51
|
Wallon D, Nicolas G. Genetica della malattia di Alzheimer. Neurologia 2022. [DOI: 10.1016/s1634-7072(22)47093-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
|
52
|
Bateman RJ, Cummings J, Schobel S, Salloway S, Vellas B, Boada M, Black SE, Blennow K, Fontoura P, Klein G, Assunção SS, Smith J, Doody RS. Gantenerumab: an anti-amyloid monoclonal antibody with potential disease-modifying effects in early Alzheimer's disease. Alzheimers Res Ther 2022; 14:178. [PMID: 36447240 PMCID: PMC9707418 DOI: 10.1186/s13195-022-01110-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND This review describes the research and development process of gantenerumab, a fully human anti-amyloid monoclonal antibody in development to treat early symptomatic and asymptomatic Alzheimer's disease (AD). Anti-amyloid monoclonal antibodies can substantially reverse amyloid plaque pathology and may modify the course of the disease by slowing or stopping its clinical progression. Several molecules targeting amyloid have failed in clinical development due to drug-related factors (e.g., treatment-limiting adverse events, low potency, poor brain penetration), study design/methodological issues (e.g., disease stage, lack of AD pathology confirmation), and other factors. The US Food and Drug Administration's approval of aducanumab, an anti-amyloid monoclonal antibody as the first potential disease-modifying therapy for AD, signaled the value of more than 20 years of drug development, adding to the available therapies the first nominal success since cholinesterase inhibitors and memantine were approved. BODY: Here, we review over 2 decades of gantenerumab development in the context of scientific discoveries in the broader AD field. Key learnings from the field were incorporated into the gantenerumab phase 3 program, including confirmed amyloid positivity as an entry criterion, an enriched clinical trial population to ensure measurable clinical decline, data-driven exposure-response models to inform a safe and efficacious dosing regimen, and the use of several blood-based biomarkers. Subcutaneous formulation for more pragmatic implementation was prioritized as a key feature from the beginning of the gantenerumab development program. CONCLUSION The results from the gantenerumab phase 3 programs are expected by the end of 2022 and will add critical information to the collective knowledge on the search for effective AD treatments.
Collapse
Affiliation(s)
- Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Dominantly Inherited Alzheimer's Network (DIAN) and The Knight Family DIAN Trials Unit (DIAN-TU), St. Louis, MO, USA.
| | - Jeffrey Cummings
- Department of Brain Health, Chambers-Grundy Center for Transformative Neuroscience, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Scott Schobel
- Product Development, F. Hoffmann-La Roche, Basel, Switzerland
| | - Stephen Salloway
- Departments of Psychiatry, Human Behavior, and Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Neurology, Butler Hospital, Providence, RI, USA
- Brown University Center for Alzheimer's Disease Research, Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI, USA
| | - Bruno Vellas
- Department of Geriatric Internal Medicine, UMR 1295 Mixed Unit INSERM - Université Toulouse III Paul Sabatier, Toulouse University Hospital, Toulouse, France
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra E Black
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
- LC Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Paulo Fontoura
- Product Development, F. Hoffmann-La Roche, Basel, Switzerland
| | - Gregory Klein
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche, Basel, Switzerland
| | - Sheila Seleri Assunção
- US Medical Affairs, Genentech Inc., a member of the Roche Group, South San Francisco, CA, USA
| | - Janice Smith
- Product Development, Roche Products Ltd., Welwyn Garden City, UK
| | - Rachelle S Doody
- Product Development, F. Hoffmann-La Roche, Basel, Switzerland
- Product Development, Genentech Inc., a member of the Roche Group, South San Francisco, CA, USA
| |
Collapse
|
53
|
Saunders S, Gregory S, Clement MHS, Birck C, van der Geyten S, Ritchie CW. The European Prevention of Alzheimer's Dementia Programme: An Innovative Medicines Initiative-funded partnership to facilitate secondary prevention of Alzheimer's disease dementia. Front Neurol 2022; 13:1051543. [PMID: 36484017 PMCID: PMC9723139 DOI: 10.3389/fneur.2022.1051543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/28/2022] [Indexed: 08/08/2023] Open
Abstract
INTRODUCTION Tens of millions of people worldwide will develop Alzheimer's disease (AD), and only by intervening early in the preclinical disease can we make a fundamental difference to the rates of late-stage disease where clinical symptoms and societal burden manifest. However, collectively utilizing data, samples, and knowledge amassed by large-scale projects such as the Innovative Medicines Initiative (IMI)-funded European Prevention of Alzheimer's Dementia (EPAD) program will enable the research community to learn, adapt, and implement change. METHOD In the current article, we define and discuss the substantial assets of the EPAD project for the scientific community, patient population, and industry, describe the EPAD structure with a focus on how the public and private sector interacted and collaborated within the project, reflect how IMI specifically supported the achievements of the above, and conclude with a view for future. RESULTS The EPAD project was a €64-million investment to facilitate secondary prevention of AD dementia research. The project recruited over 2,000 research participants into the EPAD longitudinal cohort study (LCS) and included over 400 researchers from 39 partners. The EPAD LCS data and biobank are freely available and easily accessible via the Alzheimer's Disease Data Initiative's (ADDI) AD Workbench platform and the University of Edinburgh's Sample Access Committee. The trial delivery network established within the EPAD program is being incorporated into the truly global offering from the Global Alzheimer's Platform (GAP) for trial delivery, and the almost 100 early-career researchers who were part of the EPAD Academy will take forward their experience and learning from EPAD to the next stage of their careers. DISCUSSION Through GAP, IMI-Neuronet, and follow-on funding from the Alzheimer's Association for the data and sample access systems, the EPAD assets will be maintained and, as and when sponsors seek a new platform trial to be established, the learnings from EPAD will ensure that this can be developed to be even more successful than this first pan-European attempt.
Collapse
Affiliation(s)
- Stina Saunders
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah Gregory
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Serge van der Geyten
- Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Craig W. Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Brain Health Scotland, Edinburgh, United Kingdom
| |
Collapse
|
54
|
Wang G, Li Y, Xiong C, McDade E, Clifford DB, Mills SL, Santacruz AM, Aschenbrenner AJ, Hassenstab J, Benzinger TL, Gordon BA, Fagan AM, Coalier KA, Libre‐Guerra JJ, McCullough A, Joseph‐Mathurin N, Chen CD, Mummery C, Wendelberger BA, Gauthier S, Masellis M, Holdridge KC, Yaari R, Chatterjee S, Sims J, Delmar P, Kerchner GA, Bittner T, Hofmann C, Bateman RJ. Evaluation of dose-dependent treatment effects after mid-trial dose escalation in biomarker, clinical, and cognitive outcomes for gantenerumab or solanezumab in dominantly inherited Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12367. [PMID: 36348972 PMCID: PMC9632865 DOI: 10.1002/dad2.12367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
Introduction While the Dominantly Inherited Alzheimer Network Trials Unit (DIAN-TU) was ongoing, external data suggested higher doses were needed to achieve targeted effects; therefore, doses of gantenerumab were increased 5-fold, and solanezumab was increased 4-fold. We evaluated to what extent mid-trial dose increases produced a dose-dependent treatment effect. Methods Using generalized linear mixed effects (LME) models, we estimated the annual low- and high-dose treatment effects in clinical, cognitive, and biomarker outcomes. Results Both gantenerumab and solanezumab demonstrated dose-dependent treatment effects (significant for gantenerumab, non-significant for solanezumab) in their respective target amyloid biomarkers (Pittsburgh compound B positron emission tomography standardized uptake value ratio and cerebrospinal fluid amyloid beta 42), with gantenerumab demonstrating additional treatment effects in some downstream biomarkers. No dose-dependent treatment effects were observed in clinical or cognitive outcomes. Conclusions Mid-trial dose escalation can be implemented as a remedy for an insufficient initial dose and can be more cost effective and less burdensome to participants than starting a new trial with higher doses, especially in rare diseases. Highlights We evaluated the dose-dependent treatment effect of two different amyloid-specific immunotherapies.Dose-dependent treatment effects were observed in some biomarkers.No dose-dependent treatment effects were observed in clinical/cognitive outcomes, potentially due to the fact that the modified study may not have been powered to detect such treatment effects in symptomatic subjects at a mild stage of disease exposed to high (or maximal) doses of medication for prolonged durations.
Collapse
Affiliation(s)
- Guoqiao Wang
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Yan Li
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Chengjie Xiong
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Eric McDade
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - David B. Clifford
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Susan L. Mills
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Anna M. Santacruz
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | | | - Jason Hassenstab
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | | | - Brian A. Gordon
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Anne M. Fagan
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | - Kelley A. Coalier
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | | | - Austin McCullough
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | | | - Charles D. Chen
- Washington University St LouisSchool of MedicineSt. LouisMissouriUSA
| | | | | | - Serge Gauthier
- McGill University Centre for Studies on Aging in MontrealMontrealQuebecCanada
| | - Mario Masellis
- University of TorontoSunnybrook Health Sciences CentreTorontoOntarioCanada
| | | | - Roy Yaari
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | - John Sims
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | | | | | | | | | | |
Collapse
|
55
|
Aschenbrenner AJ, Li Y, Henson RL, Volluz K, Hassenstab J, Verghese P, West T, Meyer MR, Kirmess KM, Fagan AM, Xiong C, Holtzman D, Morris JC, Bateman RJ, Schindler SE. Comparison of plasma and CSF biomarkers in predicting cognitive decline. Ann Clin Transl Neurol 2022; 9:1739-1751. [PMID: 36183195 PMCID: PMC9639639 DOI: 10.1002/acn3.51670] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVES Concentrations of amyloid-β peptides (Aβ42/Aβ40) and neurofilament light (NfL) can be measured in plasma or cerebrospinal fluid (CSF) and are associated with Alzheimer's disease brain pathology and cognitive impairment. This study directly compared plasma and CSF measures of Aβ42/Aβ40 and NfL as predictors of cognitive decline. METHODS Participants were 65 years or older and cognitively normal at baseline with at least one follow-up cognitive assessment. Analytes were measured with the following types of assays: plasma Aβ42/Aβ40, immunoprecipitation-mass spectrometry; plasma NfL, Simoa; CSF Aβ42/Aβ40, automated immunoassay; CSF NfL plate-based immunoassay. Mixed effects models evaluated the global cognitive composite score over a maximum of 6 years as predicted by the fluid biomarkers. RESULTS Analyses included 371 cognitively normal participants, aged 72.7 ± 5.2 years (mean ± standard deviation) with an average length of follow-up of 3.9 ± 1.6 years. Standardized concentrations of biomarkers were associated with annualized cognitive change: plasma Aβ42/Aβ40, 0.014 standard deviations (95% confidence intervals 0.002 to 0.026); CSF Aβ42/Aβ40, 0.020 (0.008 to 0.032); plasma Nfl, -0.018 (-0.030 to -0.005); and CSF NfL, -0.024 (-0.036 to -0.012). Power analyses estimated that 266 individuals in each treatment arm would be needed to detect a 50% slowing of decline if identified by abnormal plasma measures versus 229 for CSF measures. INTERPRETATION Both plasma and CSF measures of Aβ42/Aβ40 and NfL predicted cognitive decline. A clinical trial that enrolled individuals based on abnormal plasma Aβ42/Aβ40 and NfL levels would require only a marginally larger cohort than if CSF measures were used.
Collapse
Affiliation(s)
- Andrew J. Aschenbrenner
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Yan Li
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - Rachel L. Henson
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Katherine Volluz
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Jason Hassenstab
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | | | | | | | | | - Anne M. Fagan
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - David Holtzman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - John C. Morris
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
56
|
Joseph‐Mathurin N, Llibre‐Guerra JJ, Li Y, McCullough AA, Hofmann C, Wojtowicz J, Park E, Wang G, Preboske GM, Wang Q, Gordon BA, Chen CD, Flores S, Aggarwal NT, Berman SB, Bird TD, Black SE, Borowski B, Brooks WS, Chhatwal JP, Clarnette R, Cruchaga C, Fagan AM, Farlow M, Fox NC, Gauthier S, Hassenstab J, Hobbs DA, Holdridge KC, Honig LS, Hornbeck RC, Hsiung GR, Jack CR, Jimenez‐Velazquez IZ, Jucker M, Klein G, Levin J, Mancini M, Masellis M, McKay NS, Mummery CJ, Ringman JM, Shimada H, Snider BJ, Suzuki K, Wallon D, Xiong C, Yaari R, McDade E, Perrin RJ, Bateman RJ, Salloway SP, Benzinger TL, Clifford DB. Amyloid-Related Imaging Abnormalities in the DIAN-TU-001 Trial of Gantenerumab and Solanezumab: Lessons from a Trial in Dominantly Inherited Alzheimer Disease. Ann Neurol 2022; 92:729-744. [PMID: 36151869 PMCID: PMC9828339 DOI: 10.1002/ana.26511] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To determine the characteristics of participants with amyloid-related imaging abnormalities (ARIA) in a trial of gantenerumab or solanezumab in dominantly inherited Alzheimer disease (DIAD). METHODS 142 DIAD mutation carriers received either gantenerumab SC (n = 52), solanezumab IV (n = 50), or placebo (n = 40). Participants underwent assessments with the Clinical Dementia Rating® (CDR®), neuropsychological testing, CSF biomarkers, β-amyloid positron emission tomography (PET), and magnetic resonance imaging (MRI) to monitor ARIA. Cross-sectional and longitudinal analyses evaluated potential ARIA-related risk factors. RESULTS Eleven participants developed ARIA-E, including 3 with mild symptoms. No ARIA-E was reported under solanezumab while gantenerumab was associated with ARIA-E compared to placebo (odds ratio [OR] = 9.1, confidence interval [CI][1.2, 412.3]; p = 0.021). Under gantenerumab, APOE-ɛ4 carriers were more likely to develop ARIA-E (OR = 5.0, CI[1.0, 30.4]; p = 0.055), as were individuals with microhemorrhage at baseline (OR = 13.7, CI[1.2, 163.2]; p = 0.039). No ARIA-E was observed at the initial 225 mg/month gantenerumab dose, and most cases were observed at doses >675 mg. At first ARIA-E occurrence, all ARIA-E participants were amyloid-PET+, 60% were CDR >0, 60% were past their estimated year to symptom onset, and 60% had also incident ARIA-H. Most ARIA-E radiologically resolved after dose adjustment and developing ARIA-E did not significantly increase odds of trial discontinuation. ARIA-E was more frequently observed in the occipital lobe (90%). ARIA-E severity was associated with age at time of ARIA-E. INTERPRETATION In DIAD, solanezumab was not associated with ARIA. Gantenerumab dose over 225 mg increased ARIA-E risk, with additional risk for individuals APOE-ɛ4(+) or with microhemorrhage. ARIA-E was reversible on MRI in most cases, generally asymptomatic, without additional risk for trial discontinuation. ANN NEUROL 2022;92:729-744.
Collapse
Affiliation(s)
- Nelly Joseph‐Mathurin
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | | | - Yan Li
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | - Austin A. McCullough
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Carsten Hofmann
- Pharmaceutical Sciences, Roche Innovation Center BaselF. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Jakub Wojtowicz
- Product Development, Clinical SafetyF. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Ethan Park
- Division of BiostatisticsWashington University School of MedicineSt. LouisMO
| | - Guoqiao Wang
- Division of BiostatisticsWashington University School of MedicineSt. LouisMO
| | | | - Qing Wang
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Charles D. Chen
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Shaney Flores
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Neelum T. Aggarwal
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| | - Sarah B. Berman
- Departments of Neurology and Clinical and Translational ScienceUniversity of PittsburghPittsburghPA
| | - Thomas D. Bird
- Department of NeurologyUniversity of WashingtonSeattleWA
| | - Sandra E. Black
- Department of Medicine (Neurology), Sunnybrook Health Sciences CentreSunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | | | - William S. Brooks
- Neuroscience Research AustraliaUniversity of New South WalesNew South WalesAustralia
| | - Jasmeer P. Chhatwal
- Department of NeurologyBrigham and Women's Hospital, Massachusetts General HospitalBostonMA
| | - Roger Clarnette
- Department of Internal Medicine, Medical SchoolUniversity of Western AustraliaCrawleyAustralia
| | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSt. LouisMO
| | - Anne M. Fagan
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | - Martin Farlow
- Department of NeurologyIndiana University School of MedicineIndianapolisIN
| | - Nick C. Fox
- UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Serge Gauthier
- McGill Center for Studies in AgingMcGill UniversityMontrealQuebecCanada
| | - Jason Hassenstab
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
- Psychological and Brain SciencesWashington University School of MedicineSt. LouisMO
| | - Diana A. Hobbs
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | | | | | - Russ C. Hornbeck
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - Ging‐Yuek R. Hsiung
- Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | | | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE)Hertie Institute for Clinical Brain Research, University of TübingenTübingenGermany
| | - Gregory Klein
- Clinical Imaging, Biomarkers & Translational TechnologiesF. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Department of Neurology, Ludwig‐Maximilians‐Universität MünchenMunich Cluster for Systems Neurology (SyNergy)MunichGermany
| | | | - Mario Masellis
- Department of Medicine (Neurology), Sunnybrook Health Sciences CentreSunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Nicole S. McKay
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | | | - John M. Ringman
- Department of Neurology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCA
| | - Hiroyuki Shimada
- Diagnostic and Interventional Radiology, Graduate School of MedicineOsaka City UniversityOsakaJapan
| | - B. Joy Snider
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | - Kazushi Suzuki
- Department of Internal MedicineNational Defense Medical CollegeSaitamaJapan
| | | | - Chengjie Xiong
- Division of BiostatisticsWashington University School of MedicineSt. LouisMO
| | | | - Eric McDade
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | - Richard J. Perrin
- Department of NeurologyWashington University School of MedicineSt. LouisMO
- Department of Pathology & ImmunologyWashington University School of MedicineSt. LouisMO
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | - Stephen P. Salloway
- Department of NeurologyAlpert Medical School of Brown University, Butler HospitalProvidenceRI
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt. LouisMO
| | - David B. Clifford
- Department of NeurologyWashington University School of MedicineSt. LouisMO
| | | |
Collapse
|
57
|
Morris JC, Weiner M, Xiong C, Beckett L, Coble D, Saito N, Aisen PS, Allegri R, Benzinger TLS, Berman SB, Cairns NJ, Carrillo MC, Chui HC, Chhatwal JP, Cruchaga C, Fagan AM, Farlow M, Fox NC, Ghetti B, Goate AM, Gordon BA, Graff-Radford N, Day GS, Hassenstab J, Ikeuchi T, Jack CR, Jagust WJ, Jucker M, Levin J, Massoumzadeh P, Masters CL, Martins R, McDade E, Mori H, Noble JM, Petersen RC, Ringman JM, Salloway S, Saykin AJ, Schofield PR, Shaw LM, Toga AW, Trojanowski JQ, Vöglein J, Weninger S, Bateman RJ, Buckles VD. Autosomal dominant and sporadic late onset Alzheimer's disease share a common in vivo pathophysiology. Brain 2022; 145:3594-3607. [PMID: 35580594 PMCID: PMC9989348 DOI: 10.1093/brain/awac181] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
The extent to which the pathophysiology of autosomal dominant Alzheimer's disease corresponds to the pathophysiology of 'sporadic' late onset Alzheimer's disease is unknown, thus limiting the extrapolation of study findings and clinical trial results in autosomal dominant Alzheimer's disease to late onset Alzheimer's disease. We compared brain MRI and amyloid PET data, as well as CSF concentrations of amyloid-β42, amyloid-β40, tau and tau phosphorylated at position 181, in 292 carriers of pathogenic variants for Alzheimer's disease from the Dominantly Inherited Alzheimer Network, with corresponding data from 559 participants from the Alzheimer's Disease Neuroimaging Initiative. Imaging data and CSF samples were reprocessed as appropriate to guarantee uniform pipelines and assays. Data analyses yielded rates of change before and after symptomatic onset of Alzheimer's disease, allowing the alignment of the ∼30-year age difference between the cohorts on a clinically meaningful anchor point, namely the participant age at symptomatic onset. Biomarker profiles were similar for both autosomal dominant Alzheimer's disease and late onset Alzheimer's disease. Both groups demonstrated accelerated rates of decline in cognitive performance and in regional brain volume loss after symptomatic onset. Although amyloid burden accumulation as determined by PET was greater after symptomatic onset in autosomal dominant Alzheimer's disease than in late onset Alzheimer's disease participants, CSF assays of amyloid-β42, amyloid-β40, tau and p-tau181 were largely overlapping in both groups. Rates of change in cognitive performance and hippocampal volume loss after symptomatic onset were more aggressive for autosomal dominant Alzheimer's disease participants. These findings suggest a similar pathophysiology of autosomal dominant Alzheimer's disease and late onset Alzheimer's disease, supporting a shared pathobiological construct.
Collapse
Affiliation(s)
- John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Weiner
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Laurel Beckett
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Dean Coble
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Naomi Saito
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Paul S Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nigel J Cairns
- College of Medicine and Health and the Living Systems Institute, University of Exeter, Exeter, UK
| | | | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Martin Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nick C Fox
- Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Institute of Neurology, London, UK
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Mathias Jucker
- Cell Biology of Neurological Diseases Group, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Johannes Levin
- DZNE Munich, Munich Cluster of Systems Neurology (SyNergy) and Ludwig-Maximilians-Universität, Munich, Germany
| | - Parinaz Massoumzadeh
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- Florey Institute, University of Melbourne, Melbourne, Australia
| | - Ralph Martins
- Sir James McCusker Alzheimer’s Disease Research Unit, Edith Cowan University, Nedlands, Australia
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hiroshi Mori
- Department of Neuroscience, Osaka City University Medical School, Osaka City, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | | | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Salloway
- Department of Neurology, Butler Hospital and Alpert Medical School of Brown University, Providence, RI, 02906, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter R Schofield
- Neuroscience Research Australia and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE) and Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Virginia D Buckles
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
58
|
Ouma LO, Wason JMS, Zheng H, Wilson N, Grayling M. Design and analysis of umbrella trials: Where do we stand? Front Med (Lausanne) 2022; 9:1037439. [PMID: 36313987 PMCID: PMC9596938 DOI: 10.3389/fmed.2022.1037439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background The efficiencies that master protocol designs can bring to modern drug development have seen their increased utilization in oncology. Growing interest has also resulted in their consideration in non-oncology settings. Umbrella trials are one class of master protocol design that evaluates multiple targeted therapies in a single disease setting. Despite the existence of several reviews of master protocols, the statistical considerations of umbrella trials have received more limited attention. Methods We conduct a systematic review of the literature on umbrella trials, examining both the statistical methods that are available for their design and analysis, and also their use in practice. We pay particular attention to considerations for umbrella designs applied outside of oncology. Findings We identified 38 umbrella trials. To date, most umbrella trials have been conducted in early phase settings (73.7%, 28/38) and in oncology (92.1%, 35/38). The quality of statistical information available about conducted umbrella trials to date is poor; for example, it was impossible to ascertain how sample size was determined in the majority of trials (55.3%, 21/38). The literature on statistical methods for umbrella trials is currently sparse. Conclusions Umbrella trials have potentially great utility to expedite drug development, including outside of oncology. However, to enable lessons to be effectively learned from early use of such designs, there is a need for higher-quality reporting of umbrella trials. Furthermore, if the potential of umbrella trials is to be realized, further methodological research is required.
Collapse
Affiliation(s)
- Luke O. Ouma
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - James M. S. Wason
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Haiyan Zheng
- Medical Research Council (MRC) Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nina Wilson
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael Grayling
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
59
|
Saville BR, Berry DA, Berry NS, Viele K, Berry SM. The Bayesian Time Machine: Accounting for temporal drift in multi-arm platform trials. Clin Trials 2022; 19:490-501. [PMID: 35993547 DOI: 10.1177/17407745221112013] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Multi-arm platform trials investigate multiple agents simultaneously, typically with staggered entry and exit of experimental treatment arms versus a shared control arm. In such settings, there is considerable debate whether to limit analyses for a treatment arm to concurrent randomized control subjects or to allow comparisons to both concurrent and non-concurrent (pooled) control subjects. The potential bias from temporal drift over time is at the core of this debate. METHODS We propose time-adjusted analyses, including a "Bayesian Time Machine," to model potential temporal drift in the entire study population, such that primary analyses can incorporate all randomized control subjects from the platform trial. We conduct a simulation study to assess performance relative to utilizing concurrent or pooled controls. RESULTS In multi-arm platform trials with staggered entry, analyses adjusting for temporal drift (either Bayesian or frequentist) have superior estimation of treatment effects and favorable testing properties compared to analyses using either concurrent or pooled controls. The Bayesian Time Machine generally provides estimates with greater precision and smaller mean square error than alternative approaches, at the risk of small bias and small Type I error inflation. CONCLUSIONS The Bayesian Time Machine provides a compromise between bias and precision by smoothing estimates across time and leveraging all available data for the estimation of treatment effects. Prior distributions controlling the behavior of dynamic smoothing across time must be pre-specified and carefully calibrated to the unique context of each trial, appropriately accounting for the population, disease, and endpoints.
Collapse
Affiliation(s)
- Benjamin R Saville
- Berry Consultants, LLC, Austin, TX, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Donald A Berry
- Berry Consultants, LLC, Austin, TX, USA
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, TX, USA
| | | | - Kert Viele
- Berry Consultants, LLC, Austin, TX, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Scott M Berry
- Berry Consultants, LLC, Austin, TX, USA
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
60
|
Zhao P, Zhang N, An Z. Engineering antibody and protein therapeutics to cross the blood-brain barrier. Antib Ther 2022; 5:311-331. [PMID: 36540309 PMCID: PMC9759110 DOI: 10.1093/abt/tbac028] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Diseases in the central nervous system (CNS) are often difficult to treat. Antibody- and protein-based therapeutics hold huge promises in CNS disease treatment. However, proteins are restricted from entering the CNS by the blood-brain barrier (BBB). To achieve enhanced BBB crossing, antibody-based carriers have been developed by utilizing the endogenous macromolecule transportation pathway, known as receptor-mediated transcytosis. In this report, we first provided an overall review on key CNS diseases and the most promising antibody- or protein-based therapeutics approved or in clinical trials. We then reviewed the platforms that are being explored to increase the macromolecule brain entry to combat CNS diseases. Finally, we have analyzed the lessons learned from past experiences and have provided a perspective on the future engineering of novel delivery vehicles for antibody- and protein-based therapies for CNS diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| |
Collapse
|
61
|
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, 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] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [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
|
62
|
Bruno F, Laganà V, Di Lorenzo R, Bruni AC, Maletta R. Calabria as a Genetic Isolate: A Model for the Study of Neurodegenerative Diseases. Biomedicines 2022; 10:biomedicines10092288. [PMID: 36140389 PMCID: PMC9496333 DOI: 10.3390/biomedicines10092288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/08/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Although originally multi-ethnic in its structure, nowadays the Calabria region of southern Italy represents an area with low genetic heterogeneity and a high level of consanguinity that allows rare mutations to be maintained due to the founder effect. A complex research methodology—ranging from clinical activity to the genealogical reconstruction of families/populations across the centuries, the creation of databases, and molecular/genetic research—was modelled on the characteristics of the Calabrian population for more than three decades. This methodology allowed the identification of several novel genetic mutations or variants associated with neurodegenerative diseases. In addition, a higher prevalence of several hereditary neurodegenerative diseases has been reported in this population, such as Alzheimer’s disease, frontotemporal dementia, Parkinson’s disease, Niemann–Pick type C disease, spinocerebellar ataxia, Creutzfeldt–Jakob disease, and Gerstmann–Straussler–Scheinker disease. Here, we summarize and discuss the results of research data supporting the view that Calabria could be considered as a genetic isolate and could represent a model, a sort of outdoor laboratory—similar to very few places in the world—useful for the advancement of knowledge on neurodegenerative diseases.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy
- Correspondence: (F.B.); (A.C.B.)
| | - Valentina Laganà
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy
| | | | - Amalia C. Bruni
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy
- Correspondence: (F.B.); (A.C.B.)
| | - Raffaele Maletta
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy
| |
Collapse
|
63
|
Gao G, Gajewski BJ, Wick J, Beall J, Saver JL, Meinzer C. Optimizing a Bayesian hierarchical adaptive platform trial design for stroke patients. Trials 2022; 23:754. [PMID: 36068547 PMCID: PMC9446515 DOI: 10.1186/s13063-022-06664-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Platform trials are well-known for their ability to investigate multiple arms on heterogeneous patient populations and their flexibility to add/drop treatment arms due to efficacy/lack of efficacy. Because of their complexity, it is important to develop highly optimized, transparent, and rigorous designs that are cost-efficient, offer high statistical power, maximize patient benefit, and are robust to changes over time. METHODS To address these needs, we present a Bayesian platform trial design based on a beta-binomial model for binary outcomes that uses three key strategies: (1) hierarchical modeling of subgroups within treatment arms that allows for borrowing of information across subgroups, (2) utilization of response-adaptive randomization (RAR) schemes that seek a tradeoff between statistical power and patient benefit, and (3) adjustment for potential drift over time. Motivated by a proposed clinical trial that aims to find the appropriate treatment for different subgroup populations of ischemic stroke patients, extensive simulation studies were performed to validate the approach, compare different allocation rules, and study the model operating characteristics. RESULTS AND CONCLUSIONS Our proposed approach achieved high statistical power and good patient benefit and was also robust against population drift over time. Our design provided a good balance between the strengths of both the traditional RAR scheme and fixed 1:1 allocation and may be a promising choice for dichotomous outcomes trials investigating multiple subgroups.
Collapse
Affiliation(s)
- Guangyi Gao
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Byron J Gajewski
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jo Wick
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jonathan Beall
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jeffrey L Saver
- Department of Neurology and Comprehensive Stroke Center, University of California, Los Angeles, CA, 90095, USA
| | - Caitlyn Meinzer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | | |
Collapse
|
64
|
Li Y, Izem R. Novel clinical trial design and analytic methods to tackle challenges in therapeutic development in rare diseases. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1034. [PMID: 36267797 PMCID: PMC9577738 DOI: 10.21037/atm-21-5496] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/28/2022] [Indexed: 12/03/2022]
Abstract
While only a fraction of the worldwide population may have a particular rare disorder, millions of people worldwide are affected across the over 6,000 rare disorders and do not have a safe and effective approved therapy to help them live or manage complications from the disorder. Challenges to clinical development of new therapies in rare disorders include difficulty in powering and recruiting into a study in small and often heterogenous population, scarcity of natural history data informing critical design elements such as endpoint selection and study duration, and ethical and recruitment challenges in randomizing patients to a placebo arm. In this review, we describe some existing and novel strategies to tackle these challenges, by efficient utilization of available resources. We discuss the role of natural history studies and endpoint selection as they remain critical features that apply across designs and disorders. We also review some novel clinical trial designs including incorporating external control and/or longitudinal measures, master protocol designs, and adaptive designs. Additionally, we review some analytic strategies that are often associated with these designs, such as the use of causal inference methods, and Bayesian methods. We hope this review will raise awareness of these novel approaches and encourage their use in studies of rare diseases.
Collapse
Affiliation(s)
- Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, PA, USA
| | - Rima Izem
- Statistical Methodology and Consulting, Novartis, Basel, Switzerland
| |
Collapse
|
65
|
Dokholyan NV, Mohs RC, Bateman RJ. Challenges and progress in research, diagnostics, and therapeutics in Alzheimer's disease and related dementias. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12330. [PMID: 35910674 PMCID: PMC9322822 DOI: 10.1002/trc2.12330] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
The health, well-being, and financial security of Americans are greatly impacted by Alzheimer's disease. The forecast paints an upward trajectory with the number of Americans suffering from Alzheimer's disease and related dementia. To discuss the Alzheimer's crisis, The Senate Committee on Finance, Subcommittee on Health Care, held a hearing titled, "The Alzheimer's Crisis: Examining, Testing, and Treatment Pipelines and Fiscal Implications," on December 16, 2020. Here, we summarize and expand on the discussion of the panel and its review of recent progress, ongoing challenges associated with Alzheimer's disease, and potential initiatives that promise to speed progress in developing treatments and improving care.
Collapse
Affiliation(s)
- Nikolay V. Dokholyan
- Departments of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Departments of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | | | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
66
|
Aschenbrenner AJ, Hassenstab J, Wang G, Li Y, Xiong C, McDade E, Clifford DB, Salloway S, Farlow M, Yaari R, Cheng EYJ, Holdridge KC, Mummery CJ, Masters CL, Hsiung GY, Surti G, Day GS, Weintraub S, Honig LS, Galvin JE, Ringman JM, Brooks WS, Fox NC, Snyder PJ, Suzuki K, Shimada H, Gräber S, Bateman RJ. Avoid or Embrace? Practice Effects in Alzheimer's Disease Prevention Trials. Front Aging Neurosci 2022; 14:883131. [PMID: 35783127 PMCID: PMC9244171 DOI: 10.3389/fnagi.2022.883131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022] Open
Abstract
Demonstrating a slowing in the rate of cognitive decline is a common outcome measure in clinical trials in Alzheimer's disease (AD). Selection of cognitive endpoints typically includes modeling candidate outcome measures in the many, richly phenotyped observational cohort studies available. An important part of choosing cognitive endpoints is a consideration of improvements in performance due to repeated cognitive testing (termed "practice effects"). As primary and secondary AD prevention trials are comprised predominantly of cognitively unimpaired participants, practice effects may be substantial and may have considerable impact on detecting cognitive change. The extent to which practice effects in AD prevention trials are similar to those from observational studies and how these potential differences impact trials is unknown. In the current study, we analyzed data from the recently completed DIAN-TU-001 clinical trial (TU) and the associated DIAN-Observational (OBS) study. Results indicated that asymptomatic mutation carriers in the TU exhibited persistent practice effects on several key outcomes spanning the entire trial duration. Critically, these practice related improvements were larger on certain tests in the TU relative to matched participants from the OBS study. Our results suggest that the magnitude of practice effects may not be captured by modeling potential endpoints in observational studies where assessments are typically less frequent and drug expectancy effects are absent. Using alternate instrument forms (represented in our study by computerized tasks) may partly mitigate practice effects in clinical trials but incorporating practice effects as outcomes may also be viable. Thus, investigators must carefully consider practice effects (either by minimizing them or modeling them directly) when designing cognitive endpoint AD prevention trials by utilizing trial data with similar assessment frequencies.
Collapse
Affiliation(s)
| | - Jason Hassenstab
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Guoqiao Wang
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Yan Li
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Chengjie Xiong
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Eric McDade
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - David B. Clifford
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Stephen Salloway
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Martin Farlow
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Roy Yaari
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | | | | | | | | | - Ghulam Surti
- The University of Rhode Island, Kingston, RI, United States
| | | | - Sandra Weintraub
- Feiniberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lawrence S. Honig
- Columbia University Irving Medical Center, New York, NY, United States
| | - James E. Galvin
- Miller School of Medicine, University of Miami, Miami, FL, United States
| | - John M. Ringman
- University of Southern California, Los Angeles, CA, United States
| | - William S. Brooks
- Neuroscience Research Australia, University of New South Wales Medicine, Randwick, NSW, Australia
| | - Nick C. Fox
- Dementia Research Center, University College London, London, United Kingdom
| | | | | | | | - Susanne Gräber
- German Center for Neurodegenerative Disease (DZNE), Tübingen, Germany
| | - Randall J. Bateman
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
67
|
Strain JF, Barthelemy N, Horie K, Gordon BA, Kilgore C, Aschenbrenner A, Cruchaga C, Xiong C, Joseph-Mathurin N, Hassenstab J, Fagan AM, Li Y, Karch CM, Perrin RJ, Berman SB, Chhatwal JP, Graff-Radford NR, Mori H, Levin J, Noble JM, Allegri R, Schofield PR, Marcus DS, Holtzman DM, Morris JC, Benzinger TLS, McDade EM, Bateman RJ, Ances BM. CSF Tau phosphorylation at Thr205 is associated with loss of white matter integrity in autosomal dominant Alzheimer disease. Neurobiol Dis 2022; 168:105714. [PMID: 35358703 PMCID: PMC9701560 DOI: 10.1016/j.nbd.2022.105714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/26/2022] [Accepted: 03/25/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Hyperphosphorylation of tau leads to conformational changes that destabilize microtubules and hinder axonal transport in Alzheimer's disease (AD). However, it remains unknown whether white matter (WM) decline due to AD is associated with specific Tau phosphorylation site(s). METHODS In autosomal dominant AD (ADAD) mutation carriers (MC) and non-carriers (NC) we compared cerebrospinal fluid (CSF) phosphorylation at tau sites (pT217, pT181, pS202, and pT205) and total tau with WM measures, as derived from diffusion tensor imaging (DTI), and cognition. A WM composite metric, derived from a principal component analysis, was used to identify spatial decline seen in ADAD. RESULTS The WM composite explained over 70% of the variance in MC. WM regions that strongly contributed to the spatial topography were located in callosal and cingulate regions. Loss of integrity within the WM composite was strongly associated with AD progression in MC as defined by the estimated years to onset (EYO) and cognitive decline. A linear regression demonstrated that amyloid, gray matter atrophy and phosphorylation at CSF tau site pT205 each uniquely explained a reduction in the WM composite within MC that was independent of vascular changes (white matter hyperintensities), and age. Hyperphosphorylation of CSF tau at other sites and total tau did not significantly predict WM composite loss. CONCLUSIONS We identified a site-specific relationship between CSF phosphorylated tau and WM decline within MC. The presence of both amyloid deposition and Tau phosphorylation at pT205 were associated with WM composite loss. These findings highlight a primary AD-specific mechanism for WM dysfunction that is tightly coupled to symptom manifestation and cognitive decline.
Collapse
Affiliation(s)
- Jeremy F Strain
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Nicolas Barthelemy
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Kanta Horie
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA; Department of Psychological & Brain Sciences, Washington University, St. Louis, MO 63110, USA
| | - Collin Kilgore
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | | | - Carlos Cruchaga
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Chengjie Xiong
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA; Osaka City University School of Medicine Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Nelly Joseph-Mathurin
- Department of Radiology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA; Osaka City University School of Medicine Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Anne M Fagan
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Yan Li
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Celeste M Karch
- Department of Radiology, Washington University, St. Louis, MO 63110, USA
| | - Richard J Perrin
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Hiroshi Mori
- Osaka City University School of Medicine Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Johannes Levin
- German Center for Neurodegenerative Disease (DZNE) Munich, Munich, Germany
| | - James M Noble
- Department of Neurology, Columbia University, New York, NY 100310, USA
| | - Ricardo Allegri
- School of Medicine, Universidad de Buenos Aires, Viamonte 430, C1053 CABA, Argentina
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia; Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63100, USA
| | - Daniel S Marcus
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Eric M McDade
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University, St. Louis, MO 63110, USA; Department of Radiology, Washington University, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
68
|
Marion J, Ruiz J, Saville BR. Bayesian model of disease progression in mucopolysaccaridosis IIIA. Stat Med 2022; 41:3579-3595. [PMID: 35567343 DOI: 10.1002/sim.9435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/25/2022] [Accepted: 04/22/2022] [Indexed: 11/07/2022]
Abstract
Mucopolysaccaridosis IIIA (MPS IIIA) is a rare genetic disease that afflicts children and leads to neurocognitive degeneration. We develop a Bayesian disease progression model (DPM) of MPS IIIA that characterizes the pattern of cognitive growth and decline in this disease. The DPM is a repeated measures model that incorporates a nonlinear developmental trajectory and shape-invariant random effects. This approach quantifies the pattern of cognitive development in MPS IIIA and addresses differences in biological age, length of follow-up, and clinical outcomes across natural history subjects. The DPM can be used in clinical trials to estimate the percent slowing in disease progression for treatment relative to natural history. Simulations demonstrate that the DPM provides substantial improvements in power relative to alternative analyses.
Collapse
Affiliation(s)
| | - Juan Ruiz
- Forge Biologics, Grove City, Ohio, USA.,Abeona Therapeutics, Madrid, Spain
| | - Benjamin R Saville
- Berry Consultants, Austin, Texas, USA.,Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
69
|
Kidwell KM, Roychoudhury S, Wendelberger B, Scott J, Moroz T, Yin S, Majumder M, Zhong J, Huml RA, Miller V. Application of Bayesian methods to accelerate rare disease drug development: scopes and hurdles. Orphanet J Rare Dis 2022; 17:186. [PMID: 35526036 PMCID: PMC9077995 DOI: 10.1186/s13023-022-02342-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background Design and analysis of clinical trials for rare and ultra-rare disease pose unique challenges to the practitioners. Meeting conventional power requirements is infeasible for diseases where sample sizes are inherently very small. Moreover, rare disease populations are generally heterogeneous and widely dispersed, which complicates study enrollment and design. Leveraging all available information in rare and ultra-rare disease trials can improve both drug development and informed decision-making processes. Main text Bayesian statistics provides a formal framework for combining all relevant information at all stages of the clinical trial, including trial design, execution, and analysis. This manuscript provides an overview of different Bayesian methods applicable to clinical trials in rare disease. We present real or hypothetical case studies that address the key needs of rare disease drug development highlighting several specific Bayesian examples of clinical trials. Advantages and hurdles of these approaches are discussed in detail. In addition, we emphasize the practical and regulatory aspects in the context of real-life applications.
Conclusion The use of innovative trial designs such as master protocols and complex adaptive designs in conjunction with a Bayesian approach may help to reduce sample size, select the correct treatment and population, and accurately and reliably assess the treatment effect in the rare disease setting. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02342-5.
Collapse
Affiliation(s)
- Kelley M Kidwell
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | | | | | - John Scott
- Food and Drug Administration, Washington, DC, USA
| | | | - Shaoming Yin
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | | | | | | | - Veronica Miller
- Forum for Collaborative Research, University of California School of Public Health, Berkeley, CA, USA
| |
Collapse
|
70
|
Salehipour A, Bagheri M, Sabahi M, Dolatshahi M, Boche D. Combination Therapy in Alzheimer’s Disease: Is It Time? J Alzheimers Dis 2022; 87:1433-1449. [DOI: 10.3233/jad-215680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia globally. There is increasing evidence showing AD has no single pathogenic mechanism, and thus treatment approaches focusing only on one mechanism are unlikely to be meaningfully effective. With only one potentially disease modifying treatment approved, targeting amyloid-β (Aβ), AD is underserved regarding effective drug treatments. Combining multiple drugs or designing treatments that target multiple pathways could be an effective therapeutic approach. Considering the distinction between added and combination therapies, one can conclude that most trials fall under the category of added therapies. For combination therapy to have an actual impact on the course of AD, it is likely necessary to target multiple mechanisms including but not limited to Aβ and tau pathology. Several challenges have to be addressed regarding combination therapy, including choosing the correct agents, the best time and stage of AD to intervene, designing and providing proper protocols for clinical trials. This can be achieved by a cooperation between the pharmaceutical industry, academia, private research centers, philanthropic institutions, and the regulatory bodies. Based on all the available information, the success of combination therapy to tackle complicated disorders such as cancer, and the blueprint already laid out on how to implement combination therapy and overcome its challenges, an argument can be made that the field has to move cautiously but quickly toward designing new clinical trials, further exploring the pathological mechanisms of AD, and re-examining the previous studies with combination therapies so that effective treatments for AD may be finally found.
Collapse
Affiliation(s)
- Arash Salehipour
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Motahareh Bagheri
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadmahdi Sabahi
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Dolatshahi
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom
| |
Collapse
|
71
|
Wang G, Liu L, Li Y, Aschenbrenner AJ, Bateman RJ, Delmar P, Schneider LS, Kennedy RE, Cutter GR, Xiong C. Proportional constrained longitudinal data analysis models for clinical trials in sporadic Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12286. [PMID: 35415211 PMCID: PMC8984094 DOI: 10.1002/trc2.12286] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 11/07/2022]
Abstract
Introduction Clinical trials for sporadic Alzheimer's disease generally use mixed models for repeated measures (MMRM) or, to a lesser degree, constrained longitudinal data analysis models (cLDA) as the analysis model with time since baseline as a categorical variable. Inferences using MMRM/cLDA focus on the between-group contrast at the pre-determined, end-of-study assessments, thus are less efficient (eg, less power). Methods The proportional cLDA (PcLDA) and proportional MMRM (pMMRM) with time as a categorical variable are proposed to use all the post-baseline data without the linearity assumption on disease progression. Results Compared with the traditional cLDA/MMRM models, PcLDA or pMMRM lead to greater gain in power (up to 20% to 30%) while maintaining type I error control. Discussion The PcLDA framework offers a variety of possibilities to model longitudinal data such as proportional MMRM (pMMRM) and two-part pMMRM which can model heterogeneous cohorts more efficiently and model co-primary endpoints simultaneously.
Collapse
Affiliation(s)
- Guoqiao Wang
- Division of BiostatisticsWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Lei Liu
- Division of BiostatisticsWashington University School of MedicineSt. LouisMissouriUSA
| | - Yan Li
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Lon S. Schneider
- Department of Psychiatry and The Behavioral SciencesDepartment of NeurologyKeck School of MedicineUniversity of Southern CaliforniaLos AngelesUSA
| | | | - Gary R. Cutter
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamUSA
| | - Chengjie Xiong
- Division of BiostatisticsWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
72
|
Aisen PS, Jimenez-Maggiora GA, Rafii MS, Walter S, Raman R. Early-stage Alzheimer disease: getting trial-ready. Nat Rev Neurol 2022; 18:389-399. [PMID: 35379951 PMCID: PMC8978175 DOI: 10.1038/s41582-022-00645-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Slowing the progression of Alzheimer disease (AD) might be the greatest unmet medical need of our time. Although one AD therapeutic has received a controversial accelerated approval from the FDA, more effective and accessible therapies are urgently needed. Consensus is growing that for meaningful disease modification in AD, therapeutic intervention must be initiated at very early (preclinical or prodromal) stages of the disease. Although the methods for such early-stage clinical trials have been developed, identification and recruitment of the required asymptomatic or minimally symptomatic study participants takes many years and requires substantial funds. As an example, in the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s Disease Trial (the first phase III trial to be performed in preclinical AD), 3.5 years and more than 5,900 screens were required to recruit and randomize 1,169 participants. A new clinical trials infrastructure is required to increase the efficiency of recruitment and accelerate therapeutic progress. Collaborations in North America, Europe and Asia are now addressing this need by establishing trial-ready cohorts of individuals with preclinical and prodromal AD. These collaborations are employing innovative methods to engage the target population, assess risk of brain amyloid accumulation, select participants for biomarker studies and determine eligibility for trials. In the future, these programmes could provide effective tools for pursuing the primary prevention of AD. Here, we review the lessons learned from the AD trial-ready cohorts that have been established to date, with the aim of informing ongoing and future efforts towards efficient, cost-effective trial recruitment. Consensus is growing that intervention in the very early stages of Alzheimer disease is necessary for disease modification. Here, the authors discuss the challenges of recruiting asymptomatic or mildly symptomatic participants for clinical trials, focusing on ‘trial-ready’ cohorts as a potential solution. Trial-ready cohorts are an effective strategy for the identification of participants eligible for clinical trials in early-stage Alzheimer disease (AD). Building these cohorts requires considerable planning and technological infrastructure to facilitate recruitment, remote longitudinal assessment, data management and data storage. Trial-ready cohorts exist for genetically determined populations at risk of AD, such as those with familial AD and Down syndrome; the longitudinal data from these cohorts is improving our understanding of the disease progression in early stages, informing clinical trial design and accelerating recruitment to intervention studies. So far, the challenges experienced by trial-ready cohorts for early-stage AD have included difficulties recruiting an ethnically and racially representative cohort; and for online cohorts, difficulty retaining participants. The results of ongoing work will reveal the success of strategies to improve cohort diversity and retention, and the rates of referral to clinical trials.
Collapse
|
73
|
Morenas-Rodríguez E, Li Y, Nuscher B, Franzmeier N, Xiong C, Suárez-Calvet M, Fagan AM, Schultz S, Gordon BA, Benzinger TLS, Hassenstab J, McDade E, Feederle R, Karch CM, Schlepckow K, Morris JC, Kleinberger G, Nellgard B, Vöglein J, Blennow K, Zetterberg H, Ewers M, Jucker M, Levin J, Bateman RJ, Haass C, Allegri R, Araki A, Barthelemy N, Bechara J, Berman S, Bodge C, Brandon S, Brooks W(B, Brosch J, Buck J, Buckles V, Carter K, Cash L, Chen C, Chhatwal J, Chrem P, Chua J, Chui H, Cruchaga C, Day GS, De La Cruz C, Denner D, Diffenbacher A, Dincer A, Donahue T, Douglas J, Duong D, Egido N, Esposito B, Farlow M, Feldman B, Fitzpatrick C, Flores S, Fox N, Franklin E, Friedrichsen N, Fujii H, Gardener S, Ghetti B, Goate A, Goldberg S, Goldman J, Gonzalez A, Gräber-Sultan S, Graff-Radford N, Graham M, Gray J, Gremminger E, Grilo M, Groves A, Häsler L, Hellm C, Herries E, Hoechst-Swisher L, Hofmann A, Holtzman D, Hornbeck R, Igor Y, Ihara R, Ikeuchi T, Ikonomovic S, Ishii K, Jack C, Jerome G, Johnson E, Käser S, Kasuga K, Keefe S, Klunk W(B, Koeppe R, Koudelis D, Kuder-Buletta E, Laske C, Levey A, Lopez O, Marsh J, Martinez R, Martins R, Mason NS, Masters C, Mawuenyega K, McCullough A, Mejia A, MountzMD J, Mummery C, Nadkarni N, Nagamatsu A, Neimeyer K, Niimi Y, Noble J, Norton J, Nuscher B, O'Connor A, Obermüller U, Patira R, Perrin R, Ping L, Preische O, Renton A, Ringman J, Salloway S, Schofield P, Senda M, Seyfried N, Shady K, Shimada H, Sigurdson W, Smith J, Smith L, Snitz B, Sohrabi H, Stephens S, Taddei K, Thompson S, Wang P, Wang Q, Weamer E, Xu J, Xu X. Soluble TREM2 in CSF and its association with other biomarkers and cognition in autosomal-dominant Alzheimer's disease: a longitudinal observational study. Lancet Neurol 2022; 21:329-341. [PMID: 35305339 PMCID: PMC8926925 DOI: 10.1016/s1474-4422(22)00027-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Therapeutic modulation of TREM2-dependent microglial function might provide an additional strategy to slow the progression of Alzheimer's disease. Although studies in animal models suggest that TREM2 is protective against Alzheimer's pathology, its effect on tau pathology and its potential beneficial role in people with Alzheimer's disease is still unclear. Our aim was to study associations between the dynamics of soluble TREM2, as a biomarker of TREM2 signalling, and amyloid β (Aβ) deposition, tau-related pathology, neuroimaging markers, and cognitive decline, during the progression of autosomal dominant Alzheimer's disease. METHODS We did a longitudinal analysis of data from the Dominantly Inherited Alzheimer Network (DIAN) observational study, which includes families with a history of autosomal dominant Alzheimer's disease. Participants aged over 18 years who were enrolled in DIAN between Jan 1, 2009, and July 31, 2019, were categorised as either carriers of pathogenic variants in PSEN1, PSEN2, and APP genes (n=155) or non-carriers (n=93). We measured amounts of cleaved soluble TREM2 using a novel immunoassay in CSF samples obtained every 2 years from participants who were asymptomatic (Clinical Dementia Rating [CDR]=0) and annually for those who were symptomatic (CDR>0). CSF concentrations of Aβ40, Aβ42, total tau (t-tau), and tau phosphorylated on threonine 181 (p-tau) were measured by validated immunoassays. Predefined neuroimaging measurements were total cortical uptake of Pittsburgh compound B PET (PiB-PET), cortical thickness in the precuneus ascertained by MRI, and hippocampal volume determined by MRI. Cognition was measured using a validated cognitive composite (including DIAN word list test, logical memory delayed recall, digit symbol coding test [total score], and minimental status examination). We based our statistical analysis on univariate and bivariate linear mixed effects models. FINDINGS In carriers of pathogenic variants, a high amyloid burden at baseline, represented by low CSF Aβ42 (β=-4·28 × 10-2 [SE 0·013], p=0·0012), but not high cortical uptake in PiB-PET (β=-5·51 × 10-3 [0·011], p=0·63), was the only predictor of an augmented annual rate of subsequent increase in soluble TREM2. Augmented annual rates of increase in soluble TREM2 were associated with a diminished rate of decrease in amyloid deposition, as measured by Aβ42 in CSF (r=0·56 [0·22], p=0·011), in presymptomatic carriers of pathogenic variants, and with diminished annual rate of increase in PiB-PET (r=-0·67 [0·25], p=0·0060) in symptomatic carriers of pathogenic variants. Presymptomatic carriers of pathogenic variants with annual rates of increase in soluble TREM2 lower than the median showed a correlation between enhanced annual rates of increase in p-tau in CSF and augmented annual rates of increase in PiB-PET signal (r=0·45 [0·21], p=0·035), that was not observed in those with rates of increase in soluble TREM2 higher than the median. Furthermore, presymptomatic carriers of pathogenic variants with rates of increase in soluble TREM2 above or below the median had opposite associations between Aβ42 in CSF and PiB-PET uptake when assessed longitudinally. Augmented annual rates of increase in soluble TREM2 in presymptomatic carriers of pathogenic variants correlated with decreased cortical shrinkage in the precuneus (r=0·46 [0·22]), p=0·040) and diminished cognitive decline (r=0·67 [0·22], p=0·0020). INTERPRETATION Our findings in autosomal dominant Alzheimer's disease position the TREM2 response within the amyloid cascade immediately after the first pathological changes in Aβ aggregation and further support the role of TREM2 on Aβ plaque deposition and compaction. Furthermore, these findings underpin a beneficial effect of TREM2 on Aβ deposition, Aβ-dependent tau pathology, cortical shrinkage, and cognitive decline. Soluble TREM2 could, therefore, be a key marker for clinical trial design and interpretation. Efforts to develop TREM2-boosting therapies are ongoing. FUNDING German Research Foundation, US National Institutes of Health.
Collapse
Affiliation(s)
- Estrella Morenas-Rodríguez
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany.
| | - Yan Li
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA
| | - Brigitte Nuscher
- German Center for Neurodegenerative Diseases, Munich, Germany,Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain,Servei de Neurologia, Hospital del Mar Medical Research Institute, Barcelona, Spain,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephanie Schultz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Regina Feederle
- German Center for Neurodegenerative Diseases, Munich, Germany,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany,Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center, Munich, Germany,German Research Center for Environmental Health, Neuherberg, Germany
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases, Munich, Germany,Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Gernot Kleinberger
- German Center for Neurodegenerative Diseases, Munich, Germany,Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Bengt Nellgard
- Department of Anesthesiology and Intensive Care, Sahlgrenska University Hospital, Mölndal, Sweden,Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases, Munich, Germany,Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,Department of Neurodegenerative Disease, UCL Queens Square Institute of Neurology, University College London, London, UK,UK Dementia Research Institute, University College London, London, UK,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, China
| | - Michael Ewers
- German Center for Neurodegenerative Diseases, Munich, Germany,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases, Tübingen, Germany,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases, Munich, Germany,Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases, Munich, Germany,Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Benatar M, Wuu J, McHutchison C, Postuma RB, Boeve BF, Petersen R, Ross CA, Rosen H, Arias JJ, Fradette S, McDermott MP, Shefner J, Stanislaw C, Abrahams S, Cosentino S, Andersen PM, Finkel RS, Granit V, Grignon AL, Rohrer JD, McMillan CT, Grossman M, Al-Chalabi A, Turner MR. Preventing amyotrophic lateral sclerosis: insights from pre-symptomatic neurodegenerative diseases. Brain 2022; 145:27-44. [PMID: 34677606 PMCID: PMC8967095 DOI: 10.1093/brain/awab404] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/16/2021] [Accepted: 10/08/2021] [Indexed: 11/12/2022] Open
Abstract
Significant progress has been made in understanding the pre-symptomatic phase of amyotrophic lateral sclerosis. While much is still unknown, advances in other neurodegenerative diseases offer valuable insights. Indeed, it is increasingly clear that the well-recognized clinical syndromes of Alzheimer's disease, Parkinson's disease, Huntington's disease, spinal muscular atrophy and frontotemporal dementia are also each preceded by a pre-symptomatic or prodromal period of varying duration, during which the underlying disease process unfolds, with associated compensatory changes and loss of inherent system redundancy. Key insights from these diseases highlight opportunities for discovery in amyotrophic lateral sclerosis. The development of biomarkers reflecting amyloid and tau has led to a shift in defining Alzheimer's disease based on inferred underlying histopathology. Parkinson's disease is unique among neurodegenerative diseases in the number and diversity of non-genetic biomarkers of pre-symptomatic disease, most notably REM sleep behaviour disorder. Huntington's disease benefits from an ability to predict the likely timing of clinically manifest disease based on age and CAG-repeat length alongside reliable neuroimaging markers of atrophy. Spinal muscular atrophy clinical trials have highlighted the transformational value of early therapeutic intervention, and studies in frontotemporal dementia illustrate the differential role of biomarkers based on genotype. Similar advances in amyotrophic lateral sclerosis would transform our understanding of key events in pathogenesis, thereby dramatically accelerating progress towards disease prevention. Deciphering the biology of pre-symptomatic amyotrophic lateral sclerosis relies on a clear conceptual framework for defining the earliest stages of disease. Clinically manifest amyotrophic lateral sclerosis may emerge abruptly, especially among those who harbour genetic mutations associated with rapidly progressive amyotrophic lateral sclerosis. However, the disease may also evolve more gradually, revealing a prodromal period of mild motor impairment preceding phenoconversion to clinically manifest disease. Similarly, cognitive and behavioural impairment, when present, may emerge gradually, evolving through a prodromal period of mild cognitive impairment or mild behavioural impairment before progression to amyotrophic lateral sclerosis. Biomarkers are critically important to studying pre-symptomatic amyotrophic lateral sclerosis and essential to efforts to intervene therapeutically before clinically manifest disease emerges. The use of non-genetic biomarkers, however, presents challenges related to counselling, informed consent, communication of results and limited protections afforded by existing legislation. Experiences from pre-symptomatic genetic testing and counselling, and the legal protections against discrimination based on genetic data, may serve as a guide. Building on what we have learned-more broadly from other pre-symptomatic neurodegenerative diseases and specifically from amyotrophic lateral sclerosis gene mutation carriers-we present a road map to early intervention, and perhaps even disease prevention, for all forms of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Joanne Wuu
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Caroline McHutchison
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Ronald B Postuma
- Department of Neurology, Montreal Neurological Institute, McGill University, Montreal, Canada
| | | | | | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Howard Rosen
- Department of Neurology, University of California San Francisco, CA, USA
| | - Jalayne J Arias
- Department of Neurology, University of California San Francisco, CA, USA
| | | | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Sharon Abrahams
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | | | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, Sweden
| | - Richard S Finkel
- Department of Pediatric Medicine, Center for Experimental Neurotherapeutics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Volkan Granit
- Department of Neurology, University of Miami, Miami, FL, USA
| | | | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
75
|
Cullen N, Janelidze S, Palmqvist S, Stomrud E, Mattsson-Carlgren N, Hansson O. Association of CSF Aβ 38 Levels With Risk of Alzheimer Disease-Related Decline. Neurology 2022; 98:e958-e967. [PMID: 34937781 PMCID: PMC8901176 DOI: 10.1212/wnl.0000000000013228] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/08/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Experimental studies suggest that the balance between short and long β-amyloid (Aβ) species might modulate the toxic effects of Aβ in Alzheimer disease (AD), but clinical evidence is lacking. We studied whether Aβ38 levels in CSF relate to risk of AD dementia and cognitive decline. METHODS CSF Aβ38 levels were measured in 656 individuals across 2 clinical cohorts: the Swedish BioFINDER study and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Cox regression models were used to evaluate the association between baseline Aβ38 levels and risk of AD dementia in AD biomarker-positive individuals (AD+; determined by CSF phosphorylated tau [P-tau]/Aβ42 ratio) with subjective cognitive decline (SCD) or mild cognitive impairment (MCI). Linear mixed-effects models were used to evaluate the association between baseline Aβ38 levels and cognitive decline as measured by the Mini-Mental State Examination (MMSE) in AD+ participants with SCD, MCI, or AD dementia. RESULTS In the BioFINDER cohort, high Aβ38 levels were associated with slower decline in MMSE score (β = 0.30 points per SD, p = 0.001) and with lower risk of conversion to AD dementia (hazard ratio 0.83 per SD, p = 0.03). In the ADNI cohort, higher Aβ38 levels were associated with less decline in MMSE score (β = 0.27, p = 0.01) but not risk of conversion to AD dementia (p = 0.66). Aβ38 levels in both cohorts were significantly associated with both cognitive and clinical outcomes when further adjusted for CSF P-tau or CSF Aβ42 levels. DISCUSSION Higher CSF Aβ38 levels are associated with lower risk of AD-related changes in 2 independent clinical cohorts. These findings suggest that γ-secretase modulators could be effective as disease-altering therapy. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT03174938.
Collapse
Affiliation(s)
- Nicholas Cullen
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden.
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| |
Collapse
|
76
|
Nagata T, Shinagawa S, Nakajima S, Noda Y, Mimura M. Pharmacotherapeutic combinations for the treatment of Alzheimer's disease. Expert Opin Pharmacother 2022; 23:727-737. [PMID: 35230200 DOI: 10.1080/14656566.2022.2042514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common form of dementia, and four medications are currently available as symptomatic therapies: three cholinesterase inhibitors (ChEI) and memantine. In June 2021, aducanumab was approved in the United States under an accelerated approval pathway as the first novel putative disease-modifying therapy (p-DMT) targeting the β-amyloid (Aβ) cascade in the brain. The combination of several monotherapies to address the multifactorial pathogenesis of neurodegenerative diseases is an anticipated next step. AREAS COVERED The cholinergic hypothesis and the amyloid cascade hypothesis have been proposed as explanations for the pathogenesis of AD. Given the limited effectiveness of monotherapies based on these hypotheses, approaches using combination therapy are attempting to address the complexity of AD pathogenesis, including putative causative proteins-related neurodegeneration, neurotransmitters, and neuroinflammation, in a comprehensive manner. EXPERT OPINION The efficacy of an initial or add-on combination approach to counteracting neurodegenerative processes and functional deterioration has been investigated. The combination of symptomatic therapies with approved anti-dementia medicines (one ChEI and memantine) has been found to be functionally effective for a moderately severe disease stage. Furthermore, combination strategies involving p-DMTs, symptomatic therapies, and neuro-regeneration may be useful in the future.
Collapse
Affiliation(s)
- Tomoyuki Nagata
- Department of Psychiatry, The Jikei University School of Medicine, Tokyo, Japan.,Department of Psychiatry, Airanomori Hospital, Kagoshima, Japan
| | | | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
77
|
Schindler SE. Predicting Symptom Onset in Sporadic Alzheimer's Disease: "How Long Do I Have?". J Alzheimers Dis 2022; 90:975-979. [PMID: 35213383 DOI: 10.3233/jad-215722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Predicting not just if but when cognitively normal individuals will develop the onset of Alzheimer's disease (AD) dementia seems increasingly feasible, as evidenced by converging findings from several approaches and cohorts. These estimates may improve the efficiency of clinical trials by better identifying cognitively normal individuals at high risk of developing AD symptoms. As models are refined, the implications of disclosing estimates of the age of AD symptom onset must be examined, since telling a cognitively normal individual the age they are expected to develop AD symptoms may have different implications than disclosing increased risk for AD dementia.
Collapse
Affiliation(s)
- Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
78
|
Karran E, De Strooper B. The amyloid hypothesis in Alzheimer disease: new insights from new therapeutics. Nat Rev Drug Discov 2022; 21:306-318. [PMID: 35177833 DOI: 10.1038/s41573-022-00391-w] [Citation(s) in RCA: 340] [Impact Index Per Article: 113.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/14/2022]
Abstract
Many drugs that target amyloid-β (Aβ) in Alzheimer disease (AD) have failed to demonstrate clinical efficacy. However, four anti-Aβ antibodies have been shown to mediate the removal of amyloid plaque from brains of patients with AD, and the FDA has recently granted accelerated approval to one of these, aducanumab, using reduction of amyloid plaque as a surrogate end point. The rationale for approval and the extent of the clinical benefit from these antibodies are under intense debate. With the aim of informing this debate, we review clinical trial data for drugs that target Aβ from the perspective of the temporal interplay between the two pathognomonic protein aggregates in AD - Aβ plaques and tau neurofibrillary tangles - and their relationship to cognitive impairment, highlighting differences in drug properties that could affect their clinical performance. On this basis, we propose that Aβ pathology drives tau pathology, that amyloid plaque would need to be reduced to a low level (~20 centiloids) to reveal significant clinical benefit and that there will be a lag between the removal of amyloid and the potential to observe a clinical benefit. We conclude that the speed of amyloid removal from the brain by a potential therapy will be important in demonstrating clinical benefit in the context of a clinical trial.
Collapse
Affiliation(s)
- Eric Karran
- Cambridge Research Center, AbbVie, Inc., Cambridge, MA, USA.
| | - Bart De Strooper
- VIB Centre for Brain Disease Research, KU Leuven, Leuven, Belgium.,UK Dementia Research Institute, University College London, London, UK
| |
Collapse
|
79
|
Golde TE. Disease-Modifying Therapies for Alzheimer's Disease: More Questions than Answers. Neurotherapeutics 2022; 19:209-227. [PMID: 35229269 PMCID: PMC8885119 DOI: 10.1007/s13311-022-01201-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Scientific advances over the last four decades have steadily infused the Alzheimer's disease (AD) field with great optimism that therapies targeting Aβ, amyloid, tau, and innate immune activation states in the brain would provide disease modification. Unfortunately, this optimistic scenario has not yet played out. Though a recent approval of the anti-Aβ aggregate binding antibody, Aduhelm (aducanumab), as a "disease-modifying therapy for AD" is viewed by some as a breakthrough, many remain unconvinced by the data underlying this approval. Collectively, we have not succeeded in changing AD from a largely untreatable, inevitable, and incurable disease to a treatable, preventable, and curable one. Here, I will review the major foci of the AD "disease-modifying" therapeutic pipeline and some of the "open questions" that remain in terms of these therapeutic approaches. I will conclude the review by discussing how we, as a field, might adjust our approach, learning from our past failures to ensure future success.
Collapse
Affiliation(s)
- Todd E Golde
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, Evelyn F. and William L. McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
80
|
Savelieff MG, Noureldein MH, Feldman EL. Systems Biology to Address Unmet Medical Needs in Neurological Disorders. Methods Mol Biol 2022; 2486:247-276. [PMID: 35437727 PMCID: PMC9446424 DOI: 10.1007/978-1-0716-2265-0_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Neurological diseases are highly prevalent and constitute a significant cause of mortality and disability. Neurological disorders encompass a heterogeneous group of neurodegenerative conditions, broadly characterized by injury to the peripheral and/or central nervous system. Although the etiology of neurological diseases varies greatly, they share several characteristics, such as heterogeneity of clinical presentation, non-cell autonomous nature, and diversity of cellular, subcellular, and molecular pathways. Systems biology has emerged as a valuable platform for addressing the challenges of studying heterogeneous neurological diseases. Systems biology has manifold applications to address unmet medical needs for neurological illness, including integrating and correlating different large datasets covering the transcriptome, epigenome, proteome, and metabolome associated with a specific condition. This is particularly useful for disentangling the heterogeneity and complexity of neurological conditions. Hence, systems biology can help in uncovering pathophysiology to develop novel therapeutic targets and assessing the impact of known treatments on disease progression. Additionally, systems biology can identify early diagnostic biomarkers, to help diagnose neurological disease preceded by a long subclinical phase, as well as define the exposome, the collection of environmental toxicants that increase risk of certain neurological diseases. In addition to these current applications, there are numerous potential emergent uses, such as precision medicine.
Collapse
Affiliation(s)
- Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Mohamed H Noureldein
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eva L Feldman
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
81
|
Paganoni S, Berry JD, Quintana M, Macklin E, Saville BR, Detry MA, Chase M, Sherman A, Yu H, Drake K, Andrews J, Shefner J, Chibnik L, Vestrucci M, Cudkowicz ME. Adaptive Platform Trials to Transform ALS Therapy Development. Ann Neurol 2021; 91:165-175. [PMID: 34935174 DOI: 10.1002/ana.26285] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 11/07/2022]
Abstract
Current therapeutic development in ALS relies on individual randomized clinical trials to test a specific investigational product in a single patient population. This approach has intrinsic limitations including cost, time, and lack of flexibility. Adaptive platform trials represent a novel approach to investigate several interventions for a single disease in a continuous manner. Already in use in oncology, this approach is now being employed more often in neurology. Here, we describe a newly launched, platform trial for amyotrophic lateral sclerosis (ALS). The HEALEY ALS Platform Trial is testing multiple investigational products concurrently in people with ALS, with the goal of rapidly identifying novel treatments, biomarkers, and trial endpoints. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sabrina Paganoni
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Eric Macklin
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Benjamin R Saville
- Berry Consultants, Austin, TX.,Department of Biostatistics, Vanderbilt University School of Medicine
| | | | - Marianne Chase
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Alex Sherman
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Hong Yu
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kristin Drake
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | | | - Lori Chibnik
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Merit E Cudkowicz
- Sean M. Healey & AMG Center for ALS at Mass General and Neurological Clinical Research Insititute, Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
82
|
Iglesias-Lopez C, Agustí A, Vallano A, Obach M. Current landscape of clinical development and approval of advanced therapies. Mol Ther Methods Clin Dev 2021; 23:606-618. [PMID: 34901306 PMCID: PMC8626628 DOI: 10.1016/j.omtm.2021.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/06/2021] [Accepted: 11/07/2021] [Indexed: 01/26/2023]
Abstract
Advanced therapy medicinal products (ATMPs) are innovative therapies that mainly target orphan diseases and high unmet medical needs. The uncertainty about the product's benefit-risk balance at the time of approval, the limitations of nonclinical development, and the complex quality aspects of those highly individualized advanced therapies are playing a key role in the clinical development, approval, and post-marketing setting for these therapies. This article reviews the current landscape of clinical development of advanced therapies, its challenges, and some of the efforts several stakeholders are conducting to move forward within this field. Progressive iteration of the science, methodologically sound clinical developments, establishing new standards for ATMPs development with the aim to ensure consistency in clinical development, and the reproducibility of knowledge is required, not only to increase the evidence generation for approval but to set principles to achieve translational success in this field.
Collapse
Affiliation(s)
- Carolina Iglesias-Lopez
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonia Agustí
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Clinical Pharmacology Service, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Antoni Vallano
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Medicines Department, Catalan Healthcare Service, Barcelona, Spain
| | - Merce Obach
- Medicines Department, Catalan Healthcare Service, Barcelona, Spain
| |
Collapse
|
83
|
Limorenko G, Lashuel HA. Revisiting the grammar of Tau aggregation and pathology formation: how new insights from brain pathology are shaping how we study and target Tauopathies. Chem Soc Rev 2021; 51:513-565. [PMID: 34889934 DOI: 10.1039/d1cs00127b] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Converging evidence continues to point towards Tau aggregation and pathology formation as central events in the pathogenesis of Alzheimer's disease and other Tauopathies. Despite significant advances in understanding the morphological and structural properties of Tau fibrils, many fundamental questions remain about what causes Tau to aggregate in the first place. The exact roles of cofactors, Tau post-translational modifications, and Tau interactome in regulating Tau aggregation, pathology formation, and toxicity remain unknown. Recent studies have put the spotlight on the wide gap between the complexity of Tau structures, aggregation, and pathology formation in the brain and the simplicity of experimental approaches used for modeling these processes in research laboratories. Embracing and deconstructing this complexity is an essential first step to understanding the role of Tau in health and disease. To help deconstruct this complexity and understand its implication for the development of effective Tau targeting diagnostics and therapies, we firstly review how our understanding of Tau aggregation and pathology formation has evolved over the past few decades. Secondly, we present an analysis of new findings and insights from recent studies illustrating the biochemical, structural, and functional heterogeneity of Tau aggregates. Thirdly, we discuss the importance of adopting new experimental approaches that embrace the complexity of Tau aggregation and pathology as an important first step towards developing mechanism- and structure-based therapies that account for the pathological and clinical heterogeneity of Alzheimer's disease and Tauopathies. We believe that this is essential to develop effective diagnostics and therapies to treat these devastating diseases.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
84
|
Schindler SE, Li Y, Buckles VD, Gordon BA, Benzinger TLS, Wang G, Coble D, Klunk WE, Fagan AM, Holtzman DM, Bateman RJ, Morris JC, Xiong C. Predicting Symptom Onset in Sporadic Alzheimer Disease With Amyloid PET. Neurology 2021; 97:e1823-e1834. [PMID: 34504028 PMCID: PMC8610624 DOI: 10.1212/wnl.0000000000012775] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/12/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To predict when cognitively normal individuals with brain amyloidosis will develop symptoms of Alzheimer disease (AD). METHODS Brain amyloid burden was measured by amyloid PET with Pittsburgh compound B. The mean cortical standardized uptake value ratio (SUVR) was transformed into a timescale with the use of longitudinal data. RESULTS Amyloid accumulation was evaluated in 236 individuals who underwent >1 amyloid PET scan. The average age was 66.5 ± 9.2 years, and 12 individuals (5%) had cognitive impairment at their baseline amyloid PET scan. A tipping point in amyloid accumulation was identified at a low level of amyloid burden (SUVR 1.2), after which nearly all individuals accumulated amyloid at a relatively consistent rate until reaching a high level of amyloid burden (SUVR 3.0). The average time between levels of amyloid burden was used to estimate the age at which an individual reached SUVR 1.2. Longitudinal clinical diagnoses for 180 individuals were aligned by the estimated age at SUVR 1.2. In the 22 individuals who progressed from cognitively normal to a typical AD dementia syndrome, the estimated age at which an individual reached SUVR 1.2 predicted the age at symptom onset (R 2 = 0.54, p < 0.0001, root mean square error [RMSE] 4.5 years); the model was more accurate after exclusion of 3 likely misdiagnoses (R 2 = 0.84, p < 0.0001, RMSE 2.8 years). CONCLUSION The age at symptom onset in sporadic AD is strongly correlated with the age at which an individual reaches a tipping point in amyloid accumulation.
Collapse
Affiliation(s)
- Suzanne E Schindler
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA.
| | - Yan Li
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Virginia D Buckles
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Brian A Gordon
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Tammie L S Benzinger
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Guoqiao Wang
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Dean Coble
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - William E Klunk
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Anne M Fagan
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - David M Holtzman
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Randall J Bateman
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - John C Morris
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| | - Chengjie Xiong
- From the Department of Neurology (S.E.S., Y.L., V.D.B., A.M.F., D.M.H., R.J.B., J.C.M.), Knight Alzheimer Disease Research Center (S.E.S., V.D.B., B.A.G., T.L.S.B., G.W., D.C., A.M.F., D.M.H., R.J.B., J.C.M., C.X.), Division of Biostatistics (Y.L., G.W., D.C., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (A.M.F., D.M.H., R.J.B.), Washington University School of Medicine, St. Louis, MO; and Department of Neurology and Psychiatry (W.E.K.), University of Pittsburgh, PA
| |
Collapse
|
85
|
Ezzati A, Abdulkadir A, Jack CR, Thompson PM, Harvey DJ, Truelove-Hill M, Sreepada LP, Davatzikos C, Lipton RB. Predictive value of ATN biomarker profiles in estimating disease progression in Alzheimer's disease dementia. Alzheimers Dement 2021; 17:1855-1867. [PMID: 34870371 PMCID: PMC8842842 DOI: 10.1002/alz.12491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 07/15/2021] [Accepted: 09/07/2021] [Indexed: 01/18/2023]
Abstract
We aimed to evaluate the value of ATN biomarker classification system (amyloid beta [A], pathologic tau [T], and neurodegeneration [N]) for predicting conversion from mild cognitive impairment (MCI) to dementia. In a sample of people with MCI (n = 415) we assessed predictive performance of ATN classification using empirical knowledge-based cut-offs for each component of ATN and compared it to two data-driven approaches, logistic regression and RUSBoost machine learning classifiers, which used continuous clinical or biomarker scores. In data-driven approaches, we identified ATN features that distinguish normals from individuals with dementia and used them to classify persons with MCI into dementia-like and normal groups. Both data-driven classification methods performed better than the empirical cut-offs for ATN biomarkers in predicting conversion to dementia. Classifiers that used clinical features performed as well as classifiers that used ATN biomarkers for prediction of progression to dementia. We discuss that data-driven modeling approaches can improve our ability to predict disease progression and might have implications in future clinical trials.
Collapse
Affiliation(s)
- Ali Ezzati
- Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical center, Bronx, New York, USA
| | - Ahmed Abdulkadir
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Paul M. Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Danielle J. Harvey
- Department of Public Health Sciences, University of California-Davis, Davis, California, USA
| | - Monica Truelove-Hill
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lasya P. Sreepada
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Richard B. Lipton
- Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical center, Bronx, New York, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
86
|
Aisen PS, Bateman RJ, Carrillo M, Doody R, Johnson K, Sims JR, Sperling R, Vellas B. Platform Trials to Expedite Drug Development in Alzheimer's Disease: A Report from the EU/US CTAD Task Force. J Prev Alzheimers Dis 2021; 8:306-312. [PMID: 34101788 PMCID: PMC8136263 DOI: 10.14283/jpad.2021.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 11/11/2022]
Abstract
A diverse range of platforms has been established to increase the efficiency and speed of clinical trials for Alzheimer's disease (AD). These platforms enable parallel assessment of multiple therapeutics, treatment regimens, or participant groups; use uniform protocols and outcome measures; and may allow treatment arms to be added or dropped based on interim analyses of outcomes. The EU/US CTAD Task Force discussed the lessons learned from the Dominantly Inherited Alzheimer's Network Trials Unit (DIAN-TU) platform trial and the challenges addressed by other platform trials that have launched or are in the planning stages. The landscape of clinical trial platforms in the AD space includes those testing experimental therapies such as DIAN-TU, platforms designed to test multidomain interventions, and those designed to streamline trial recruitment by building trial-ready cohorts. The heterogeneity of the AD patient population, AD drugs, treatment regimens, and analytical methods complicates the design and execution of platform trials, yet Task Force members concluded that platform trials are essential to advance the search for effective AD treatments, including combination therapies.
Collapse
Affiliation(s)
- P S Aisen
- P.S. Aisen, University of Southern California Alzheimer's Therapeutic Research Institute, San Diego, CA, USA,
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Floeter MK, Warden D, Lange D, Wymer J, Paganoni S, Mitsumoto H. Clinical care and therapeutic trials in PLS. Amyotroph Lateral Scler Frontotemporal Degener 2021; 21:67-73. [PMID: 33602017 DOI: 10.1080/21678421.2020.1837180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Primary lateral sclerosis (PLS) is an extremely rare central nervous system degenerative disorder characterized by slowly progressive upper motor neuron loss leading to severe limb and bulbar dysfunction and disability. Although not necessarily life-shortening, PLS disease burden is substantial and improved symptomatic treatments are a major unmet need, especially for the often refractory spasticity that is a core feature of the syndrome. In Section 1, we describe clinical care needs and emphasize a highly personalized approach that can be best attained through multidisciplinary management. In Section 2, we describe progress in clinical trials in PLS that includes advances in symptomatic treatment, disease-modifying therapy, and emerging innovative trials.
Collapse
Affiliation(s)
- Mary Kay Floeter
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Deborah Warden
- Departments of Neurology and Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Dale Lange
- Department of Neurology, Hospital for Special Surgery, Weill Cornell School of Medicine, New York, NY, USA
| | - James Wymer
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Sabrina Paganoni
- Healey Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA, and
| | - Hiroshi Mitsumoto
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
88
|
Vignon A, Salvador-Prince L, Lehmann S, Perrier V, Torrent J. Deconstructing Alzheimer's Disease: How to Bridge the Gap between Experimental Models and the Human Pathology? Int J Mol Sci 2021; 22:8769. [PMID: 34445475 PMCID: PMC8395727 DOI: 10.3390/ijms22168769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Discovered more than a century ago, Alzheimer's disease (AD) is not only still present in our societies but has also become the most common dementia, with 50 million people worldwide affected by the disease. This number is expected to double in the next generation, and no cure is currently available to slow down or stop the disease progression. Recently, some advances were made due to the approval of the aducanumab treatment by the American Food and Drug Administration. The etiology of this human-specific disease remains poorly understood, and the mechanisms of its development have not been completely clarified. Several hypotheses concerning the molecular mechanisms of AD have been proposed, but the existing studies focus primarily on the two main markers of the disease: the amyloid β peptides, whose aggregation in the brain generates amyloid plaques, and the abnormally phosphorylated tau proteins, which are responsible for neurofibrillary tangles. These protein aggregates induce neuroinflammation and neurodegeneration, which, in turn, lead to cognitive and behavioral deficits. The challenge is, therefore, to create models that best reproduce this pathology. This review aims at gathering the different existing AD models developed in vitro, in cellulo, and in vivo. Many models have already been set up, but it is necessary to identify the most relevant ones for our investigations. The purpose of the review is to help researchers to identify the most pertinent disease models, from the most often used to the most recently generated and from simple to complex, explaining their specificities and giving concrete examples.
Collapse
Affiliation(s)
- Anaïs Vignon
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| | - Lucie Salvador-Prince
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| | - Sylvain Lehmann
- INM, University of Montpellier, INSERM, CHU Montpellier, 34095 Montpellier, France;
| | - Véronique Perrier
- INM, University of Montpellier, INSERM, CNRS, 34095 Montpellier, France
| | - Joan Torrent
- INM, University of Montpellier, INSERM, 34095 Montpellier, France; (A.V.); (L.S.-P.)
| |
Collapse
|
89
|
Ibanez L, Cruchaga C, Fernández MV. Advances in Genetic and Molecular Understanding of Alzheimer's Disease. Genes (Basel) 2021; 12:1247. [PMID: 34440421 PMCID: PMC8394321 DOI: 10.3390/genes12081247] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) has become a common disease of the elderly for which no cure currently exists. After over 30 years of intensive research, we have gained extensive knowledge of the genetic and molecular factors involved and their interplay in disease. These findings suggest that different subgroups of AD may exist. Not only are we starting to treat autosomal dominant cases differently from sporadic cases, but we could be observing different underlying pathological mechanisms related to the amyloid cascade hypothesis, immune dysfunction, and a tau-dependent pathology. Genetic, molecular, and, more recently, multi-omic evidence support each of these scenarios, which are highly interconnected but can also point to the different subgroups of AD. The identification of the pathologic triggers and order of events in the disease processes are key to the design of treatments and therapies. Prevention and treatment of AD cannot be attempted using a single approach; different therapeutic strategies at specific disease stages may be appropriate. For successful prevention and treatment, biomarker assays must be designed so that patients can be more accurately monitored at specific points during the course of the disease and potential treatment. In addition, to advance the development of therapeutic drugs, models that better mimic the complexity of the human brain are needed; there have been several advances in this arena. Here, we review significant, recent developments in genetics, omics, and molecular studies that have contributed to the understanding of this disease. We also discuss the implications that these contributions have on medicine.
Collapse
Affiliation(s)
- Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Maria Victoria Fernández
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| |
Collapse
|
90
|
van Eijk RPA, Nikolakopoulos S, Roes KCB, Kendall L, Han SS, Lavrov A, Epstein N, Kliest T, de Jongh AD, Westeneng HJ, Al-Chalabi A, Van Damme P, Hardiman O, Shaw PJ, McDermott CJ, Eijkemans MJC, van den Berg LH. Challenging the Established Order: Innovating Clinical Trials for Amyotrophic Lateral Sclerosis. Neurology 2021; 97:528-536. [PMID: 34315786 PMCID: PMC8456357 DOI: 10.1212/wnl.0000000000012545] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022] Open
Abstract
Development of effective treatments for amyotrophic lateral sclerosis (ALS) has been hampered by disease heterogeneity, a limited understanding of underlying pathophysiology, and methodologic design challenges. We have evaluated 2 major themes in the design of pivotal, phase 3 clinical trials for ALS—(1) patient selection and (2) analytical strategy—and discussed potential solutions with the European Medicines Agency. Several design considerations were assessed using data from 5 placebo-controlled clinical trials (n = 988), 4 population-based cohorts (n = 5,100), and 2,436 placebo-allocated patients from the Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) database. The validity of each proposed design modification was confirmed by means of simulation and illustrated for a hypothetical setting. Compared to classical trial design, the proposed design modifications reduce the sample size by 30.5% and placebo exposure time by 35.4%. By making use of prognostic survival models, one creates a potential to include a larger proportion of the population and maximize generalizability. We propose a flexible design framework that naturally adapts the trial duration when inaccurate assumptions are made at the design stage, such as enrollment or survival rate. In case of futility, the follow-up time is shortened and patient exposure to ineffective treatments or placebo is minimized. For diseases such as ALS, optimizing the use of resources, widening eligibility criteria, and minimizing exposure to futile treatments and placebo is critical to the development of effective treatments. Our proposed design modifications could circumvent important pitfalls and may serve as a blueprint for future clinical trials in this population.
Collapse
Affiliation(s)
- Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands. .,Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stavros Nikolakopoulos
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Kit C B Roes
- Department of Health Evidence, Section Biostatistics, Radboud Medical Centre Nijmegen, the Netherlands
| | | | - Steve S Han
- Neurosciences, Takeda Pharmaceuticals, Cambridge, USA.,Discovery Medicine, GlaxoSmithKline R&D, Upper Providence, USA
| | - Arseniy Lavrov
- Clinical Development, Novartis Gene Therapies, London, UK.,Clinical Translational Medicine, Future Pipeline Discovery, GlaxoSmithKline R&D, Middlesex, UK
| | - Noam Epstein
- Discovery Medicine, GlaxoSmithKline R&D, Upper Providence, USA
| | - Tessa Kliest
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Adriaan D de Jongh
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ammar Al-Chalabi
- King's College London, London, Maurice Wohl Clinical Neuroscience Institute and United Kingdom Dementia Research Institute Centre, Department of Basic and Clinical Neuroscience, UK.,Department of Neurology, King's College Hospital, London, UK
| | - Philip Van Damme
- Department of Neurosciences, Laboratory for Neurobiology, KU Leuven and Center for Brain & Disease Research, VIB, Leuven Brain Institute, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Orla Hardiman
- Department of Neurology, National Neuroscience Centre, Beaumont Hospital, Dublin, Ireland.,FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Pamela J Shaw
- Department of Neuroscience, University of Sheffield, Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - Christopher J McDermott
- Department of Neuroscience, University of Sheffield, Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - Marinus J C Eijkemans
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
91
|
A trial of gantenerumab or solanezumab in dominantly inherited Alzheimer's disease. Nat Med 2021; 27:1187-1196. [PMID: 34155411 PMCID: PMC8988051 DOI: 10.1038/s41591-021-01369-8] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Dominantly inherited Alzheimer's disease (DIAD) causes predictable biological changes decades before the onset of clinical symptoms, enabling testing of interventions in the asymptomatic and symptomatic stages to delay or slow disease progression. We conducted a randomized, placebo-controlled, multi-arm trial of gantenerumab or solanezumab in participants with DIAD across asymptomatic and symptomatic disease stages. Mutation carriers were assigned 3:1 to either drug or placebo and received treatment for 4-7 years. The primary outcome was a cognitive end point; secondary outcomes included clinical, cognitive, imaging and fluid biomarker measures. Fifty-two participants carrying a mutation were assigned to receive gantenerumab, 52 solanezumab and 40 placebo. Both drugs engaged their Aβ targets but neither demonstrated a beneficial effect on cognitive measures compared to controls. The solanezumab-treated group showed a greater cognitive decline on some measures and did not show benefits on downstream biomarkers. Gantenerumab significantly reduced amyloid plaques, cerebrospinal fluid total tau, and phospho-tau181 and attenuated increases of neurofilament light chain. Amyloid-related imaging abnormalities edema was observed in 19.2% (3 out of 11 were mildly symptomatic) of the gantenerumab group, 2.5% of the placebo group and 0% of the solanezumab group. Gantenerumab and solanezumab did not slow cognitive decline in symptomatic DIAD. The asymptomatic groups showed no cognitive decline; symptomatic participants had declined before reaching the target doses.
Collapse
|
92
|
Stoiljkovic M, Horvath TL, Hajós M. Therapy for Alzheimer's disease: Missing targets and functional markers? Ageing Res Rev 2021; 68:101318. [PMID: 33711510 PMCID: PMC8131215 DOI: 10.1016/j.arr.2021.101318] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
The development of the next generation therapy for Alzheimer's disease (AD) presents a huge challenge given the number of promising treatment candidates that failed in trials, despite recent advancements in understanding of genetic, pathophysiologic and clinical characteristics of the disease. This review reflects some of the most current concepts and controversies in developing disease-modifying and new symptomatic treatments. It elaborates on recent changes in the AD research strategy for broadening drug targets, and potentials of emerging non-pharmacological treatment interventions. Established and novel biomarkers are discussed, including emerging cerebrospinal fluid and plasma biomarkers reflecting tau pathology, neuroinflammation and neurodegeneration. These fluid biomarkers together with neuroimaging findings can provide innovative objective assessments of subtle changes in brain reflecting disease progression. A particular emphasis is given to neurophysiological biomarkers which are well-suited for evaluating the brain overall neural network integrity and function. Combination of multiple biomarkers, including target engagement and outcome biomarkers will empower translational studies and facilitate successful development of effective therapies.
Collapse
Affiliation(s)
- Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Pharmacology, University of Nis School of Medicine, Nis, Serbia.
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Mihály Hajós
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA; Cognito Therapeutics, Cambridge, MA, 02138, USA
| |
Collapse
|
93
|
Zannad F, Cotter G, Alonso Garcia A, George S, Davison B, Figtree G, Prasad K, Rockhold F, Schilsky RL, Stockbridge N, Pitt B, Butler J. What can heart failure trialists learn from oncology trialists? Eur Heart J 2021; 42:2373-2383. [PMID: 34076243 DOI: 10.1093/eurheartj/ehab236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/24/2021] [Accepted: 04/30/2021] [Indexed: 12/27/2022] Open
Abstract
Globally, there has been little change in mortality rates from cardiovascular (CV) diseases or cancers over the past two decades (1997-2018). This is especially true for heart failure (HF) where 5-year mortality rates remain as high as 45-55%. In the same timeframe, the proportion of drug revenue, and regulatory drug approvals for cancer drugs, far out paces those for CV drugs. In 2018, while cancer drugs made 27% of Food and Drug Administration drug approvals, only 1% of drug approvals was for a CV drug, and over this entire 20 year span, only four drugs were approved for HF in the USA. Cardiovascular trialists need to reassess the design, execution, and purpose of CV clinical trials. In the area of oncology research, trials are much smaller, follow-up is shorter, and targeted therapies are common. Cardiovascular diseases and cancer are the two most common causes of death globally, and although they differ substantially, this review evaluates whether some elements of oncology research may be applicable in the CV arena. As one of the most underserved CV diseases, the review focuses on aspects of cancer research that may be applicable to HF research with the aim of streamlining the clinical trial process and decreasing the time and cost required to bring safe, effective, treatments to patients who need them. The paper is based on discussions among clinical trialists, industry representatives, regulatory authorities, and patients, which took place at the Cardiovascular Clinical Trialists Workshop in Washington, DC, on 8 December 2019 (https://www.globalcvctforum.com/2019 (14 September 2020)).
Collapse
Affiliation(s)
- Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center 1439 at Institut Lorrain du Coeur et des Vaisseaux, CHU 54500, University Hospital of Nancy, Nancy, France
| | - Gad Cotter
- 2Momentum Research, Inc., 3100 Tower Blvd, Durham, NC, 27707, USA, Inserm, Paris, 942 Mascot, France
| | - Angeles Alonso Garcia
- Medicines and Healthcare products Regulatory Agency (MHRA), 10 South Colonnade, London, E14 4PU, UK
| | - Suzanne George
- Sarcoma Center, Dana-Farber Cancer Center, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Beth Davison
- 2Momentum Research, Inc., 3100 Tower Blvd, Durham, NC, 27707, USA, Inserm, Paris, 942 Mascot, France
| | - Gemma Figtree
- Northern Clinical School, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia, Reserve Road, St Leonards, NSW 2065
| | - Krishna Prasad
- Medicines and Healthcare products Regulatory Agency (MHRA), 10 South Colonnade, London, E14 4PU, UK
| | - Frank Rockhold
- Department of Biostatistics & Bioinformatics, Duke University Medical Center, 2424 Erwin Rd, Durham, NC, 27710, USA
| | | | - Norman Stockbridge
- Division of Cardiovascular and Renal Products, FDA Center for Drug Evaluation and Research (CDER), 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Bertram Pitt
- Division of Cardiology, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216, USA
| |
Collapse
|
94
|
Practical Considerations and Recommendations for Master Protocol Framework: Basket, Umbrella and Platform Trials. Ther Innov Regul Sci 2021; 55:1145-1154. [PMID: 34160785 PMCID: PMC8220876 DOI: 10.1007/s43441-021-00315-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/07/2021] [Indexed: 11/05/2022]
Abstract
Master protocol, categorized as basket trial, umbrella trial or platform trial, is an innovative clinical trial framework that aims to expedite clinical drug development, enhance trial efficiency, and eventually bring medicines to patients faster. Despite a clear uptake on the advantages in the concepts and designs, master protocols are still yet to be widely used. Part of that may be due to the fact that the master protocol framework comes with the need for new statistical designs and considerations for analyses and operational challenges. In this article, we provide an overview of the master protocol framework, unify the definitions with some examples, review the statistical methods for the designs and analyses, and focus our discussions on some practical considerations and recommendations of master protocols to help practitioners better design and implement such studies.
Collapse
|
95
|
Xiao X, Liu H, Liu X, Zhang W, Zhang S, Jiao B. APP, PSEN1, and PSEN2 Variants in Alzheimer's Disease: Systematic Re-evaluation According to ACMG Guidelines. Front Aging Neurosci 2021; 13:695808. [PMID: 34220489 PMCID: PMC8249733 DOI: 10.3389/fnagi.2021.695808] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/31/2021] [Indexed: 01/18/2023] Open
Abstract
The strategies of classifying APP, PSEN1, and PSEN2 variants varied substantially in the previous studies. We aimed to re-evaluate these variants systematically according to the American college of medical genetics and genomics and the association for molecular pathology (ACMG-AMP) guidelines. In our study, APP, PSEN1, and PSEN2 variants were collected by searching Alzforum and PubMed database with keywords “PSEN1,” “PSEN2,” and “APP.” These variants were re-evaluated based on the ACMG-AMP guidelines. We compared the number of pathogenic/likely pathogenic variants of APP, PSEN1, and PSEN2. In total, 66 APP variants, 323 PSEN1 variants, and 63 PSEN2 variants were re-evaluated in our study. 94.91% of previously reported pathogenic variants were re-classified as pathogenic/likely pathogenic variants, while 5.09% of them were variants of uncertain significance (VUS). PSEN1 carried the most prevalent pathogenic/likely pathogenic variants, followed by APP and PSEN2. Significant statistically difference was identified among these three genes when comparing the number of pathogenic/likely pathogenic variants (P < 2.2 × 10–16). Most of the previously reported pathogenic variants were re-classified as pathogenic/likely pathogenic variants while the others were re-evaluated as VUS, highlighting the importance of interpreting APP, PSEN1, and PSEN2 variants with caution according to ACMG-AMP guidelines.
Collapse
Affiliation(s)
- Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Sizhe Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
96
|
Fowler C, Rainey-Smith SR, Bird S, Bomke J, Bourgeat P, Brown BM, Burnham SC, Bush AI, Chadunow C, Collins S, Doecke J, Doré V, Ellis KA, Evered L, Fazlollahi A, Fripp J, Gardener SL, Gibson S, Grenfell R, Harrison E, Head R, Jin L, Kamer A, Lamb F, Lautenschlager NT, Laws SM, Li QX, Lim L, Lim YY, Louey A, Macaulay SL, Mackintosh L, Martins RN, Maruff P, Masters CL, McBride S, Milicic L, Peretti M, Pertile K, Porter T, Radler M, Rembach A, Robertson J, Rodrigues M, Rowe CC, Rumble R, Salvado O, Savage G, Silbert B, Soh M, Sohrabi HR, Taddei K, Taddei T, Thai C, Trounson B, Tyrrell R, Vacher M, Varghese S, Villemagne VL, Weinborn M, Woodward M, Xia Y, Ames D. Fifteen Years of the Australian Imaging, Biomarkers and Lifestyle (AIBL) Study: Progress and Observations from 2,359 Older Adults Spanning the Spectrum from Cognitive Normality to Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:443-468. [PMID: 34368630 PMCID: PMC8293663 DOI: 10.3233/adr-210005] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: The Australian Imaging, Biomarkers and Lifestyle (AIBL) Study commenced in 2006 as a prospective study of 1,112 individuals (768 cognitively normal (CN), 133 with mild cognitive impairment (MCI), and 211 with Alzheimer’s disease dementia (AD)) as an ‘Inception cohort’ who underwent detailed ssessments every 18 months. Over the past decade, an additional 1247 subjects have been added as an ‘Enrichment cohort’ (as of 10 April 2019). Objective: Here we provide an overview of these Inception and Enrichment cohorts of more than 8,500 person-years of investigation. Methods: Participants underwent reassessment every 18 months including comprehensive cognitive testing, neuroimaging (magnetic resonance imaging, MRI; positron emission tomography, PET), biofluid biomarkers and lifestyle evaluations. Results: AIBL has made major contributions to the understanding of the natural history of AD, with cognitive and biological definitions of its three major stages: preclinical, prodromal and clinical. Early deployment of Aβ-amyloid and tau molecular PET imaging and the development of more sensitive and specific blood tests have facilitated the assessment of genetic and environmental factors which affect age at onset and rates of progression. Conclusion: This fifteen-year study provides a large database of highly characterized individuals with longitudinal cognitive, imaging and lifestyle data and biofluid collections, to aid in the development of interventions to delay onset, prevent or treat AD. Harmonization with similar large longitudinal cohort studies is underway to further these aims.
Collapse
Affiliation(s)
- Christopher Fowler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| | - Sabine Bird
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Julia Bomke
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Pierrick Bourgeat
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Belinda M Brown
- Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Samantha C Burnham
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Ashley I Bush
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Carolyn Chadunow
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Steven Collins
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - James Doecke
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia.,Cooperative Research Council for Mental Health, Melbourne, VIC, Australia
| | - Vincent Doré
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Kathryn A Ellis
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, VIC, Australia.,Melbourne School of Psychological Sciences, Melbourne, VIC, Australia
| | - Lis Evered
- Department of Anaesthesia and Acute Pain Medicine, St Vincent's Hospital Melbourne, Victoria Parade, Fitzroy, VIC, Australia
| | - Amir Fazlollahi
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Jurgen Fripp
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Simon Gibson
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Robert Grenfell
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Elise Harrison
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Richard Head
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Liang Jin
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Adrian Kamer
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Fiona Lamb
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | | | - Simon M Laws
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Lucy Lim
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Yen Ying Lim
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Andrea Louey
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - S Lance Macaulay
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Lucy Mackintosh
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | | | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Simon McBride
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Madeline Peretti
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Kelly Pertile
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Morgan Radler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Alan Rembach
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne Robertson
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Rodrigues
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Rebecca Rumble
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | | | - Greg Savage
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Brendan Silbert
- Department of Anaesthesia and Acute Pain Medicine, St Vincent's Hospital Melbourne, Victoria Parade, Fitzroy, VIC, Australia
| | - Magdalene Soh
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Tania Taddei
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Christine Thai
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Brett Trounson
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Regan Tyrrell
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Michael Vacher
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Shiji Varghese
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Weinborn
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| | - Michael Woodward
- Department of Geriatric Medicine Austin Hospital, Heidelberg, VIC, Australia
| | - Ying Xia
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - David Ames
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, VIC, Australia.,National Ageing Research Institute (NARI), Parkville, VIC, Australia
| | | |
Collapse
|
97
|
Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, Ropele S, Fialova L, Kramberger M, Paulenka-Ivanovova N, Smisek M, Hanes J, Stevens E, Kovac A, Sutovsky S, Parrak V, Koson P, Prcina M, Galba J, Cente M, Hromadka T, Filipcik P, Piestansky J, Samcova M, Prenn-Gologranc C, Sivak R, Froelich L, Fresser M, Rakusa M, Harrison J, Hort J, Otto M, Tosun D, Ondrus M, Winblad B, Novak M, Zilka N. ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer's disease. NATURE AGING 2021; 1:521-534. [PMID: 37117834 DOI: 10.1038/s43587-021-00070-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/28/2021] [Indexed: 04/30/2023]
Abstract
Alzheimer's disease (AD) pathology is partly characterized by accumulation of aberrant forms of tau protein. Here we report the results of ADAMANT, a 24-month double-blinded, parallel-arm, randomized phase 2 multicenter placebo-controlled trial of AADvac1, an active peptide vaccine designed to target pathological tau in AD (EudraCT 2015-000630-30). Eleven doses of AADvac1 were administered to patients with mild AD dementia at 40 μg per dose over the course of the trial. The primary objective was to evaluate the safety and tolerability of long-term AADvac1 treatment. The secondary objectives were to evaluate immunogenicity and efficacy of AADvac1 treatment in slowing cognitive and functional decline. A total of 196 patients were randomized 3:2 between AADvac1 and placebo. AADvac1 was safe and well tolerated (AADvac1 n = 117, placebo n = 79; serious adverse events observed in 17.1% of AADvac1-treated individuals and 24.1% of placebo-treated individuals; adverse events observed in 84.6% of AADvac1-treated individuals and 81.0% of placebo-treated individuals). The vaccine induced high levels of IgG antibodies. No significant effects were found in cognitive and functional tests on the whole study sample (Clinical Dementia Rating-Sum of the Boxes scale adjusted mean point difference -0.360 (95% CI -1.306, 0.589)), custom cognitive battery adjusted mean z-score difference of 0.0008 (95% CI -0.169, 0.172). We also present results from exploratory and post hoc analyses looking at relevant biomarkers and clinical outcomes in specific subgroups. Our results show that AADvac1 is safe and immunogenic, but larger stratified studies are needed to better evaluate its potential clinical efficacy and impact on disease biomarkers.
Collapse
Affiliation(s)
- Petr Novak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia.
| | | | | | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Philip Scheltens
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Stefan Ropele
- Clinical Division of General Neurology, Department of Neurology, Medical University Graz, Graz, Austria
| | | | - Milica Kramberger
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | | | - Jozef Hanes
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Eva Stevens
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Andrej Kovac
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | | | - Peter Koson
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Michal Prcina
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | | | - Martin Cente
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Tomas Hromadka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | - Maria Samcova
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | | | - Roman Sivak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Lutz Froelich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, Medical Faculty Mannheim University of Heidelberg, Heidelberg, Germany
| | | | - Martin Rakusa
- Department of Neurological Diseases, University Medical Centre Maribor, Maribor, Slovenia
| | - John Harrison
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Matej Ondrus
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | | | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| |
Collapse
|
98
|
Cummings J, Bauzon J, Lee G. Who funds Alzheimer's disease drug development? ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12185. [PMID: 34095442 PMCID: PMC8145442 DOI: 10.1002/trc2.12185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Despite the increase in Alzheimer's disease (AD) cases in the United States, no new treatments have been approved in the United States since 2003. The costs associated with drug development programs are high and serve as a significant deterrent to AD therapeutic investigations. In this study, we analyze the sponsorship data for AD clinical trials conducted since 2016 to assess the fiscal support for AD clinical trials. METHODS We analyzed the funding sources of all AD trials over the past 5 years as reported on ClinicalTrials.gov. RESULTS There were 136 trials being conducted for treatments in the US AD therapeutic pipeline on the index date of this study. Among non-prevention trials, disease-modifying therapies (DMT) in Phase 3 were almost entirely sponsored by the biopharmaceutical industry; Phase 2 DMT trials were split between the biopharmaceutical industry and funding from the National Institutes of Health (NIH) to academic medical centers (AMCs). The majority of prevention trials received sponsorship from public-private partnerships (PPP). Trials of symptomatic agents are equally likely to have biopharmaceutical or NIH/AMC sponsorship. Most trials with repurposed agents had NIH/AMC funding (89%). Since 2016, there has been consistent growth in the number of trials sponsored both in part and fully by NIH/AMC sources and in PPP, and there has been a reduction in biopharmaceutical company-sponsored trials. DISCUSSION The number of trials supported by the biopharmaceutical industry has decreased over the past 5 years; trials supported from federal sources and PPP have increased. Repurposed compounds are mostly in Phase 2 trials and provide critical mechanistic information.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of NevadaLas Vegas (UNLV)Las VegasNevadaUSA
| | - Justin Bauzon
- University of NevadaLas Vegas (UNLV)School of MedicineLas VegasNevadaUSA
| | - Garam Lee
- Biogen, Medical AffairsWestonMassachusettsUSA
| |
Collapse
|
99
|
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer's disease. Lancet 2021; 397:1577-1590. [PMID: 33667416 PMCID: PMC8354300 DOI: 10.1016/s0140-6736(20)32205-4] [Citation(s) in RCA: 2047] [Impact Index Per Article: 511.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/21/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
In this Seminar, we highlight the main developments in the field of Alzheimer's disease. The most recent data indicate that, by 2050, the prevalence of dementia will double in Europe and triple worldwide, and that estimate is 3 times higher when based on a biological (rather than clinical) definition of Alzheimer's disease. The earliest phase of Alzheimer's disease (cellular phase) happens in parallel with accumulating amyloid β, inducing the spread of tau pathology. The risk of Alzheimer's disease is 60-80% dependent on heritable factors, with more than 40 Alzheimer's disease-associated genetic risk loci already identified, of which the APOE alleles have the strongest association with the disease. Novel biomarkers include PET scans and plasma assays for amyloid β and phosphorylated tau, which show great promise for clinical and research use. Multidomain lifestyle-based prevention trials suggest cognitive benefits in participants with increased risk of dementia. Lifestyle factors do not directly affect Alzheimer's disease pathology, but can still contribute to a positive outcome in individuals with Alzheimer's disease. Promising pharmacological treatments are poised at advanced stages of clinical trials and include anti-amyloid β, anti-tau, and anti-inflammatory strategies.
Collapse
Affiliation(s)
- Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Life Science Partners, Amsterdam, Netherlands.
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurology, Leuven, Belgium; Dementia Research Institute, University College London, London, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Ageing and Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Henne Holstege
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gael Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Groupement d'Intérêt Public Cyceron, Caen, France
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, University of Nevada, Las Vegas, NV, USA; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Wiesje M van der Flier
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Epidemiology and Datascience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
100
|
Perrone F, Cacace R, van der Zee J, Van Broeckhoven C. Emerging genetic complexity and rare genetic variants in neurodegenerative brain diseases. Genome Med 2021; 13:59. [PMID: 33853652 PMCID: PMC8048219 DOI: 10.1186/s13073-021-00878-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge of the molecular etiology of neurodegenerative brain diseases (NBD) has substantially increased over the past three decades. Early genetic studies of NBD families identified rare and highly penetrant deleterious mutations in causal genes that segregate with disease. Large genome-wide association studies uncovered common genetic variants that influenced disease risk. Major developments in next-generation sequencing (NGS) technologies accelerated gene discoveries at an unprecedented rate and revealed novel pathways underlying NBD pathogenesis. NGS technology exposed large numbers of rare genetic variants of uncertain significance (VUS) in coding regions, highlighting the genetic complexity of NBD. Since experimental studies of these coding rare VUS are largely lacking, the potential contributions of VUS to NBD etiology remain unknown. In this review, we summarize novel findings in NBD genetic etiology driven by NGS and the impact of rare VUS on NBD etiology. We consider different mechanisms by which rare VUS can act and influence NBD pathophysiology and discuss why a better understanding of rare VUS is instrumental for deriving novel insights into the molecular complexity and heterogeneity of NBD. New knowledge might open avenues for effective personalized therapies.
Collapse
Affiliation(s)
- Federica Perrone
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp – CDE, Universiteitsplein 1, BE-2610 Antwerp, Belgium
| | - Rita Cacace
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp – CDE, Universiteitsplein 1, BE-2610 Antwerp, Belgium
| | - Julie van der Zee
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp – CDE, Universiteitsplein 1, BE-2610 Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp – CDE, Universiteitsplein 1, BE-2610 Antwerp, Belgium
| |
Collapse
|