1
|
de Boer L, Poos JM, Van Den Berg E, De Houwer JFH, Swartenbroekx T, Dopper EGP, Boesjes P, Tahboun N, Bouzigues A, Foster PH, Ferry-Bolder E, Adams-Carr K, Russell LL, Convery RS, Rohrer JD, Seelaar H, Jiskoot LC. Montreal Cognitive Assessment vs the Mini-Mental State Examination as a Screening Tool for Patients With Genetic Frontotemporal Dementia. Neurology 2025; 104:e213401. [PMID: 39951678 PMCID: PMC11837847 DOI: 10.1212/wnl.0000000000213401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND AND OBJECTIVES With upcoming clinical trials targeting preclinical stages of genetic frontotemporal dementia (FTD), early detection through cognitive screening is crucial. The Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) have potential as screening instruments for early-stage genetic FTD. However, no comparative evaluation has been performed. We aimed to compare MMSE and MoCA performance among presymptomatic, prodromal, and symptomatic pathogenic variant carriers to analyze which screening test has superior discriminative abilities. METHODS We used cross-sectional and longitudinal data from 2 longitudinal genetic FTD cohort studies in the Netherlands and the United Kingdom, collected between 2021 and 2024. Participants were either presymptomatic, prodromal, or symptomatic pathogenic variant carriers or healthy controls (first-degree family members without pathogenic variants for FTD). Grouping was based on the global CDR-plus-NACC-FTLD score. Participants were assessed with both MoCA and MMSE. Statistical analyses compared total and subscores between groups and evaluated predictive and classification accuracy of both tests. RESULTS A total of 243 participants (mean age 49.9 ± 13.1 years, mean education 14.5 ± 3.0 years, 56% female), 157 of whom were pathogenic variant carriers (MAPT, GRN, C9orf72, TARDBP, and TBK1) and 86 controls, were included. Carriers were classified as presymptomatic (n = 119), prodromal (n = 18), or symptomatic (n = 20). Both MoCA [F(3,239) = 16.565, p < 0.001] and MMSE [F(3,239) = 13.529, p < 0.001] total scores differed significantly between groups, with controls (median MoCA 28.5, 95% CI 28.0-29.0; median MMSE 30, 95% CI 30.0-30.0) outperforming prodromal (median MoCA 26, 95% CI 23.0-27.0; median MMSE 29, 95% CI 27.5-29.5) and symptomatic (median MoCA 20.5, 95% CI 17.0-24.0; median MMSE 26, 95% CI 23.5-29.0) carriers. MoCA distinguished between presymptomatic carriers and controls (median MoCA 28, 95% CI 27.0-29.0), but MMSE did not. MoCA demonstrated superior discriminative ability compared with MMSE (MoCA area under the curve [AUC] = 0.87, 95% CI 0.81-0.94; MMSE AUC = 0.80, 95% CI 0.72-0.89). DISCUSSION Its higher sensitivity and better discriminative power make MoCA a more valuable tool for cognitive screening in upcoming clinical trials targeting preclinical FTD. Future studies should aim for larger sample sizes from additional study centers.
Collapse
Affiliation(s)
- Liset de Boer
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Jackie M Poos
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Esther Van Den Berg
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Julie F H De Houwer
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Tine Swartenbroekx
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Elise G P Dopper
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Pam Boesjes
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Najlae Tahboun
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | | | - Phoebe H Foster
- Dementia Research Centre, University College London, United Kingdom
| | - Eve Ferry-Bolder
- Dementia Research Centre, University College London, United Kingdom
| | | | - Lucy L Russell
- Dementia Research Centre, University College London, United Kingdom
| | - Rhian S Convery
- Dementia Research Centre, University College London, United Kingdom
| | | | - Harro Seelaar
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Centre, Rotterdam, the Netherlands; and
| |
Collapse
|
2
|
Nóbrega PR, Paiva ARB, Amorim Junior AD, Lima PLGSB, Cabral KSS, Barcelos IP, Pessoa ALS, Souza-Lima CFL, Castro MAA, Freua F, Santos EDS, Rocha MMV, Maia RE, Araújo RS, Ramos JDG, Resende RG, Carvalho GDS, Valença LPA, Lima de Carvalho JR, Melo ES, Pedroso JL, Barsottini OGP, Houlden H, Kok F, Lynch DS. Further description of the phenotypic spectrum of neuronal ceroid lipofuscinosis type 11. Genet Med 2025; 27:101291. [PMID: 39394881 DOI: 10.1016/j.gim.2024.101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
PURPOSE Ceroid lipofuscinosis type 11 (CLN11) is a very rare disease, being reported in only 13 unrelated families so far. Further reports are necessary to comprehend the clinical phenotype of this condition. This article aims to report 9 additional cases of CLN11 from 9 unrelated Latin American families presenting with relatively slow disease progression. METHODS This was a retrospective observational study including patients with CLN11. Patients were identified through an active search for granulin precursor gene (GRN) pathogenic variants across the entire database of next-generation sequencing of a commercial laboratory and by contacting attending physicians to check for clinical and radiologic findings compatible with a neuronal ceroid lipofuscinosis phenotype. RESULTS Nine CLN11 patients from unrelated families were evaluated. Age of onset varied between 3 to 17 years. The most common findings were visual impairment, cerebellar ataxia, seizures, myoclonus, and cognitive decline. One patient had a previously unreported finding of cervical, perioral, and tongue myoclonus. Most of the patients were able to walk unassisted after an average of 14.2 years (SD 4.76 y) from disease onset. CONCLUSION We describe 9 new cases of a very rare type of neuronal ceroid lipofuscinosis (CLN11) from Latin America with a recurrent p.(Gln257ProfsTer27) and a novel p.(Cys83Ter) nonsense variant. Our findings suggest that a slowly progressive neuronal ceroid lipofuscinosis might be a clue for the diagnosis of CLN11.
Collapse
Affiliation(s)
- Paulo Ribeiro Nóbrega
- Division of Neurology, Universidade Federal do Ceara, Fortaleza, CE, Brazil; Centro Universitário Christus, Fortaleza, CE, Brazil
| | - Anderson Rodrigues Brandão Paiva
- Neurogenetics Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Mendelics Genomic Analysis, São Paulo, SP, Brazil
| | - Antonio Duarte Amorim Junior
- Neurology Department, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Katiane Sayão Souza Cabral
- Neurology Department, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Isabella Peixoto Barcelos
- Neurology Department, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - André Luis Santos Pessoa
- Hospital Infantil Albert Sabin, Fortaleza, CE, Brazil; Universidade Estadual do Ceara, Fortaleza, CE, Brazil
| | | | - Matheus Augusto Araújo Castro
- Neurogenetics Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Mendelics Genomic Analysis, São Paulo, SP, Brazil
| | - Fernando Freua
- Neurogenetics Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Rayana Elias Maia
- Departamento de Pediatria e Genética, Universidade Federal da Paraíba, João Pessoa, PB, Brazil
| | - Rodrigo Santos Araújo
- Hospital Universitário de Lagarto, Universidade Federal de Sergipe, Aracaju, SE, Brazil; Hospital de Urgência de Sergipe, Aracaju, SE, Brazil
| | | | | | - Gerson da Silva Carvalho
- Centro de Referência em Doenças Raras do Distrito Federal, Hospital de Apoio de Brasília, Brasília, DF, Brazil
| | | | | | - Eduardo Sousa Melo
- Centro de Ciências Médicas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - José Luiz Pedroso
- Universidade Federal de São Paulo, Departamento de Neurologia, São Paulo, SP, Brazil
| | | | - Henry Houlden
- Dept of Neuromuscular Disease, UCL Institute of Neurology, Queen Sq, London; National Hospital for Neurology and Neurosurgery, Queen Sq, London
| | - Fernando Kok
- Neurogenetics Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Mendelics Genomic Analysis, São Paulo, SP, Brazil
| | - David S Lynch
- Dept of Neuromuscular Disease, UCL Institute of Neurology, Queen Sq, London; National Hospital for Neurology and Neurosurgery, Queen Sq, London; National Institute for Health and Care Research (NIHR) and University College London Hospitals, Biomedical Research Centre, London.
| |
Collapse
|
3
|
Sevigny J, Uspenskaya O, Heckman LD, Wong LC, Hatch DA, Tewari A, Vandenberghe R, Irwin DJ, Saracino D, Le Ber I, Ahmed R, Rohrer JD, Boxer AL, Boland S, Sheehan P, Brandes A, Burstein SR, Shykind BM, Kamalakaran S, Daniels CW, David Litwack E, Mahoney E, Velaga J, McNamara I, Sondergaard P, Sajjad SA, Kobayashi YM, Abeliovich A, Hefti F. Progranulin AAV gene therapy for frontotemporal dementia: translational studies and phase 1/2 trial interim results. Nat Med 2024; 30:1406-1415. [PMID: 38745011 PMCID: PMC11108785 DOI: 10.1038/s41591-024-02973-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .
Collapse
Affiliation(s)
- Jeffrey Sevigny
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA.
| | - Olga Uspenskaya
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Laura Dean Heckman
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Li Chin Wong
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Daniel A Hatch
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ambika Tewari
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Leuven, Belgium and Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Rebekah Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Center, UCL Queen Square Institute of Neurology, London, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sebastian Boland
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sheehan
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Alissa Brandes
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Suzanne R Burstein
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Benjamin M Shykind
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Sitharthan Kamalakaran
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Carter W Daniels
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - E David Litwack
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Erin Mahoney
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jenny Velaga
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ilan McNamara
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sondergaard
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Syed A Sajjad
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Yvonne M Kobayashi
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Asa Abeliovich
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
4
|
Staffaroni AM, Clark AL, Taylor JC, Heuer HW, Sanderson-Cimino M, Wise AB, Dhanam S, Cobigo Y, Wolf A, Manoochehri M, Forsberg L, Mester C, Rankin KP, Appleby BS, Bayram E, Bozoki A, Clark D, Darby RR, Domoto-Reilly K, Fields JA, Galasko D, Geschwind D, Ghoshal N, Graff-Radford N, Grossman M, Hsiung GY, Huey ED, Jones DT, Lapid MI, Litvan I, Masdeu JC, Massimo L, Mendez MF, Miyagawa T, Pascual B, Pressman P, Ramanan VK, Ramos EM, Rascovsky K, Roberson ED, Tartaglia MC, Wong B, Miller BL, Kornak J, Kremers W, Hassenstab J, Kramer JH, Boeve BF, Rosen HJ, Boxer AL. Reliability and Validity of Smartphone Cognitive Testing for Frontotemporal Lobar Degeneration. JAMA Netw Open 2024; 7:e244266. [PMID: 38558141 PMCID: PMC10985553 DOI: 10.1001/jamanetworkopen.2024.4266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
Importance Frontotemporal lobar degeneration (FTLD) is relatively rare, behavioral and motor symptoms increase travel burden, and standard neuropsychological tests are not sensitive to early-stage disease. Remote smartphone-based cognitive assessments could mitigate these barriers to trial recruitment and success, but no such tools are validated for FTLD. Objective To evaluate the reliability and validity of smartphone-based cognitive measures for remote FTLD evaluations. Design, Setting, and Participants In this cohort study conducted from January 10, 2019, to July 31, 2023, controls and participants with FTLD performed smartphone application (app)-based executive functioning tasks and an associative memory task 3 times over 2 weeks. Observational research participants were enrolled through 18 centers of a North American FTLD research consortium (ALLFTD) and were asked to complete the tests remotely using their own smartphones. Of 1163 eligible individuals (enrolled in parent studies), 360 were enrolled in the present study; 364 refused and 439 were excluded. Participants were divided into discovery (n = 258) and validation (n = 102) cohorts. Among 329 participants with data available on disease stage, 195 were asymptomatic or had preclinical FTLD (59.3%), 66 had prodromal FTLD (20.1%), and 68 had symptomatic FTLD (20.7%) with a range of clinical syndromes. Exposure Participants completed standard in-clinic measures and remotely administered ALLFTD mobile app (app) smartphone tests. Main Outcomes and Measures Internal consistency, test-retest reliability, association of smartphone tests with criterion standard clinical measures, and diagnostic accuracy. Results In the 360 participants (mean [SD] age, 54.0 [15.4] years; 209 [58.1%] women), smartphone tests showed moderate-to-excellent reliability (intraclass correlation coefficients, 0.77-0.95). Validity was supported by association of smartphones tests with disease severity (r range, 0.38-0.59), criterion-standard neuropsychological tests (r range, 0.40-0.66), and brain volume (standardized β range, 0.34-0.50). Smartphone tests accurately differentiated individuals with dementia from controls (area under the curve [AUC], 0.93 [95% CI, 0.90-0.96]) and were more sensitive to early symptoms (AUC, 0.82 [95% CI, 0.76-0.88]) than the Montreal Cognitive Assessment (AUC, 0.68 [95% CI, 0.59-0.78]) (z of comparison, -2.49 [95% CI, -0.19 to -0.02]; P = .01). Reliability and validity findings were highly similar in the discovery and validation cohorts. Preclinical participants who carried pathogenic variants performed significantly worse than noncarrier family controls on 3 app tasks (eg, 2-back β = -0.49 [95% CI, -0.72 to -0.25]; P < .001) but not a composite of traditional neuropsychological measures (β = -0.14 [95% CI, -0.42 to 0.14]; P = .32). Conclusions and Relevance The findings of this cohort study suggest that smartphones could offer a feasible, reliable, valid, and scalable solution for remote evaluations of FTLD and may improve early detection. Smartphone assessments should be considered as a complementary approach to traditional in-person trial designs. Future research should validate these results in diverse populations and evaluate the utility of these tests for longitudinal monitoring.
Collapse
Affiliation(s)
- Adam M. Staffaroni
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Annie L. Clark
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Jack C. Taylor
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Hilary W. Heuer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Mark Sanderson-Cimino
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Amy B. Wise
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Sreya Dhanam
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Yann Cobigo
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Amy Wolf
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | | | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Carly Mester
- Department of Quantitative Health Sciences, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Katherine P. Rankin
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Brian S. Appleby
- Department of Neurology, Case Western Reserve University, Cleveland, Ohio
| | - Ece Bayram
- Department of Neurosciences, University of California, San Diego, La Jolla
| | - Andrea Bozoki
- Department of Radiology, University of North Carolina, Chapel Hill
| | - David Clark
- Department of Neurology, Indiana University, Indianapolis
| | - R. Ryan Darby
- Department of Neurology, Vanderbilt University, Nashville, Tennessee
| | | | - Julie A. Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla
| | - Daniel Geschwind
- Department of Neurology, Institute for Precision Health, University of California, Los Angeles
| | - Nupur Ghoshal
- Department of Neurology, Knight Alzheimer Disease Research Center, Washington University, Saint Louis, Missouri
- Department of Psychiatry, Knight Alzheimer Disease Research Center, Washington University, Saint Louis, Missouri
| | | | - Murray Grossman
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Ging-Yuek Hsiung
- Division of Neurology, University of British Columbia, Musqueam, Squamish & Tsleil-Waututh Traditional Territory, Vancouver, Canada
| | - Edward D. Huey
- Department of Neurology, Columbia University, New York, New York
| | - David T. Jones
- Department of Quantitative Health Sciences, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Maria I. Lapid
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, La Jolla
| | - Joseph C. Masdeu
- Department of Neurology, Nantz National Alzheimer Center, Houston Methodist and Weill Cornell Medicine, Houston Methodist, Houston, Texas
| | - Lauren Massimo
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Mario F. Mendez
- Department of Neurology, UCLA (University of California, Los Angeles)
| | - Toji Miyagawa
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Belen Pascual
- Department of Neurology, Nantz National Alzheimer Center, Houston Methodist and Weill Cornell Medicine, Houston Methodist, Houston, Texas
| | | | | | | | - Katya Rascovsky
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | - M. Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, Division of Neurology, University of Toronto, Toronto, Ontario, Canada
| | - Bonnie Wong
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Bruce L. Miller
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Walter Kremers
- Department of Quantitative Health Sciences, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Jason Hassenstab
- Department of Neurology, Knight Alzheimer Disease Research Center, Washington University, Saint Louis, Missouri
- Department of Psychological & Brain Sciences, Washington University, Saint Louis, Missouri
| | - Joel H. Kramer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | | | - Howard J. Rosen
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| | - Adam L. Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco
| |
Collapse
|
5
|
Benussi A, Premi E, Grassi M, Alberici A, Cantoni V, Gazzina S, Archetti S, Gasparotti R, Fumagalli GG, Bouzigues A, Russell LL, Samra K, Cash DM, Bocchetta M, Todd EG, Convery RS, Swift I, Sogorb-Esteve A, Heller C, van Swieten JC, Jiskoot LC, Seelaar H, Sanchez-Valle R, Moreno F, Laforce RJ, Graff C, Synofzik M, Galimberti D, Rowe JB, Masellis M, Tartaglia MC, Finger E, Vandenberghe R, Mendonça A, Tiraboschi P, Butler CR, Santana I, Gerhard A, Le Ber I, Pasquier F, Ducharme S, Levin J, Sorbi S, Otto M, Padovani A, Rohrer JD, Borroni B. Diagnostic accuracy of research criteria for prodromal frontotemporal dementia. Alzheimers Res Ther 2024; 16:10. [PMID: 38216961 PMCID: PMC10785469 DOI: 10.1186/s13195-024-01383-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND The Genetic Frontotemporal Initiative Staging Group has proposed clinical criteria for the diagnosis of prodromal frontotemporal dementia (FTD), termed mild cognitive and/or behavioral and/or motor impairment (MCBMI). The objective of the study was to validate the proposed research criteria for MCBMI-FTD in a cohort of genetically confirmed FTD cases against healthy controls. METHODS A total of 398 participants were enrolled, 117 of whom were carriers of an FTD pathogenic variant with mild clinical symptoms, while 281 were non-carrier family members (healthy controls (HC)). A subgroup of patients underwent blood neurofilament light (NfL) levels and anterior cingulate atrophy assessment. RESULTS The core clinical criteria correctly classified MCBMI vs HC with an AUC of 0.79 (p < 0.001), while the addition of either blood NfL or anterior cingulate atrophy significantly increased the AUC to 0.84 and 0.82, respectively (p < 0.001). The addition of both markers further increased the AUC to 0.90 (p < 0.001). CONCLUSIONS The proposed MCBMI criteria showed very good classification accuracy for identifying the prodromal stage of FTD.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Enrico Premi
- Vascular Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Mario Grassi
- Department of Brain and Behavioral Science, Medical and Genomic Statistics Unit, University of Pavia, 27100, Pavia, Italy
| | - Antonella Alberici
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Valentina Cantoni
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Stefano Gazzina
- Department of Neurological and Vision Sciences, Neurophysiology Unit, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Silvana Archetti
- Biotechnology Laboratory, Department of Diagnostics, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Roberto Gasparotti
- Department of Medical and Surgical Specialties, Neuroradiology Unit, University of Brescia, 25123, Brescia, Italy
| | - Giorgio G Fumagalli
- Center for Mind/Brain Sciences-CIMeC, University of Trento, 38068, Rovereto, Italy
| | - Arabella Bouzigues
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Lucy L Russell
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Kiran Samra
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - David M Cash
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Martina Bocchetta
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Emily G Todd
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Rhian S Convery
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Imogen Swift
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Aitana Sogorb-Esteve
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Carolin Heller
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, 3015 GD, The Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, 3015 GD, The Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, 3015 GD, The Netherlands
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, 08036, Barcelona, Spain
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, 20014, San Sebastian, Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Institute, 20014, San Sebastian, Gipuzkoa, Spain
| | - Robert Jr Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Facultéde Médecine, Université Laval, Quebec City, Québec, G1V 0A6, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, 141 52, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, 141 52, Solna, Sweden
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tubingen, 72076, Tubingen, Germany
- Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5S, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3001, Leuven, Belgium
- Neurology Service, University Hospitals Leuven, 3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3001, Leuven, Belgium
| | | | - Pietro Tiraboschi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, OX3 9DU, UK
- Department of Brain Sciences, Imperial College London, London, SW7 2BX, UK
| | - Isabel Santana
- Neurology Service, Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra, 3004-561, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, 3004-561, Coimbra, Portugal
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, M20 3LJ, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, 47057, Essen, Germany
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, M6 8HD, UK
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, 75013, Paris, France
- Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, 75013, Paris, France
- Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, 75013, Paris, France
| | - Florence Pasquier
- Univ Lille, 59000, Lille, France
- , Inserm 1172, 59000, Lille, France
- CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, 59000, Lille, France
| | - Simon Ducharme
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, H3A 1A1, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, 80539, Munich, Germany
- Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster of Systems Neurology, 81377, Munich, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, 50139, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, 50143, Florence, Italy
| | - Markus Otto
- Department of Neurology, University of Ulm, 89081, Ulm, Germany
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy.
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, 25123, Brescia, Italy.
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Frontotemporal dementia (FTD) is a clinically, pathologically and genetically heterogeneous disorder. Whilst disease modifying therapy trials are mostly focused on the symptomatic phase, future studies will move earlier in the disease aiming to prevent symptom onset. This review summarizes the recent work to better understand this presymptomatic period. RECENT FINDINGS The presymptomatic phase can be split into preclinical and prodromal stages. The onset of the preclinical phase is defined by the first presence of pathological inclusions of tau, TDP-43 or fused in sarcoma in the brain. Definitive biomarkers of these pathologies do not yet exist for FTD. The prodromal phase is defined by the onset of mild symptoms. Recent work has highlighted the wide phenotypic spectrum that occurs, with the concept of mild cognitive ± behavioural ± motor impairment (MCBMI) being put forward, and additions to scales such as the CDR plus NACC FTLD now incorporating neuropsychiatric and motor symptoms. SUMMARY It will be important to better characterize the presymptomatic period moving forward and develop robust biomarkers that can be used both for stratification and outcome measures in prevention trials. The work of the FTD Prevention Initiative aims to facilitate this by bringing together data from natural history studies across the world.
Collapse
Affiliation(s)
- Lucy L Russell
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | |
Collapse
|
7
|
Jiskoot LC, Russell LL, Peakman G, Convery RS, Greaves CV, Bocchetta M, Poos JM, Seelaar H, Giannini LAA, van Swieten JC, van Minkelen R, Pijnenburg YAL, Rowe JB, Borroni B, Galimberti D, Masellis M, Tartaglia C, Finger E, Butler CR, Graff C, Laforce R, Sanchez-Valle R, de Mendonça A, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, le Ber I, Levin J, Otto M, Pasquier F, Santana I, Cash DM, Thomas D, Rohrer JD. The Benson Complex Figure Test detects deficits in visuoconstruction and visual memory in symptomatic familial frontotemporal dementia: A GENFI study. J Neurol Sci 2023; 446:120590. [PMID: 36812822 DOI: 10.1016/j.jns.2023.120590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVE Sensitive cognitive markers are still needed for frontotemporal dementia (FTD). The Benson Complex Figure Test (BCFT) is an interesting candidate test, as it assesses visuospatial, visual memory, and executive abilities, allowing the detection of multiple mechanisms of cognitive impairment. To investigate differences in BCFT Copy, Recall and Recognition in presymptomatic and symptomatic FTD mutation carriers, and to explore its cognitive and neuroimaging correlates. METHOD We included cross-sectional data from 332 presymptomatic and 136 symptomatic mutation carriers (GRN, MAPT or C9orf72 mutations), and 290 controls in the GENFI consortium. We examined gene-specific differences between mutation carriers (stratified by CDR® NACC-FTLD score) and controls using Quade's / Pearson Χ2 tests. We investigated associations with neuropsychological test scores and grey matter volume using partial correlations and multiple regression models respectively. RESULTS No significant differences were found between groups at CDR® NACC-FTLD 0-0.5. Symptomatic GRN and C9orf72 mutation carriers had lower Copy scores at CDR® NACC-FTLD ≥2. All three groups had lower Recall scores at CDR® NACC-FTLD ≥2, with MAPT mutation carriers starting at CDR® NACC-FTLD ≥1. All three groups had lower Recognition scores at CDR® NACC FTLD ≥2. Performance correlated with tests for visuoconstruction, memory, and executive function. Copy scores correlated with frontal-subcortical grey matter atrophy, while Recall scores correlated with temporal lobe atrophy. CONCLUSIONS In the symptomatic stage, the BCFT identifies differential mechanisms of cognitive impairment depending on the genetic mutation, corroborated by gene-specific cognitive and neuroimaging correlates. Our findings suggest that impaired performance on the BCFT occurs relatively late in the genetic FTD disease process. Therefore its potential as cognitive biomarker for upcoming clinical trials in presymptomatic to early-stage FTD is most likely limited.
Collapse
Affiliation(s)
- Lize C Jiskoot
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Dementia Research Centre, University College London, London, UK.
| | - Lucy L Russell
- Dementia Research Centre, University College London, London, UK.
| | - Georgia Peakman
- Dementia Research Centre, University College London, London, UK.
| | - Rhian S Convery
- Dementia Research Centre, University College London, London, UK.
| | | | | | - Jackie M Poos
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Lucia A A Giannini
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Rick van Minkelen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Yolande A L Pijnenburg
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, Amsterdam, the Netherlands.
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- University of Milan, Centro Dino Ferrari, Milan, Italy; Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy.
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada..
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada.
| | - Chris R Butler
- Department of Clinical Neurology, University of Oxford, Oxford, UK.
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital-Huddinge, Stockholm, Sweden..
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Université Laval, Québec, Canada.
| | - Raquel Sanchez-Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain.
| | | | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec, Canada.
| | - Isabelle le Ber
- Paris Brain Institute - Institut du Cerveau - Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France; Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, Hôpital Pitié-Salpêtrière, Paris, France; Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
| | - Florence Pasquier
- University of Lille, Lille, France; Inserm, 1172 Lille, France; CHU, CNR-MAJ, Labex Distalz, LiCEND, Lille, France.
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - David M Cash
- Dementia Research Centre, University College London, London, UK.
| | - David Thomas
- Dementia Research Centre, University College London, London, UK.
| | | |
Collapse
|
8
|
Staffaroni AM, Quintana M, Wendelberger B, Heuer HW, Russell LL, Cobigo Y, Wolf A, Goh SYM, Petrucelli L, Gendron TF, Heller C, Clark AL, Taylor JC, Wise A, Ong E, Forsberg L, Brushaber D, Rojas JC, VandeVrede L, Ljubenkov P, Kramer J, Casaletto KB, Appleby B, Bordelon Y, Botha H, Dickerson BC, Domoto-Reilly K, Fields JA, Foroud T, Gavrilova R, Geschwind D, Ghoshal N, Goldman J, Graff-Radford J, Graff-Radford N, Grossman M, Hall MGH, Hsiung GY, Huey ED, Irwin D, Jones DT, Kantarci K, Kaufer D, Knopman D, Kremers W, Lago AL, Lapid MI, Litvan I, Lucente D, Mackenzie IR, Mendez MF, Mester C, Miller BL, Onyike CU, Rademakers R, Ramanan VK, Ramos EM, Rao M, Rascovsky K, Rankin KP, Roberson ED, Savica R, Tartaglia MC, Weintraub S, Wong B, Cash DM, Bouzigues A, Swift IJ, Peakman G, Bocchetta M, Todd EG, Convery RS, Rowe JB, Borroni B, Galimberti D, Tiraboschi P, Masellis M, Finger E, van Swieten JC, Seelaar H, Jiskoot LC, Sorbi S, Butler CR, Graff C, Gerhard A, Langheinrich T, Laforce R, Sanchez-Valle R, de Mendonça A, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, Le Ber I, Levin J, Danek A, Otto M, Pasquier F, Santana I, Kornak J, et alStaffaroni AM, Quintana M, Wendelberger B, Heuer HW, Russell LL, Cobigo Y, Wolf A, Goh SYM, Petrucelli L, Gendron TF, Heller C, Clark AL, Taylor JC, Wise A, Ong E, Forsberg L, Brushaber D, Rojas JC, VandeVrede L, Ljubenkov P, Kramer J, Casaletto KB, Appleby B, Bordelon Y, Botha H, Dickerson BC, Domoto-Reilly K, Fields JA, Foroud T, Gavrilova R, Geschwind D, Ghoshal N, Goldman J, Graff-Radford J, Graff-Radford N, Grossman M, Hall MGH, Hsiung GY, Huey ED, Irwin D, Jones DT, Kantarci K, Kaufer D, Knopman D, Kremers W, Lago AL, Lapid MI, Litvan I, Lucente D, Mackenzie IR, Mendez MF, Mester C, Miller BL, Onyike CU, Rademakers R, Ramanan VK, Ramos EM, Rao M, Rascovsky K, Rankin KP, Roberson ED, Savica R, Tartaglia MC, Weintraub S, Wong B, Cash DM, Bouzigues A, Swift IJ, Peakman G, Bocchetta M, Todd EG, Convery RS, Rowe JB, Borroni B, Galimberti D, Tiraboschi P, Masellis M, Finger E, van Swieten JC, Seelaar H, Jiskoot LC, Sorbi S, Butler CR, Graff C, Gerhard A, Langheinrich T, Laforce R, Sanchez-Valle R, de Mendonça A, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, Le Ber I, Levin J, Danek A, Otto M, Pasquier F, Santana I, Kornak J, Boeve BF, Rosen HJ, Rohrer JD, Boxer AL. Temporal order of clinical and biomarker changes in familial frontotemporal dementia. Nat Med 2022; 28:2194-2206. [PMID: 36138153 PMCID: PMC9951811 DOI: 10.1038/s41591-022-01942-9] [Show More Authors] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 07/08/2022] [Indexed: 01/17/2023]
Abstract
Unlike familial Alzheimer's disease, we have been unable to accurately predict symptom onset in presymptomatic familial frontotemporal dementia (f-FTD) mutation carriers, which is a major hurdle to designing disease prevention trials. We developed multimodal models for f-FTD disease progression and estimated clinical trial sample sizes in C9orf72, GRN and MAPT mutation carriers. Models included longitudinal clinical and neuropsychological scores, regional brain volumes and plasma neurofilament light chain (NfL) in 796 carriers and 412 noncarrier controls. We found that the temporal ordering of clinical and biomarker progression differed by genotype. In prevention-trial simulations using model-based patient selection, atrophy and NfL were the best endpoints, whereas clinical measures were potential endpoints in early symptomatic trials. f-FTD prevention trials are feasible but will likely require global recruitment efforts. These disease progression models will facilitate the planning of f-FTD clinical trials, including the selection of optimal endpoints and enrollment criteria to maximize power to detect treatment effects.
Collapse
Affiliation(s)
- Adam M Staffaroni
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | | | | | - Hilary W Heuer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Yann Cobigo
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Amy Wolf
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sheng-Yang Matt Goh
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | | | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Carolin Heller
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Annie L Clark
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jack Carson Taylor
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Amy Wise
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Elise Ong
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Danielle Brushaber
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Julio C Rojas
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lawren VandeVrede
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Ljubenkov
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joel Kramer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Appleby
- Department of Neurology, Case Western Reserve University, Cleveland, OH, USA
| | - Yvette Bordelon
- Department of Neurology, University of California, Los Angeles, Los Angeles, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tatiana Foroud
- Indiana University School of Medicine, National Centralized Repository for Alzheimer's, Indianapolis, IN, USA
| | | | - Daniel Geschwind
- Department of Neurology, University of California, Los Angeles, Los Angeles, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nupur Ghoshal
- Departments of Neurology and Psychiatry, Washington University School of Medicine, Washington University, St. Louis, MO, USA
| | - Jill Goldman
- Department of Neurology, Columbia University, New York, NY, USA
| | | | | | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew G H Hall
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ging-Yuek Hsiung
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Edward D Huey
- Department of Neurology, Columbia University, New York, NY, USA
| | - David Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Kejal Kantarci
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Daniel Kaufer
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - David Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Walter Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Argentina Lario Lago
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Maria I Lapid
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Diane Lucente
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ian R Mackenzie
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mario F Mendez
- Department of Neurology, University of California, Los Angeles, Los Angeles, USA
| | - Carly Mester
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chiadi U Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Eliana Marisa Ramos
- Department of Neurology, University of California, Los Angeles, Los Angeles, USA
| | - Meghana Rao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Katya Rascovsky
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine P Rankin
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - M Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, Division of Neurology, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Weintraub
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Bonnie Wong
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Imogen J Swift
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Mario Masellis
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | | | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen, Essen, Germany
| | - Tobias Langheinrich
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, UK
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Raquel Sanchez-Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | | | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa, Spain
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
- Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Simon Ducharme
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montreal, Québec, Canada
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
- Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
- Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
- Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Florence Pasquier
- University of Lille, Lille, France
- Inserm, Lille, France
- CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, Lille, France
| | - Isabel Santana
- Neurology Service, Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square London, London, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|