasculopathy 0.5 (1/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
Essential tremor 0.0 (0/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
Depression 0.0 (0/182) 0.0 (0/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)
Mixed 8.2 (15/182) 17.6 (9/51) 7.8 (4/51) 3.9 (2/51) 10.1 (7/69)
Other 2.7 (5/182) 3.9 (2/51) 0.0 (0/51) 0.0 (0/51) 0.0 (0/69)

Initial and final clinical diagnosis of autopsy-confirmed patients with PSP, CBD, MSA-P, PD, and FTLD-bvFTD. Data are % (N with specific clinical diagnosis / N with any record of clinical diagnosis) of patients per group. Values in bold indicate the correct clinical diagnosis. Abbreviations: AD, Alzheimer's dementia; bvFTD, behavioral variant of frontotemporal dementia; CBD, corticobasal degeneration; CBS, corticobasal syndrome; FTD, frontotemporal dementia; FTD-MND, frontotemporal dementia with motor neuron disease; FTLD, frontotemporal lobar degeneration; LBD, Lewy body dementia; MSA, multiple system atrophy; MSA-P, multiple system atrophy with predominant parkinsonism; NPH, normal pressure hydrocephalus; PD, Parkinson's disease; PSP, progressive supranuclear palsy.

The frequency of 37 clinical features throughout the disease course is shown in Supplemental Table 3. Based on the literature review we selected symptoms of putative diagnostic value and calculated their sensitivity, PPV, and specificity in our clinico-pathological cohort (Table 4).

Table 4. Sensitivity, positive predictive value and specificity of clinical features for PSP.

Clinical features Sensitivity for PPV for PSP Spec. for PSP
PSP CBD MSA-P PD FTLD-bvFTD
Ocular Motor Dysfunction
Supranuclear gaze palsy 70.9% 14.8% 7.8% 11.3% 2.7% 88% 91%
Supranuclear gaze palsy within 3 years 29.6% 9.3% 2.0% 3.8% 0% 88% 97%
Abnormal saccades 65.5% 25.9% 21.6% 7.5% 6.8% 80% 85%
Abnormal saccades within 3 years 30.6% 13.0% 7.8% 1.9% 2.7% 82% 94%
Nonspecific ocular symptoms 35.9% 11.1% 2.0% 5.7% 5.5% 84% 94%
Postural Instability
Postural Instability 82.0% 48.1% 90.2% 75.5% 16.4% 58% 46%
Postural Instability within 3 years 53.9% 20.1% 45.1% 11.3% 5.5% 72% 81%
Postural Instability within 1 year 44.7% 13% 23.5% 5.7% 1.4% 74% 90%
Falls 78.6% 37.0% 66.7% 66.0% 8.2% 64% 59%
Falls within 3 years 51.0% 16.7% 29.4% 7.5% 5.5% 77% 86%
Falls within 1 year 37.4% 11.1% 13.7% 3.8% 0% 84% 94%
Akinesia
Parkinsonism, akinetic-rigid, predominantly axial & levodopa-resistant 28.6% 7.4% 9.8% 7.5% 8.2% 76% 92%
Parkinsonism, with tremor and/or asymmetric and/or levodopa-responsive 44.2% 38.9% 78.4% 86.8% 21.9% 43% 47%
Progressive gait freezing within 3 years 1.9% 0% 0% 0% 0% 100% 100%
Cognitive Dysfunction
Non-fluent/agrammatic primary progressive aphasia 18.9% 27.8% 0% 1.9% 16.4% 58% 88%
Non-fluent/agrammatic primary progressive aphasia within 3 years 9.2% 20.4% 0% 0% 12.3% 49% 91%
Apraxia of speech 4.4% 18.5% 0% 3.8% 2.7% 39% 94%
Apraxia of speech within 3 years 1.0% 7.4% 0 (0% 0 (0% 2.7% 25% 97%
Frontal dysfunction 57% 66.7% 21.6% 28.3% 86.8% 50% 46%
Frontal dysfunction within 3 years 30% 40.7% 3.9% 7.5% 79.5% 45% 63%
Corticobasal Syndrome
at least one of limb rigidity/akinesia/ dystonia/myoclonus + at least one of apraxia/cortical sensory deficit/alien limb 12.6% 22.2% 2.0% 1.9% 2.7% 62% 93%
Bulbar Dysfunction
Dysarthia 69.4% 35.2% 66.7% 66.0% 26.0% 57% 54%
Dysarthia within 3 years 32.5% 14.8% 21.6% 11.3% 17.8% 64% 84%
Dysphagia 65.0% 37.0% 62.7% 43.4% 41.1% 56% 55%
Dysphagia within 3 years 23.8% 11.1% 11.8% 5.7% 20.5% 62% 87%

Sensitivity, PPV, and specificity of selected symptoms in the clinico-pathological cohort of patients with PSP, CBD, MSA-P, and FTLD-bvFTD. Data are %.

Abbreviations: PSP, progressive supranuclear palsy; CBD, corticobasal degeneration; MSA-P, multiple system atrophy with predominant parkinsonism; PD, Parkinson's disease; FTLD, frontotemporal lobar degeneration; bvFTD, behavioral variant of frontotemporal dementia; PPV, positive predictive value; Spec., specificity.

Ocular motor dysfunction

Presence of vSNP throughout the disease had a specificity of 91% for definite PSP, increasing to 97% when present within 3 years after disease onset; sensitivity was 71% throughout, but only 30% within 3 years. Because the retrospective analysis did not provide reliable data on slowing of saccades for many patients, the term was generalized to abnormal saccades. These were recorded in 66% of PSP patients throughout the disease, yielding a specificity of 85%. When present within 3 years, sensitivity for PSP decreased to 31% but specificity increased to 94%. Non-specific ocular symptoms (defined as any of painful eyes, dry eyes, visual blurring, diplopia, blepharospasm, ptosis, reduced blinking rate, or “apraxia of eyelid opening”) were recorded in 36% of PSP patients throughout the disease, but only 2-11% in control groups, yielding a high specificity (94%).

Postural instability

PI throughout the disease course was the most frequent symptom in PSP (82%), but had low specificity (46%), also being common in CBD, MSA-P and PD. PI within 3 years was observed in only 54% of PSP patients, but had considerably better specificity (81%). PI within 1 year had even higher specificity (90%), but lower sensitivity (45%).

Falls throughout the disease course had moderate sensitivity (79%) and specificity (59%) for PSP. Falls within 3 years had reduced sensitivity (51%), but improved specificity (86%). Falls within 1 year resulted in even lower sensitivity (37%) and only slightly improved specificity (94%).

Akinesia and gait freezing

Akinetic-rigid, predominantly axial, and levodopa-resistant parkinsonism distinguished PSP with a sensitivity of only 29% and specificity of 92%. Parkinsonism with tremor and/or asymmetry and/or levodopa-responsiveness identified PSP with higher sensitivity (44%) but lower specificity (47%). The most specific symptom for PSP (100%) was progressive gait freezing within 3 years, defined as gait freezing or start hesitation in absence of limb rigidity, tremor, or dementia and without response to levodopa34; however it was only present in very few PSP patients (2%).

Cognitive dysfunction

Non-fluent/agrammatic primary progressive aphasia (nfaPPA) was present in 19% of definite PSP cases. nfaPPA also occurred in FTLD-bvFTD (16%) and in CBD (28%). Specificity of nfaPPA for PSP in our cohort was 88%. When noted within 3 years, nfaPPA had lower sensitivity (9%), but higher specificity (91%) for PSP. Apraxia of speech (AOS) had low sensitivity (4%) but high specificity (94%) for PSP. When considering AOS within 3 years only, sensitivity was even lower (1%) and specificity higher (97%). However, it should be noted that only cases of FTLD-bvFTD patients were included in our study and thus, including FTLD-PPA cases could have reduced specificity of these findings for PSP.

Frontal dysfunction was defined as presence of at least 1 of the following: personality change, frontal behavior, social dysfunction, executive dysfunction, and frontal physical signs. Information on the type of frontal dysfunction was too limited for differential analysis. Presence of frontal dysfunction throughout the course had a sensitivity for PSP of 57% and a specificity of 46%. When present within 3 years, sensitivity was only 30% and specificity was 63%.

Corticobasal syndrome (CBS), defined as at least one cortical and one movement disorder sign76 occurred in 13% of our PSP cases, yielding a specificity of 93% for PSP. As expected, CBD was the most relevant differential diagnosis.

Other features

Dysarthria and dysphagia throughout the disease course had limited specificity for PSP (54% and 55%, respectively). Specificity increased markedly for both symptoms if these were present within 3 years of disease (84% and 87%, respectively).

Ten PSP cases (5%) and 55 control cases (24%) had clinical features considered to be supportive for diagnoses other than PSP. These were 1) impairment of episodic memory within 1 year, suggestive of AD (3 PSP, 8 CBD, 2 PD, 1 FTLD), 2) unexplained autonomic failure within 1 year, suggestive of MSA (5 PSP, 10 MSA-P, 1 PD, 1 CBD), 3) unexplained visual hallucinations within 1 year, suggestive of dementia with Lewy bodies (1 PSP), 4) unexplained multisegmental upper and lower motor neuron signs, suggestive of motor neuron disease (17 FTLD), 5) appendicular ataxia (1 PSP, 2 MSA-P), and 6) hereditary cases with mutations other than MAPT (21 FTLD).

Discussion

Here we sought to identify ante-mortem clinical features that individually predict PSP pathology, by analyzing retrospective clinical data from a large autopsy cohort of PSP, CBD, MSA-P, PD and FTLD patients. For the same purpose, we conducted an extensive systematic literature review on features relevant for the diagnosis of PSP, published since 1996. The need for this work is apparent from the low rate of correct clinical diagnoses observed in this very cohort. Clinical diagnosis of PSP was correct in only 25% of cases at first visit, and in 63% at last visit, highlighting that PSP is under diagnosed. One reason for clinical under diagnosis of pathologically defined PSP is its phenotypic variability9-12,45. In vPSP syndromes other than PSP-RS, key features may be missing, especially early in the disease course. Indeed, of the PSP patients reported here, 33% never developed vSNP, and 23% did not have PI and falls, similar to previous observations4,9,10,33,46. This explains suboptimal sensitivity of the NINDS-SPSP clinical diagnostic criteria, as confirmed in our literature review9-11. The results of this study should serve as a framework to develop new clinical diagnostic criteria for PSP.

As demonstrated in our clinic-pathological analysis, key features of PSP-RS are highly specific for PSP pathology. VSNP, abnormal saccades, PI, and falls were highly specific for PSP, in agreement with previous reports7,28,38. Notably, CBS was also very specific for PSP (93%). A limited specificity of CBS for CBD was reported previously26, challenging the concept of CBS as the hallmark of CBD. Other features with high specificity for PSP included progressive gait freezing within 3 years (100% specificity)16,34, as well as AOS and nfaPPA15,17,20. However, since only FTLD-bvFTD was included in our cohort, the high specificity of the latter features must be interpreted with caution. Addition of cases with FTLD-PPA would have reduced specificity of PPA features for PSP. Thus, in patients with AOS and nfaPPA, a diagnosis of PSP should be suspected, however, additional PSP-specific features should be present to reliably predict PSP pathology, as evident in the literature15,22,31,48. Interestingly, predominantly axial and levodopa-resistant parkinsonism, a feature not mentioned in the NINDS-SPSP criteria, was reasonably specific for PSP pathology. Thus, to diagnose PSP with high specificity, the aforementioned features should be considered when designing new criteria.

A major shortcoming of the NINDS-SPSP criteria is low sensitivity9-11. With regard to key features of PSP-RS, expanding the time window for onset of PI and falls from 1 year (as required in the NINDS-SPSP criteria) to 3 years after disease onset resulted in improved sensitivity for a diagnosis of PSP (falls: from 37 to 51%; PI: from 45 to 54%). Similarly, presence of frontal dysfunction, dysarthria and/or dysphagia, and parkinsonism with tremor and/or asymmetry and/or levodopa-response had good sensitivity; however, specificity of these findings for PSP was, as expected, limited. Frontal dysfunction was also common in CBD and bvFTD, and dysphagia and dysarthria were frequently present in MSA-P and bvFTD, often early in the disease course. In summary, the results of this cohort study show that 1) there are a variety of features that may be acknowledged in order to increase sensitivity of diagnosing PSP, and 2) not unexpectedly, this increase in sensitivity comes at the expense of specificity.

Ideally, diagnostic investigations should be added to the clinical diagnostic criteria of PSP to increase both specificity and sensitivity. In this context, neuroimaging merits a separate discussion, and is addressed in detail in a comprehensive review of the MDS-PSPSG in an accompanying paper44. In short, brain imaging is useful to rule out differential diagnosis (Supplemental Table 1) and may be useful to support a clinical diagnosis of PSP-RS44. While midbrain atrophy amongst other markers reliably discriminates PSP-RS from disease controls, this seems not the case for other PSP phenotypes44. Imaging markers for atypical PSP phenotypes, e.g., to predict PSP pathology in CBS and FTD would be most desirable, but studies with autopsy confirmation are missing so far44.

Our literature review on other diagnostic investigations highlights the lack of any in vivo investigation that reliably predicts PSP pathology and might be useful for the clinical diagnostic criteria. While autopsy confirmation was available in a reasonable number of retrospective studies that reported on natural history of PSP, it was missing in most studies related to additional diagnostic investigations, including autonomic testing, neuropsychological testing, oculomotor analysis, and biomarker assessments. In these studies, only PSP-RS patients were evaluated, and the use/extent of additional diagnostic investigations in a more challenging diagnostic context is unclear. With regard to genetic testing, homozygosity for MAPT H1 haplotype polymorphism is frequent in, but not diagnostic for PSP. Homozygosity for MAPT H2 haplotype polymorphism is very rare in PSP, but does not exclude the diagnosis. Rare MAPT mutations can cause a PSP-like presentation, albeit obviously with a distinct etiology than the sporadic disease. However, genetic testing, as well as CSF biomarkers, can be helpful to identify PSP look-alikes, including prion disease, hereditary spinocerebellar ataxias, Perry syndrome, Kufor-Rakeb disease, Whipple's disease, Niemann-Pick disease type C, Gaucher's disease, progressive encephalomyelitis with rigidity and myoclonus, and AD. AD rarely presents clinically as PSP-RS or levodopa-resistant parkinsonism, but can clinically mimic other PSP-phenotypes, most notably CBS (in up to 25% of cases), but also nfaPPA and bvFTD; thus AD biomarkers (CSF Aβ and tau, amyloid PET) may be indicative of primary AD pathology in these syndromes. Research should be encouraged to identify investigations useful for early clinical diagnosis of PSP, as is the case in other neurodegenerative disorders such as AD77. It remains to be seen whether tau-PET imaging will qualify as a useful ancillary test.

Lastly, we were unable to identify specific features that could convincingly describe a characteristic prodromal phase of PSP, although individual reports clearly described speech/language, behavioral, or cognitive features as pre-motor manifestations in patients diagnosed with PSP upon follow-up or autopsy. Early clinical features of PSP are yet poorly addressed in the literature. It will be crucial to prospectively study putative PSP patients presenting with non-specific, albeit suggestive features in a prospective setting, starting at the earliest clinical stages of the disease course.

There are several limitations of our work. Data were obtained from clinical charts and may be incomplete, as in any retrospective clinico-pathological study. The numbers of cases per diagnosis did not present relative frequencies expected in the general population. To our knowledge, the only community-based autopsy series focusing pathologically defined diseases of interest for our study, found the following relative frequencies in 233 autopsy cases: 19% PD, 13% TDP-43 proteinopathy, 3% PSP (including anatomically restricted forms), 0.9% PSP sensu stricto, 0.9% MSA, and 0.4% CBD78. Any approach to mimic the population-based prevalence of diseases would have required introducing a massive distortion of brain bank frequencies. Hence, the relatively low numbers of PD cases included in this cohort introduces a bias that needs to be recognized when interpreting PPV and specificity. A selection bias over-representing cases with unusual clinical features cannot be excluded either. Thus PPV and specificity of clinical features for PSP might be underestimated in this cohort. The presence of some features, such as slowing of vertical saccades, may not have been documented, or missed entirely. However, the presented data was extracted from the largest clinico-pathological cohort of PSP published to date, and compared with a substantial number of pathologically confirmed patients with the most relevant differential diagnoses. In future, multivariate statistical models may increase diagnostic accuracy by considering clinical variables jointly, rather than singly, but such models are less easy to apply in the clinical setting.

In conclusion, our work provides a strong rationale for developing new diagnostic criteria for PSP. The various phenotypes and symptoms from 4 functional domains (“ocular motor dysfunction”, “postural instability”, “akinesia”, “cognitive dysfunction”), with characteristic clinical features each, should be considered when conceptualizing new criteria. Different levels of diagnostic certainty will need to be implemented into the criteria to allow for inclusion of symptoms with differing sensitivity and specificity, including features which are non-specific, but relevant for early and sensitive diagnosis of PSP.

Supplementary Material

Supp info

Acknowledgments

Founding source for study We thank all brain donors and their families for their generous donation allowing to advance our knowledge about PSP, clinicians for providing reliable clinical data, Ina B. Kopp for guidance in the methods of evidence-based medicine, Judith Dams for conducting the database inquiry. GUH was supported by the Deutsche Forschungsgemeinschaft (DFG, HO2402/6-2). WHO is senior research Professor of the charitable Hertie Foundation, Frankfurt/Main, Germany. JLW and KAJ were supported by NIH grants R01-NS89757 and R01-DC12519. Autopsy patient data from University of Pennsylvania was obtained through NIH funded program projects P01-AG017586, P50-NS053488 and P30-AG010124. The London Neurodegenerative Diseases Brain Bank, King's College London was supported by the MRC and Brains for Dementia Research - jointly funded by the Alzheimer's Society and Alzheimer's Research UK. HRM is supported by the PSP Association and CBD-Solutions. The project was supported by the Bischof Dr. Karl Golser Stiftung, CurePSP, Deutsche Forschungsgemeinschaft (DFG, HO 2402/11-1), German Center for Neurodegenerative Diseases e.V. (DZNE), German PSP Gesellschaft, Tau Consortium, UK PSP Association, and the International Parkinson & Movement Disorder Society.

Financial Disclosures of all authors for the preceding 12 months: Gesine Respondek has nothing to disclose.

Carolin Kurz has nothing to disclose.

Thomas Arzberger has nothing to disclose.

Yaroslau Compta has received funding, research support and/or honoraria in the last 5 years from UCB, Lundbeck, Medtronic, Abbvie, Novartis, GSK, Boehringer, Pfizer, Merz, Piramal Imaging and Esteve.”

Elisabet Englund has nothing to disclose.

Leslie Ferguson has nothing to disclose.

Ellen Gelpi has received research support from the Fundació Marató de TV3 (grant n° 20141610), and Fundación Tatiana Pérez de Guzmán del Bueno.

Armin Giese holds stocks and shares in and has personal links with MODAG. He also holds shares in the patent “EP2307381” and diverse national secondary patents.

David J Irwin is supported by National Institutes of Health grant K23 NS088341

Wassilios G Meissner has received fees for editorial activities with Springer, has served as advisor for Zambon France and Sanofi, has received teaching honoraria from TEVA, UCB and Aguettant, as well as research support from the Michael J Fox Foundation, the University Hospital Bordeaux, the French Health Ministry, the European Community, ANR, PSP-France, MSA Coalition, LABEX Excellence Initiative.

Christer Nilsson has received research support from the Swedish Alzheimer Fund, Skåne University Hospital grants and the Swedish Research Council ALF grants.

Alexander Pantelyat was supported by NIH grants P50NS03837701 and U01NS082133-04.

Alex Rajput has received research support from the Regina Curling Classic, Greystone Classic for Parkinson's, Inc. and the Dr. Ali Rajput Endowment for Parkinson's Disease and Movement Disorders; has been co-investigator on grant funded by International Essential Tremor Foundation (IETF) (study period 2012-2013) ; has received research funding as principal investigator for the clinical study by Teva, protocol no.: TVP-1012/501 (Aug/09 – June/13); has received speaker and travel honoraria from Teva and Allergan.

John van Swieten has nothing to disclose.

Claire Troakes has nothing to disclose.

Keith A Josephs was supported by NIH grants R01-NS89757 and R01-DC12519.

Anthony E Lang has served as an advisor for Abbvie, Acorda, Avanir Pharmaceuticals, Bristol-Myers Squibb, Ceregene, Lilly, Merck, and UCB; received honoraria from Medtronic, Teva, UCB, AbbVie; received grants from Brain Canada, Canadian Institutes of Health Research, Edmond J Safra Philanthropic Foundation, Michael J. Fox Foundation, the Ontario Brain Institute, National Parkinson Foundation, Parkinson Society Canada, Physicians Services Incorporated (PSI), W. Garfield Weston Foundation; received publishing royalties from Saunders, Wiley-Blackwell, Johns Hopkins Press, and Cambridge University Press; and has served as an expert witness in cases related to the welding industry.

Brit Mollenhauer received honoraria for consultancy from Roche, Biogen and GE Healthcare. BM is member of the executive steering committee of the Parkinson Progression Marker Initiative and the Systemic Synuclein Sampling Study of the Michael J. Fox Foundation for Parkinson's Research and has received grants from the BMBF, EU, Deutsche Parkinson Vereinigung, Parkinson Fonds Deutschland, Michael J. Fox Foundation for Parkinson's Research, Stifterverband für die deutsche Wissenschaft, and has scientific collaborations with Roche, Bristol Myers Squibb, Ely Lilly, Covance and Biogen.

Ulrich Müller has nothing to disclose.

Jennifer L Whitwell was supported by NIH grants R01-NS89757 and R01-DC12519.

Angelo Antonini received funding from Horizon2020 Project No 643706. He has also received consultancy fees and honoraria for speaker-related activities from AbbVie, UCB, Zambon, General Electric, Boston Scientific, Mundipharma, Acadia, Medtronic, Angelini Pharmaceuticals, Neuroderm.

Kailash Bhatia has received grant support from Welcome/MRC, NIHR, Parkinsons's UK and EU Horizon 2020. He has received honoraria/consulting fees for speaker related activities from Ipsen, Allergan, Merz, Sun Pharma, Teva, UCB pharma companies and from the American Academy of Neurology and Movement Disorders Society. He is an editor of MDCP journal and receives an honorary stipend from MDS for this activity and has received royalties for publications of books by Oxford University Press.

Yvette Bordelon received research support from AbbVie, C2N diagnostics and Bristol-Myers Squibb and speakers' honoraria from Teva Pharmaceuticals.

Jean Christophe Corvol received grants or research support from AP-HP (DRC-PHRC), the French drug regulation agency (ANSM), Sanofi-Aventis, the Michael J Fox Foundation; served on scientific advisory board for Zambon, Abbvie, BMS, and Amarentus; has stock option in B&A Therapeutics; received travel funding from the Movement Disorder Society, Teva, Lundbeck, and UCB.

Carlo Colosimo received consulting fees as an advisory board member and honoraria as a speaker from Ipsen, Merz, Zambon.

Richard Dodel has received research support from the following institutaions: AOK Plus, Baxter, BMBF, DFG, DGN, DPG, EU Horizon 2020, Faber-Stiftung, Hector-Stiftung, IPF, Novartis, Rhön-klinikum. He served as an advisory board member and received honoraria for scientific presentations from DZNE, Lundbeck, Med Update, Novartis, Paul-Martini-Stiftung, Studienstiftung des deutschen Volkes, Astra Zeneca, Avios Consulting, Baxter, Lilly, Med Panel, Merz, Novartis, Octapharma, Pfizer, Piramal.

Murray Grossman has nothing to disclose.

Jan Kassubek received consulting fees as an advisory board member and honoraria as a speaker from UCB Pharma, Teva Pharmaceuticals, Zambon, Medtronic, Desitin, AbbVie, Boehringer Ingelheim, GlaxoSmithKline, Merz Pharmaceuticals and Hoffmann-La Roche.

Florian Krismer has received a research grant from the MSA Coalition, travel grants from the Austrian Parkinson's disease society as well as the International Parkinson's disease and movement disorders Society and non-financial support from Fight MSA and Astra-Zeneca, outside the submitted work

Johannes Levin has received study support (third party funds) from Parkinson Fonds Deutschland GmbH (a private organization that provides grants for research into Parkinson's disease) and lecture fees from Bayer Healthcare and MSD.

Stefan Lorenzl has served on the advisory boards for TEVA, Boehringer and UCB; has received honoraria for scientific presentations from Teva, UCB, Boehringer and has received research support from the German Parkinson's Disease Foundation (DPG), German PSP Association (PSP Gesellschaft), German Research Foundation (DFG) and the German Ministry of Education and Research (BMBF).

Huw Morris has received grants from Medical Research Council UK, Wellcome Trust, Parkinson's UK, Ipsen Fund, Motor Neurone Disease Association, Welsh Assembly Government, PSP Association, CBD Solutions and Drake Foundation, and payment for advisory work/consulting and lectures from Bristol-Myers-Squibb, GE-HealthCare, Alzprotect, E-Scape Bio, Teva, AbbVie, Boehringer Ingelheim, and GSK.

Peter Nestor has nothing to disclose.

Wolfgang H Oertel has served as a consultant for Novartis, Schwarz Pharma and Neuroscience /UCB; has served on the advisory boards for Merck, Sharp & Dohme, Medtronic, Mundipharma, Novartis, Schwabe Pharma, Schwarz Pharma, Neuroscience /UCB and Teva; has owned stocks of Roche 100 and Medigene 2400; has received speaker and travel honoraria from AbbVie, Desitin, GlaxoSmithKline, Mundipharma, Novartis, Schwarz Pharma Neuroscience/UCB and Teva; has received research support from the German Ministry of Education and Health, IPF and MJFF.

Gil Rabinovici is supported by National Institutes of Health grants R01-AG045611, P50-AG23501, U54-NS092089, 2R01-AG038791, R01-AG048234, the American College of Radiology, the Association for Frontotemporal Degeneration, Michael J. Fox Foundation, Alzheimer's Association. FINANCIAL DISCLOSURES: Dr. Rabinovici received research support from Avid Radiopharmaceiticals, GE Healthcare and Piramal, and has received speaking honoraria or consulting fees from Eisai, Genentech, Lundbeck, Merck, Puntam, Roche.

James Rowe is supported by the Wellcome Trust (103838) and has received additional research grant support from AZ-Medimmune and Janssen, PSP Association, Medical Research Council, Wellcome Trust, National Institute for Health Research, McDonnell Foundation, Alzheimer Research UK, and Evelyn Trust; advised Asceneuron; and serves as Associate Editor at Brain.

Thilo van Eimeren is supported by grants of the Deutsche Forschungsgemeinschaft (DFG, EI 892/3-1), the EU Joint Programme – Neurodegenerative Disease Research (JPND, Innofond 6177-00001B) and the Leibniz Association (SAW-2013-IfW-2) and received speaking honoraria and consulting fees from Eli Lily, Shire and the CHDI Foundation.

Gregor K Wenning receives consulting and/or lecture fees from Affiris, Astra Zeneca, Boehringer Ingelheim, Ever Pharma, Lundbeck, Neuropore, Orion and UCB as well as grant support from Medical University Innsbruck, Oesterreichische National bank, FWF Austrian Science Fund, US MSA Coalition, Affiris, Astra Zeneca and Boehringer Ingelheim.

Adam Boxer is supported by National Institutes of Health grants U54NS092089, R01AG038791, U01AG045390, U01AG052943-041, U19AG010483, the University of California, the Bluefield Project to Cure FTD and the Tau Research Consortium; received research support from Avid, Biogen, Bristol Myers Squibb, C2N Diagnostics, Cortice Biosciences, Eli Lilly, Forum Pharmaceuticals, Genentech, Roche and TauRx; has served as a consultant for Abbvie, Asceneuron, Ipierian, Isis Pharmaceuticals, Janssen, Merck and Novartis; serves on a Data and Safety Monitoring Board for Neurogenetics Pharmaceuticals; has stock and/or options in Alector and Delos.

Lawrence I Golbe is supported by research funding from Bristol-Myers Squibb, AbbVie and the American Parkinson's Disease Association and consults for Bristol-Myers Squibb, AbbVie, SJO Research and the University of California.

Irene Litvan has served as an advisor for Pfizer and Biotie; received grants from Parkinson Study Group, Michael J Fox Foundation, CBD Solutions-CurePSP, AVID Pharmaceuticals, C2N Diagnostics and Bristol-Myers Squibb; receives her salary from the University of California San Diego.

Maria Stamelou has served on the editorial board of Movement Disorders Journal and Frontiers in Movement Disorders; has received speaker and travel honoraria from Actelion and Abbvie Pharmaceuticals.

Günter U. Höglinger has served on the advisory boards for AbbVie, Alzprotect, Asceneuron, Bristol-Myers Squibb, Novartis, Roche, Sellas Life Sciences Group, UCB; has received honoraria for scientific presentations from Abbvie, Roche, Teva, UCB, has received research support from CurePSP, the German Academic Exchange Service (DAAD), German Research Foundation (DFG) and the German Ministry of Education and Research (BMBF), the Sellas Life Sciences Group; has received institutional support from the German Center for Neurodegenerative Diseases (DZNE).

Appendix: The MDS endorsed PSP study group

Adam L Boxer, Alex Rajput, Alexander Pantelyat, Angelo Antonini, Anthony E Lang, Armin Giese, Brit Mollenhauer, Carlo Colosimo, Caroline Kurz, Christer Nilsson, Claire Troakes, David J Irwin, Dennis W. Dickson, Ellen Gelpi, Florian Krismer, Gerard D Schellenberg, Gesine Respondek, Gil Rabinovici, Gregor K Wenning, Günter U Höglinger, Huw R Morris, Irene Litvan, James B Rowe, Jan Kassubek, Jean-Christophe Corvol, Jennifer L Whitwell, Johannes Levin, John van Swieten, Kailash P Bhatia, Keith A Josephs, Klaus Seppi, Lawrence I Golbe, Maria Stamelou, Murray Grossman, Peter Nestor, Richard Dodel, Stefan Lorenzl, Thilo van Eimeren, Thomas Arzberger, Ulrich Müller, Wassilios G Meissner, Werner Poewe, Wolfgang H Oertel, Yaroslau Compta, Yvette Bordelon.

Footnotes

Financial Disclosure/ Conflict of Interest concerning the research related to the manuscript: none

References

  • 1.Steele JC, Richardson JC, Olszewski J. Progressive Supranuclear Palsy. A Heterogeneous Degeneration Involving the Brain Stem, Basal Ganglia and Cerebellum with Vertical Gaze and Pseudobulbar Palsy, Nuchal Dystonia and Dementia. Arch Neurol. 1964;10:333–359. doi: 10.1001/archneur.1964.00460160003001. [DOI] [PubMed] [Google Scholar]
  • 2.Coyle-Gilchrist IT, Dick KM, Patterson K, et al. Prevalence, characteristics, and survival of frontotemporal lobar degeneration syndromes. Neurology. 2016;86(18):1736–1743. doi: 10.1212/WNL.0000000000002638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schrag A, Ben-Shlomo Y, Quinn NP. Prevalence of progressive supranuclear palsy and multiple system atrophy: a cross-sectional study. Lancet. 1999;354(9192):1771–1775. doi: 10.1016/s0140-6736(99)04137-9. [DOI] [PubMed] [Google Scholar]
  • 4.Respondek G, Stamelou M, Kurz C, et al. The phenotypic spectrum of progressive supranuclear palsy: a retrospective multicenter study of 100 definite cases. Mov Disord. 2014;29(14):1758–1766. doi: 10.1002/mds.26054. [DOI] [PubMed] [Google Scholar]
  • 5.Dickson DW. Neuropathologic differentiation of progressive supranuclear palsy and corticobasal degeneration. J Neurol. 1999;246(2):II6–15. doi: 10.1007/BF03161076. [DOI] [PubMed] [Google Scholar]
  • 6.Kovacs GG. Invited review: Neuropathology of tauopathies: principles and practice. Neuropathol Appl Neurobiol. 2015;41(1):3–23. doi: 10.1111/nan.12208. [DOI] [PubMed] [Google Scholar]
  • 7.Litvan I, Agid Y, Calne D, et al. Clinical research criteria for the diagnosis of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome): report of the NINDS-SPSP international workshop. Neurology. 1996;47(1):1–9. doi: 10.1212/wnl.47.1.1. [DOI] [PubMed] [Google Scholar]
  • 8.Litvan I, Hauw JJ, Bartko JJ, et al. Validity and reliability of the preliminary NINDS neuropathologic criteria for progressive supranuclear palsy and related disorders. J Neuropathol Exp Neurol. 1996;55(1):97–105. doi: 10.1097/00005072-199601000-00010. [DOI] [PubMed] [Google Scholar]
  • 9.Respondek G, Roeber S, Kretzschmar H, et al. Accuracy of the National Institute for Neurological Disorders and Stroke/Society for Progressive Supranuclear Palsy and neuroprotection and natural history in Parkinson plus syndromes criteria for the diagnosis of progressive supranuclear palsy. Mov Disord. 2013;28(4):504–509. doi: 10.1002/mds.25327. [DOI] [PubMed] [Google Scholar]
  • 10.Birdi S, Rajput AH, Fenton M, et al. Progressive supranuclear palsy diagnosis and confounding features: report on 16 autopsied cases. Mov Disord. 2002;17(6):1255–1264. doi: 10.1002/mds.10211. [DOI] [PubMed] [Google Scholar]
  • 11.Osaki Y, Ben-Shlomo Y, Lees AJ, et al. Accuracy of clinical diagnosis of progressive supranuclear palsy. Mov Disord. 2004;19(2):181–189. doi: 10.1002/mds.10680. [DOI] [PubMed] [Google Scholar]
  • 12.Sakamoto R, Tsuchiya K, Mimura M. Clinical heterogeneity in progressive supranuclear palsy: problems of clinical diagnostic criteria of NINDS-SPSP in a retrospective study of seven Japanese autopsy cases. Neuropathology. 2010;30(1):24–35. doi: 10.1111/j.1440-1789.2009.01032.x. [DOI] [PubMed] [Google Scholar]
  • 13.Ahmed Z, Josephs KA, Gonzalez J, DelleDonne A, Dickson DW. Clinical and neuropathologic features of progressive supranuclear palsy with severe pallido-nigro-luysial degeneration and axonal dystrophy. Brain. 2008;131(Pt 2):460–472. doi: 10.1093/brain/awm301. [DOI] [PubMed] [Google Scholar]
  • 14.Boeve BF, Maraganore DM, Parisi JE, et al. Pathologic heterogeneity in clinically diagnosed corticobasal degeneration. Neurology. 1999;53(4):795–800. doi: 10.1212/wnl.53.4.795. [DOI] [PubMed] [Google Scholar]
  • 15.Boeve B, Dickson D, Duffy J, Bartleson J, Trenerry M, Petersen R. Progressive nonfluent aphasia and subsequent aphasic dementia associated with atypical progressive supranuclear palsy pathology. Eur Neurol. 2003;49(2):72–78. doi: 10.1159/000068502. [DOI] [PubMed] [Google Scholar]
  • 16.Compta Y, Valldeoriola F, Tolosa E, Rey MJ, Marti MJ, Valls-Sole J. Long lasting pure freezing of gait preceding progressive supranuclear palsy: a clinicopathological study. Mov Disord. 2007;22(13):1954–1958. doi: 10.1002/mds.21612. [DOI] [PubMed] [Google Scholar]
  • 17.Deramecourt V, Lebert F, Debachy B, et al. Prediction of pathology in primary progressive language and speech disorders. Neurology. 2010;74(1):42–49. doi: 10.1212/WNL.0b013e3181c7198e. [DOI] [PubMed] [Google Scholar]
  • 18.Donker Kaat L, Boon AJ, Kamphorst W, Ravid R, Duivenvoorden HJ, van Swieten JC. Frontal presentation in progressive supranuclear palsy. Neurology. 2007;69(8):723–729. doi: 10.1212/01.wnl.0000267643.24870.26. [DOI] [PubMed] [Google Scholar]
  • 19.Facheris MF, Maniak S, Scaravilli F, Schule B, Klein C, Pramstaller PP. Pure akinesia as initial presentation of PSP: a clinicopathological study. Parkinsonism Relat Disord. 2008;14(6):517–519. doi: 10.1016/j.parkreldis.2007.11.004. [DOI] [PubMed] [Google Scholar]
  • 20.Ferrer I, Hernandez I, Boada M, et al. Primary progressive aphasia as the initial manifestation of corticobasal degeneration and unusual tauopathies. Acta Neuropathol. 2003;106(5):419–435. doi: 10.1007/s00401-003-0756-4. [DOI] [PubMed] [Google Scholar]
  • 21.Josephs KA, Katsuse O, Beccano-Kelly DA, et al. Atypical progressive supranuclear palsy with corticospinal tract degeneration. J Neuropathol Exp Neurol. 2006;65(4):396–405. doi: 10.1097/01.jnen.0000218446.38158.61. [DOI] [PubMed] [Google Scholar]
  • 22.Josephs KA, Duffy JR. Apraxia of speech and nonfluent aphasia: a new clinical marker for corticobasal degeneration and progressive supranuclear palsy. Curr Opin Neurol. 2008;21(6):688–692. doi: 10.1097/WCO.0b013e3283168ddd. [DOI] [PubMed] [Google Scholar]
  • 23.Han HJ, Kim H, Park JH, et al. Behavioral changes as the earliest clinical manifestation of progressive supranuclear palsy. J Clin Neurol. 2010;6(3):148–151. doi: 10.3988/jcn.2010.6.3.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hassan A, Parisi JE, Josephs KA. Autopsy-proven progressive supranuclear palsy presenting as behavioral variant frontotemporal dementia. Neurocase. 2012;18(6):478–488. doi: 10.1080/13554794.2011.627345. [DOI] [PubMed] [Google Scholar]
  • 25.Karnik NS, D'Apuzzo M, Greicius M. Non-fluent progressive aphasia, depression, and OCD in a woman with progressive supranuclear palsy: neuroanatomical and neuropathological correlations. Neurocase. 2006;12(6):332–338. doi: 10.1080/13554790601125957. [DOI] [PubMed] [Google Scholar]
  • 26.Ling H, O'Sullivan SS, Holton JL, et al. Does corticobasal degeneration exist? A clinicopathological re-evaluation. Brain. 2010;133(Pt 7):2045–2057. doi: 10.1093/brain/awq123. [DOI] [PubMed] [Google Scholar]
  • 27.Ling H, de Silva R, Massey LA, et al. Characteristics of progressive supranuclear palsy presenting with corticobasal syndrome: a cortical variant. Neuropathol Appl Neurobiol. 2014;40(2):149–163. doi: 10.1111/nan.12037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Litvan I, Mangone CA, McKee A, et al. Natural history of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome) and clinical predictors of survival: a clinicopathological study. J Neurol Neurosurg Psychiatry. 1996;60(6):615–620. doi: 10.1136/jnnp.60.6.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Magdalinou NK, Ling H, Smith JD, Schott JM, Watkins LD, Lees AJ. Normal pressure hydrocephalus or progressive supranuclear palsy? A clinicopathological case series. J Neurol. 2013;260(4):1009–1013. doi: 10.1007/s00415-012-6745-6. [DOI] [PubMed] [Google Scholar]
  • 30.Mizuno T, Shiga K, Nakata Y, et al. Discrepancy between clinical and pathological diagnoses of CBD and PSP. J Neurol. 2005;252(6):687–697. doi: 10.1007/s00415-005-0718-y. [DOI] [PubMed] [Google Scholar]
  • 31.Mochizuki A, Ueda Y, Komatsuzaki Y, Tsuchiya K, Arai T, Shoji S. Progressive supranuclear palsy presenting with primary progressive aphasia--clinicopathological report of an autopsy case. Acta Neuropathol. 2003;105(6):610–614. doi: 10.1007/s00401-003-0682-5. [DOI] [PubMed] [Google Scholar]
  • 32.Nagao S, Yokota O, Nanba R, et al. Progressive supranuclear palsy presenting as primary lateral sclerosis but lacking parkinsonism, gaze palsy, aphasia, or dementia. J Neurol Sci. 2012;323(1-2):147–153. doi: 10.1016/j.jns.2012.09.005. [DOI] [PubMed] [Google Scholar]
  • 33.Williams DR, de Silva R, Paviour DC, et al. Characteristics of two distinct clinical phenotypes in pathologically proven progressive supranuclear palsy: Richardson's syndrome and PSP-parkinsonism. Brain. 2005;128(Pt 6):1247–1258. doi: 10.1093/brain/awh488. [DOI] [PubMed] [Google Scholar]
  • 34.Williams DR, Holton JL, Strand K, Revesz T, Lees AJ. Pure akinesia with gait freezing: a third clinical phenotype of progressive supranuclear palsy. Mov Disord. 2007;22(15):2235–2241. doi: 10.1002/mds.21698. [DOI] [PubMed] [Google Scholar]
  • 35.Hoglinger GU, Huppertz HJ, Wagenpfeil S, et al. Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Mov Disord. 2014;29(4):479–487. doi: 10.1002/mds.25815. [DOI] [PubMed] [Google Scholar]
  • 36.Stamelou M, de Silva R, Arias-Carrion O, et al. Rational therapeutic approaches to progressive supranuclear palsy. Brain. 2010;133(Pt 6):1578–1590. doi: 10.1093/brain/awq115. [DOI] [PubMed] [Google Scholar]
  • 37.SIGN 50: a guideline developers' handbook. Edinburgh: Scottish Intercollegiate Guidelines Network; 2008. [Google Scholar]
  • 38.Hauw JJ, Daniel SE, Dickson D, et al. Preliminary NINDS neuropathologic criteria for Steele-Richardson-Olszewski syndrome (progressive supranuclear palsy) Neurology. 1994;44(11):2015–2019. doi: 10.1212/wnl.44.11.2015. [DOI] [PubMed] [Google Scholar]
  • 39.Dickson DW, Bergeron C, Chin SS, et al. Office of Rare Diseases neuropathologic criteria for corticobasal degeneration. J Neuropathol Exp Neurol. 2002;61(11):935–946. doi: 10.1093/jnen/61.11.935. [DOI] [PubMed] [Google Scholar]
  • 40.Trojanowski JQ, Revesz T Neuropathology Working Group on MSA. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol Appl Neurobiol. 2007;33(6):615–620. doi: 10.1111/j.1365-2990.2007.00907.x. [DOI] [PubMed] [Google Scholar]
  • 41.Dickson DW, Braak H, Duda JE, et al. Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria. Lancet Neurol. 2009;8(12):1150–1157. doi: 10.1016/S1474-4422(09)70238-8. [DOI] [PubMed] [Google Scholar]
  • 42.Mackenzie IR, Neumann M, Bigio EH, et al. Nomenclature and nosology for neuropathologic subtypes of frontotemporal lobar degeneration: an update. Acta Neuropathol. 2010;119(1):1–4. doi: 10.1007/s00401-009-0612-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Neary D, Snowden JS, Gustafson L, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998;51(6):1546–1554. doi: 10.1212/wnl.51.6.1546. [DOI] [PubMed] [Google Scholar]
  • 44.Whitwell Radiological biomarkers for diagnosis in PSP: Where are we and where do we need to be? Mov Disord submitted. doi: 10.1002/mds.27038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lopez OL, Litvan I, Catt KE, et al. Accuracy of four clinical diagnostic criteria for the diagnosis of neurodegenerative dementias. Neurology. 1999;53(6):1292–1299. doi: 10.1212/wnl.53.6.1292. [DOI] [PubMed] [Google Scholar]
  • 46.Kurz C, Ebersbach G, Respondek G, Giese A, Arzberger T, Hoglinger GU. An autopsy-confirmed case of progressive supranuclear palsy with predominant postural instability. Acta Neuropathol Commun. 2016;4(1):120. doi: 10.1186/s40478-016-0391-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Factor SA, Higgins DS, Qian J. Primary progressive freezing gait: a syndrome with many causes. Neurology. 2006;66(3):411–414. doi: 10.1212/01.wnl.0000196469.52995.ab. [DOI] [PubMed] [Google Scholar]
  • 48.Josephs KA, Duffy JR, Strand EA, et al. Clinicopathological and imaging correlates of progressive aphasia and apraxia of speech. Brain. 2006;129(Pt 6):1385–1398. doi: 10.1093/brain/awl078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tsuboi Y, Josephs KA, Boeve BF, et al. Increased tau burden in the cortices of progressive supranuclear palsy presenting with corticobasal syndrome. Mov Disord. 2005;20(8):982–988. doi: 10.1002/mds.20478. [DOI] [PubMed] [Google Scholar]
  • 50.Iwasaki Y, Mori K, Ito M, Tatsumi S, Mimuro M, Yoshida M. An autopsied case of progressive supranuclear palsy presenting with cerebellar ataxia and severe cerebellar involvement. Neuropathology. 2013;33(5):561–567. doi: 10.1111/neup.12012. [DOI] [PubMed] [Google Scholar]
  • 51.Kanazawa M, Shimohata T, Toyoshima Y, et al. Cerebellar involvement in progressive supranuclear palsy: A clinicopathological study. Mov Disord. 2009;24(9):1312–1318. doi: 10.1002/mds.22583. [DOI] [PubMed] [Google Scholar]
  • 52.Aarsland D, Litvan I, Larsen JP. Neuropsychiatric symptoms of patients with progressive supranuclear palsy and Parkinson's disease. J Neuropsychiatry Clin Neurosci. 2001;13(1):42–49. doi: 10.1176/jnp.13.1.42. [DOI] [PubMed] [Google Scholar]
  • 53.Borroni B, Alberici A, Agosti C, Cosseddu M, Padovani A. Pattern of behavioral disturbances in corticobasal degeneration syndrome and progressive supranuclear palsy. Int Psychogeriatr. 2009;21(3):463–468. doi: 10.1017/S1041610209008862. [DOI] [PubMed] [Google Scholar]
  • 54.Gerstenecker A, Duff K, Mast B, Litvan I Group E-PS. Behavioral abnormalities in progressive supranuclear palsy. Psychiatry Res. 2013;210(3):1205–1210. doi: 10.1016/j.psychres.2013.08.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Litvan I, Paulsen JS, Mega MS, Cummings JL. Neuropsychiatric assessment of patients with hyperkinetic and hypokinetic movement disorders. Arch Neurol. 1998;55(10):1313–1319. doi: 10.1001/archneur.55.10.1313. [DOI] [PubMed] [Google Scholar]
  • 56.Rittman T, Ghosh BC, McColgan P, et al. The Addenbrooke's Cognitive Examination for the differential diagnosis and longitudinal assessment of patients with parkinsonian disorders. J Neurol Neurosurg Psychiatry. 2013;84(5):544–551. doi: 10.1136/jnnp-2012-303618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Boxer AL, Garbutt S, Seeley WW, et al. Saccade abnormalities in autopsy-confirmed frontotemporal lobar degeneration and Alzheimer disease. Arch Neurol. 2012;69(4):509–517. doi: 10.1001/archneurol.2011.1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Litvan I, Campbell G, Mangone CA, et al. Which clinical features differentiate progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome) from related disorders? A clinicopathological study. Brain. 1997;120(Pt 1):65–74. doi: 10.1093/brain/120.1.65. [DOI] [PubMed] [Google Scholar]
  • 59.dell'Aquila C, Zoccolella S, Cardinali V, et al. Predictors of survival in a series of clinically diagnosed progressive supranuclear palsy patients. Parkinsonism Relat Disord. 2013;19(11):980–985. doi: 10.1016/j.parkreldis.2013.06.014. [DOI] [PubMed] [Google Scholar]
  • 60.Nath U, Ben-Shlomo Y, Thomson RG, Lees AJ, Burn DJ. Clinical features and natural history of progressive supranuclear palsy: a clinical cohort study. Neurology. 2003;60(6):910–916. doi: 10.1212/01.wnl.0000052991.70149.68. [DOI] [PubMed] [Google Scholar]
  • 61.Verny M, Jellinger KA, Hauw JJ, Bancher C, Litvan I, Agid Y. Progressive supranuclear palsy: a clinicopathological study of 21 cases. Acta Neuropathol. 1996;91(4):427–431. doi: 10.1007/s004010050446. [DOI] [PubMed] [Google Scholar]
  • 62.Otero-Millan J, Serra A, Leigh RJ, Troncoso XG, Macknik SL, Martinez-Conde S. Distinctive features of saccadic intrusions and microsaccades in progressive supranuclear palsy. J Neurosci. 2011;31(12):4379–4387. doi: 10.1523/JNEUROSCI.2600-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cooper AD, Josephs KA. Photophobia, visual hallucinations, and REM sleep behavior disorder in progressive supranuclear palsy and corticobasal degeneration: a prospective study. Parkinsonism Relat Disord. 2009;15(1):59–61. doi: 10.1016/j.parkreldis.2008.01.011. [DOI] [PubMed] [Google Scholar]
  • 64.Rana AQ, Kabir A, Dogu O, Patel A, Khondker S. Prevalence of blepharospasm and apraxia of eyelid opening in patients with parkinsonism, cervical dystonia and essential tremor. Eur Neurol. 2012;68(5):318–321. doi: 10.1159/000341621. [DOI] [PubMed] [Google Scholar]
  • 65.Bologna M, Agostino R, Gregori B, et al. Voluntary, spontaneous and reflex blinking in patients with clinically probable progressive supranuclear palsy. Brain. 2009;132(Pt 2):502–510. doi: 10.1093/brain/awn317. [DOI] [PubMed] [Google Scholar]
  • 66.Borroni B, Goldwurm S, Cerini C, et al. Familial aggregation in Progressive Supranuclear Palsy and Corticobasal Syndrome. Eur J Neurol. 2011;18(1):195–197. doi: 10.1111/j.1468-1331.2010.03081.x. [DOI] [PubMed] [Google Scholar]
  • 67.Donker Kaat L, Boon AJ, Azmani A, et al. Familial aggregation of parkinsonism in progressive supranuclear palsy. Neurology. 2009;73(2):98–105. doi: 10.1212/WNL.0b013e3181a92bcc. [DOI] [PubMed] [Google Scholar]
  • 68.Hoglinger GU, Melhem NM, Dickson DW, et al. Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet. 2011;43(7):699–705. doi: 10.1038/ng.859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pastor P, Pastor E, Carnero C, et al. Familial atypical progressive supranuclear palsy associated with homozigosity for the delN296 mutation in the tau gene. Ann Neurol. 2001;49(2):263–267. doi: 10.1002/1531-8249(20010201)49:2<263::aid-ana50>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 70.Ros R, Thobois S, Streichenberger N, et al. A new mutation of the tau gene, G303V, in early-onset familial progressive supranuclear palsy. Arch Neurol. 2005;62(9):1444–1450. doi: 10.1001/archneur.62.9.1444. [DOI] [PubMed] [Google Scholar]
  • 71.Hall S, Ohrfelt A, Constantinescu R, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol. 2012;69(11):1445–1452. doi: 10.1001/archneurol.2012.1654. [DOI] [PubMed] [Google Scholar]
  • 72.Elbaz A, Bower JH, Peterson BJ, et al. Survival study of Parkinson disease in Olmsted County, Minnesota. Arch Neurol. 2003;60(1):91–96. doi: 10.1001/archneur.60.1.91. [DOI] [PubMed] [Google Scholar]
  • 73.Kansal K, Mareddy M, Sloane KL, et al. Survival in Frontotemporal Dementia Phenotypes: A Meta-Analysis. Dement Geriatr Cogn Disord. 2016;41(1-2):109–122. doi: 10.1159/000443205. [DOI] [PubMed] [Google Scholar]
  • 74.Wenning GK, Ben Shlomo Y, Magalhaes M, Daniel SE, Quinn NP. Clinical features and natural history of multiple system atrophy. An analysis of 100 cases. Brain. 1994;117(Pt 4):835–845. doi: 10.1093/brain/117.4.835. [DOI] [PubMed] [Google Scholar]
  • 75.Wenning GK, Litvan I, Jankovic J, et al. Natural history and survival of 14 patients with corticobasal degeneration confirmed at postmortem examination. J Neurol Neurosurg Psychiatry. 1998;64(2):184–189. doi: 10.1136/jnnp.64.2.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Armstrong MJ, Litvan I, Lang AE, et al. Criteria for the diagnosis of corticobasal degeneration. Neurology. 2013;80(5):496–503. doi: 10.1212/WNL.0b013e31827f0fd1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer's disease: the IWG-2 criteria. Lancet Neurol. 2014;13(6):614–629. doi: 10.1016/S1474-4422(14)70090-0. [DOI] [PubMed] [Google Scholar]
  • 78.Kovacs GG, Milenkovic I, Wohrer A, et al. Non-Alzheimer neurodegenerative pathologies and their combinations are more frequent than commonly believed in the elderly brain: a community-based autopsy series. Acta Neuropathol. 2013;126(3):365–384. doi: 10.1007/s00401-013-1157-y. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp info

RESOURCES