1
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
2
|
Perneczky R, Froelich L. Clinically meaningful benefit and real-world evidence in Alzheimer's disease research and care. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70090. [PMID: 40291121 PMCID: PMC12022225 DOI: 10.1002/trc2.70090] [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: 08/28/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
In the realm of medical research, assessing novel therapies extends beyond statistical significance. The concept of meaningful benefits plays a pivotal role in determining the practical impact of interventions on patient outcomes. Clinical trials, which form the bedrock of evidence-based medicine, guide treatment decisions and shape health-care practices. While statistical significance remains a fundamental criterion, it falls short in fully evaluating the clinical relevance of therapeutic interventions. Clinically meaningful benefits focus on tangible improvements in patient health and well-being, transcending mere statistical thresholds. Key considerations include survival rates, symptom relief, functional status, and other patient-oriented outcomes. Determining meaningful benefits varies across diseases, patient populations, and available treatments. Balancing statistical rigor with clinical relevance is crucial. Overpowered trials may detect smaller differences than anticipated, necessitating careful interpretation. Researchers must view trial results through a patient-centric lens. Beyond survival, evaluating quality of life and side effects is equally relevant. Quantifying meaningful benefits involves metrics like numbers needed to treat and progression-free survival. Consistency across outcomes matters, as clinicians weigh gains in survival against improvements in quality of life. The pursuit of meaningful benefits elevates clinical trials from mere statistical exercises to patient-centered endeavors. Researchers, clinicians, and regulators must prioritize outcomes that genuinely matter to patients, ensuring that medical progress translates into meaningful improvements for them and their families.
Collapse
Affiliation(s)
- Robert Perneczky
- Department of Psychiatry and PsychotherapyUniversity Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
- Ageing Epidemiology (AGE) Research Unit, School of Public HealthImperial College LondonLondonUK
- Sheffield Institute for Translational Neuroscience (SITraN), University of SheffieldSheffieldUK
- The International Registry for Alzheimer's Disease and Other Dementias Foundation (InRAD)DeventerThe Netherlands
| | - Lutz Froelich
- Department of Geriatric PsychiatryCentral Institute of Mental HealthMannheimGermany
| |
Collapse
|
3
|
Bosire EN, Blackmon K, Kamau LW, Udeh-Momoh C, Sokhi D, Shah J, Mbugua S, Muchungi K, Meier I, Narayan V, Nesic O, Merali Z. Healthcare providers perspectives and perceptions of dementia diagnosis and management at the Aga Khan University Hospital, Nairobi, Kenya. J Alzheimers Dis 2025; 104:862-874. [PMID: 40025713 DOI: 10.1177/13872877251320411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
BackgroundThe rising number of older people, including those living with Alzheimer's disease and related dementias (AD/ADRD) in sub-Saharan Africa (SSA) highlights the need for an improved clinical diagnosis and management of the diseases.ObjectiveTo understand and describe healthcare providers' perceptions and practices regarding AD/ADRD diagnosis and care in Kenya, not previously reported.MethodsThis was an ethnographic study involving observations and semi-structured interviews with healthcare providers working at the Aga Khan University Hospital, Nairobi (AKUHN) Kenya. Twenty-one healthcare providers were purposively recruited and interviewed in English, with the data transcribed verbatim and thematically analysed using Nvivo version 14.ResultsOur findings reveal that AKUHN's dementia diagnostic pathway aligns with universal best practice models and involves multidisciplinary care. Yet, healthcare providers noted that this level of care is not representative of most public hospitals in Kenya, where a lack of diagnostic equipment and trained staff severely limits patient access to timely dementia care. In addition, new medications that can slow AD/ADRD progression, are not readily available in Africa, including Kenya. We also identified barriers to timely diagnosis and care such as: lack of dementia policy and guidelines, limited expertise of healthcare providers, high cost of care, and sociocultural factors, including stigma.ConclusionsWe emphasize the need for the Kenyan government and relevant stakeholders to develop social and healthcare policies and allocate resources to raise awareness about dementia and combat stigma; train healthcare providers; improve early detection and service delivery through access to diagnostic tools, and establish clear guidelines/protocols for AD/ADRD care.
Collapse
Affiliation(s)
- Edna N Bosire
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Karen Blackmon
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Lucy W Kamau
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Chinedu Udeh-Momoh
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
- School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Dilraj Sokhi
- Neurology Department, Aga Khan University Hospital, Nairobi, Kenya
| | - Jasmit Shah
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Sylvia Mbugua
- Neurology Department, Aga Khan University Hospital, Nairobi, Kenya
| | - Kendi Muchungi
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Irene Meier
- Davos Alzheimer's Collaborative, Wayne, USA & Genève, Switzerland
| | - Vaibhav Narayan
- Davos Alzheimer's Collaborative, Wayne, USA & Genève, Switzerland
| | - Olivera Nesic
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Zul Merali
- Brain & Mind Institute, Aga Khan University, Nairobi, Kenya
| |
Collapse
|
4
|
Morehouse CR, Hendriks S, Rabbee N, Kim SY. Do Caregivers Value the New Antiamyloid Treatments for Alzheimer's Disease More Than Home-Based Care? Am J Geriatr Psychiatry 2025; 33:438-449. [PMID: 39709278 DOI: 10.1016/j.jagp.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE The new antiamyloid medications Lecanemab (Leqembi) and donanemab (Kisunla) are the first disease-modifying treatments for Alzheimer's disease (AD) to receive full FDA approval. However, some commentators question whether the drugs' benefits outweigh their risks, burdens, and costs to patients. This study assessed the perceived value of these medications by asking caregivers of persons with AD to compare them to a widely used intervention in AD management: home-based care. DESIGN Online survey (March 27th-April 17th, 2024) of 392 AD caregivers recruited via CloudResearch Prime Panels to match the U.S. public in education, household income, race, and ethnicity. The survey used vignettes describing antiamyloid medication and 25 hours/week of home-based care (estimated to be of similar monetary value). After rating the desirability of medication and home-based care, respondents indicated their preference for which intervention they wanted their loved one's insurance to cover. RESULTS Respondents expressed a desire for their loved ones to receive both the medication and home-based care. Over half (56.9%) favored home-based care coverage. Those preferring medication coverage were more likely to believe its benefits outweigh its risks and burdens. Preference for medication coverage was also associated with being male, Hispanic, less educated, and correctly answering fewer comprehension questions. CONCLUSIONS Our findings show most caregivers perceive modest clinical value in the novel antiamyloid therapies, and the decision to use these drugs will be preference-sensitive, pointing to the need for thorough informed consent discussions.
Collapse
Affiliation(s)
- Caroline R Morehouse
- Department of Bioethics (CRM, SH, SYK), Clinical Center, National Institutes of Health Clinical Center, Bethesda, MD
| | - Saskia Hendriks
- Department of Bioethics (CRM, SH, SYK), Clinical Center, National Institutes of Health Clinical Center, Bethesda, MD
| | - Nusrat Rabbee
- Biostatistics and Clinical Epidemiology Service (NR), Clinical Center, National Institutes of Health, Bethesda, MD
| | - Scott Yh Kim
- Department of Bioethics (CRM, SH, SYK), Clinical Center, National Institutes of Health Clinical Center, Bethesda, MD.
| |
Collapse
|
5
|
Antonioni A, Raho EM, Di Lorenzo F, Manzoli L, Flacco ME, Koch G. Blood phosphorylated Tau217 distinguishes amyloid-positive from amyloid-negative subjects in the Alzheimer's disease continuum. A systematic review and meta-analysis. J Neurol 2025; 272:252. [PMID: 40047958 PMCID: PMC11885345 DOI: 10.1007/s00415-025-12996-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia worldwide, and cost-effective tools to detect amyloid pathology are urgently needed. Blood-based Tau phosphorylated at threonine 217 (pTau217) seems promising, but its reliability as a proxy for cerebrospinal fluid (CSF) status and ability to identify patients within the AD spectrum remain unclear. METHODS We performed a systematic review and meta-analysis on the potential of blood pTau217 to differentiate amyloid-positive (A+) and amyloid-negative (A-) subjects. We included original studies reporting quantitative data on pTau217 concentrations in both blood and CSF in the AD continuum. The single-group meta-analysis computed the pooled pTau217 levels in blood and in CSF, separately in the A+ and A- groups, while the head-to-head meta-analysis compared the mean pTau217 concentrations in the A+ versus A- subjects, both in blood and CSF, stratifying by assessment method in both cases. RESULTS Ten studies (819 A+; 1055 A-) were included. The mean pTau217 levels resulted higher in CSF than in blood and, crucially, in A+ individuals than in A- ones, regardless of the laboratory method employed. Most importantly, all laboratory techniques reliably distinguished A+ from A- subjects, whether applied to CSF or blood samples. CONCLUSIONS These results confirm that blood-based pTau217 is a reliable marker of amyloid pathology with significant implications for clinical practice in the AD continuum. Indeed, pTau217 might be a non-invasive, scalable biomarker for early AD detection, reducing the reliance on more invasive, expansive, and less accessible methods. CLINICAL TRIAL REGISTRATION Prospero CRD42024565187.
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, Department of Neuroscience and Rehabilitation, University of Ferrara, Via Ludovico Ariosto, 35, 44121, Ferrara, Italy.
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy.
| | - Emanuela Maria Raho
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy
| | - Francesco Di Lorenzo
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
| | - Lamberto Manzoli
- Department of Medical and Surgical Sciences, University of Bologna, 40126, Bologna, Italy
| | - Maria Elena Flacco
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giacomo Koch
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121, Ferrara, Italy.
| |
Collapse
|
6
|
Maher P, Christopher R, Evans R, Raschke W. Safety, Pharmacokinetics, and Cardiodynamics of CMS121, a Novel Small Molecule Fisetin Derivative with Neuroprotective Properties, in Phase 1 Healthy Adult Volunteers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.28.25323123. [PMID: 40093210 PMCID: PMC11908328 DOI: 10.1101/2025.02.28.25323123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The safety, tolerability, pharmacokinetics, age-related effects of single (SD) and repeat (RD) doses of CMS121, a novel small molecule fisetin derivative, were evaluated in healthy adult volunteers. The effects of food were also evaluated in healthy young adult subjects. SD of up to 1800 mg or RD up to 900 mg/day for 7 days was generally well tolerated, with the majority of TEAEs mild in severity. Generally, the pharmacokinetics of CMS121 and its metabolites were well characterized and increased in a dose-proportional or slightly greater than dose-proportional manner across the range of doses assessed. CMS121-C2 metabolite appears to contribute the most to the presence of the molar-equivalent CMS121 in plasma than the parent compound or the other metabolites (i.e. CMS121-C1 and CMS121-C3). Urinary excretion of CMS121 metabolites was minimal, implying urinary excretion may not be a major clearance route by which CMS121 is eliminated after oral dosing. There is a significant effect of age on the pharmacokinetics of CMS121 and its metabolites, with higher systemic exposures to CMS121 and its metabolites and longer terminal elimination half-lives in elderly subjects. Systemic exposures to CMS121 were higher in the fed state by approximately 50%.
Collapse
|
7
|
Guan DX, Peters ME, Pike GB, Ballard C, Creese B, Corbett A, Pickering E, Roach P, Smith EE, Ismail Z. Cognitive, Behavioral, and Functional Outcomes of Suspected Mild Traumatic Brain Injury in Community-Dwelling Older Persons Without Mild Cognitive Impairment or Dementia. J Acad Consult Liaison Psychiatry 2025; 66:118-129. [PMID: 39746450 DOI: 10.1016/j.jaclp.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/29/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Traumatic brain injury is associated with greater risk and earlier onset of dementia. OBJECTIVE This study investigated whether later-life changes in subjective cognition and behavior - potential markers of Alzheimer disease - could be observed in cognitively unimpaired older persons with a history of suspected mild traumatic brain injury (smTBI) earlier in life and whether changes in cognition and behavior mediated the link between smTBI and daily function. METHODS Data for 1392 participants from the Canadian Platform for Research Online to Investigate Health, Quality of Life, Cognition, Behaviour, Function, and Caregiving in Aging were analyzed. A validated self-reported brain injury screening questionnaire was used to determine the history of smTBI. Outcomes were measured using the Everyday Cognition scale (for subjective cognitive decline [SCD]), Mild Behavioral Impairment (MBI) Checklist, and Standard Assessment of Global Everyday Activities (for function). Inverse probability of treatment weighted logistic and negative binomial regressions were used to model smTBI (exposure) associations with SCD and MBI statuses, and Everyday Cognition-II and MBI Checklist total scores, respectively. Mediation analyses were conducted using bootstrapping. RESULTS History of smTBI was linked to higher odds of SCD (odds ratio = 1.45, 95% confidence interval: [1.14-1.84]) or MBI (odds ratio = 1.75, 95% confidence interval: [1.54-1.98]), as well as 24% (95% confidence interval: [18%-31%]) higher Everyday Cognition-II and 52% (95% confidence interval: [41%-63%]) higher MBI Checklist total scores. Finally, SCD and MBI mediated approximately 45% and 56%, respectively, of the association between smTBI history and poorer function, as indicated by higher Standard Assessment of Global Everyday Activities total scores. CONCLUSIONS smTBI at any point in the life course is linked to poorer cognition and behavior even in community-dwelling older persons without MCI or dementia. Older persons with smTBI may benefit from early dementia risk assessment using tools that measure changes in cognition and behavior. Interventions for declining cognition and behavior may also be beneficial in this population to address functional impairment.
Collapse
Affiliation(s)
- Dylan X Guan
- Graduate Science Education, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Matthew E Peters
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - G Bruce Pike
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada; Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Clive Ballard
- Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Exeter, England, UK
| | - Byron Creese
- Department of Psychiatry, College of Health Medicine and Life Sciences, Brunel University, London, England, UK
| | - Anne Corbett
- Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Exeter, England, UK
| | - Ellie Pickering
- Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Exeter, England, UK
| | - Pamela Roach
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Family Medicine, University of Calgary, Calgary, AB, Canada; Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada; O'Brien Institute for Public Health, University of Calgary, Calgary, AB, Canada
| | - Eric E Smith
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada; Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Zahinoor Ismail
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada; Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Exeter, England, UK; Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada; O'Brien Institute for Public Health, University of Calgary, Calgary, AB, Canada; Department of Psychiatry, University of Calgary, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
8
|
Cummings JL, Atri A, Feldman HH, Hansson O, Sano M, Knop FK, Johannsen P, León T, Scheltens P. evoke and evoke+: design of two large-scale, double-blind, placebo-controlled, phase 3 studies evaluating efficacy, safety, and tolerability of semaglutide in early-stage symptomatic Alzheimer's disease. Alzheimers Res Ther 2025; 17:14. [PMID: 39780249 PMCID: PMC11708093 DOI: 10.1186/s13195-024-01666-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Disease-modifying therapies targeting the diverse pathophysiology of Alzheimer's disease (AD), including neuroinflammation, represent potentially important and novel approaches. The glucagon-like peptide-1 receptor agonist semaglutide is approved for the treatment of type 2 diabetes and obesity and has an established safety profile. Semaglutide may have a disease-modifying, neuroprotective effect in AD through multimodal mechanisms including neuroinflammatory, vascular, and other AD-related processes. Large randomized controlled trials are needed to assess the efficacy and safety of semaglutide in early-stage symptomatic AD. METHODS evoke and evoke+ are randomized, double-blind, placebo-controlled phase 3 trials investigating the efficacy, safety, and tolerability of once-daily oral semaglutide versus placebo in early-stage symptomatic AD. Eligible participants were men or women aged 55-85 years with mild cognitive impairment or mild dementia due to AD with confirmed amyloid abnormalities (assessed by positron emission tomography or cerebrospinal fluid [CSF] analysis). After a maximum 12-week screening phase, an anticipated 1840 patients in each trial are randomized (1:1) to semaglutide or placebo for 156 weeks (104-week main treatment phase and 52-week extension). Randomized participants follow an 8-week dose escalation regimen (3 mg [weeks 0-4], 7 mg [weeks 4-8], and 14 mg [weeks 8-156]). The primary endpoint is the semaglutide-placebo difference on change from baseline to week 104 in the Clinical Dementia Rating - Sum of Boxes score. Analyses of plasma biomarkers, collected from all participants, and a CSF sub-study (planned n = 210) will explore semaglutide effects on AD biomarkers and neuroinflammation. RESULTS Enrollment was undertaken between May 18, 2021, and September 8, 2023. Completion of the trials' main phase is expected in September 2025, and the 52-week extension (in which participants and investigators remain blinded to treatment assignment) will continue to October 2026. CONCLUSION evoke and evoke+ are the first large-scale trials to investigate the disease-modifying potential of semaglutide in participants with early-stage symptomatic AD, including exploration of effects on AD biomarkers and neuroinflammation. The trials will provide data on the potential disease-modifying effects of semaglutide and will be important in evaluating its utility in the treatment of early-stage symptomatic AD. TRIAL REGISTRATION Clinicaltrials.gov, NCT04777396 and NCT04777409. Date: 02/03/2021.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, NV, USA.
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, USA.
| | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, AZ, USA
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Mary Sano
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | | | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- EQT Life Sciences, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Clevenger CK, Lingler JH, Zhang Y, Seleri S, Parnas ML, Youmans-Kidder K. Role of nurse practitioners in comprehensive Alzheimer's disease care: Barriers and opportunities for timely diagnosis. Geriatr Nurs 2025; 61:400-407. [PMID: 39673910 DOI: 10.1016/j.gerinurse.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. Treatments include disease-modifying therapies (DMTs), which studies showed are most effective when initiated during the early disease stages. Timely AD diagnosis is therefore important, as DMTs can potentially extend an acceptable quality of life for people with this condition. This scoping review presents the current role of nurse practitioners (NPs) in AD care and describe the current and potential future role NPs can play in timely AD diagnosis and management. A systematic search of the PubMed and CINAHL databases identified 15 relevant articles. The literature review revealed that NPs play an active role in the initial assessment of cognitive disorders; however, there is a lack of evidence for NPs acting autonomously to diagnose and treat AD. Several opportunities to increase the autonomous role of NPs are described, which could reduce barriers to timely AD diagnosis and management.
Collapse
Affiliation(s)
- Carolyn K Clevenger
- Emory University Nell Hodgson Woodruff School of Nursing, 1520 Clifton Rd, Atlanta, GA 30322, USA.
| | - Jennifer H Lingler
- University of Pittsburgh School of Nursing, 3500 Victoria St, Pittsburgh, PA 15213, USA
| | - Yuchen Zhang
- University of Pittsburgh School of Nursing, 3500 Victoria St, Pittsburgh, PA 15213, USA
| | - Sheila Seleri
- Genentech, a member of the Roche Group, 1 DNA Way, South San Francisco, CA 94080, USA
| | - M Laura Parnas
- Roche Diagnostics Corporation, 9115 Hague Rd, Indianapolis, IN 46256, USA
| | | |
Collapse
|
10
|
Huijbers W, Pinter NK, Spaltman M, Cornelis M, Schmand B, Alnaji B, Yargeau M, Harlock S, Dorn RP, Ajtai B, Westphal ES, van Elswijk G. Clinical validity of IntelliSpace Cognition digital assessment platform in mild cognitive impairment. Front Psychol 2024; 15:1451843. [PMID: 39807355 PMCID: PMC11726315 DOI: 10.3389/fpsyg.2024.1451843] [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: 06/19/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
We evaluated a digital cognitive assessment platform, Philips IntelliSpace Cognition, in a case-control study of patients diagnosed with mild cognitive impairment (MCI) and cognitively normal (CN) older adults. Performance on individual neuropsychological tests, cognitive z-scores, and Alzheimer's disease (AD)-specific composite scores was compared between the CN and MCI groups. These groups were matched for age, sex, and education. Performance on all but two neuropsychological tests was worse in the MCI group. After ranking the cognitive scores by effect size, we found that the memory score was the most impaired, followed by executive functioning. The Early AD/MCI Alzheimer's Cognitive Composite (EMACC) and Preclinical Alzheimer's Cognitive Composite (PACC) scores were constructed from the digital tests on Philips IntelliSpace Cognition. Both AD-specific composite scores showed greater sensitivity and specificity than the Mini-Mental State Examination or individual cognitive z-scores. Together, these results demonstrate the diagnostic value of Philips IntelliSpace Cognition in patients with MCI.
Collapse
Affiliation(s)
| | - Nandor K. Pinter
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | | | - Mike Cornelis
- Digital Cognitive Dx, Philips, Eindhoven, Netherlands
| | - Ben Schmand
- Faculty of Social and Behavioral Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Baraa Alnaji
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | | | - Sarah Harlock
- Dent Neurologic Institute, Amherst, NY, United States
| | - Ryu Platinum Dorn
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Bela Ajtai
- Dent Neurologic Institute, Amherst, NY, United States
| | | | | |
Collapse
|
11
|
Tumati S, Herrmann N, Perin J, Rosenberg PB, Lerner AJ, Mintzer J, Padala PR, Brawman-Mintzer O, van Dyck CH, Porsteinsson AP, Craft S, Levey A, Shade D, Lanctôt KL. Measuring clinically relevant change in apathy symptoms in ADMET and ADMET 2. Int Psychogeriatr 2024; 36:1232-1244. [PMID: 39297292 PMCID: PMC11695175 DOI: 10.1017/s1041610224000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES Among participants with Alzheimer's disease (AD) we estimated the minimal clinically important difference (MCID) in apathy symptom severity on three scales. DESIGN Retrospective anchor- and distribution-based analyses of change in apathy symptom scores. SETTING Apathy in Dementia Methylphenidate Trial (ADMET) and ADMET 2 randomized controlled trials conducted at three and ten clinics specialized in dementia care in United States and Canada, respectively. PARTICIPANTS Two hundred and sixty participants (60 ADMET, 200 ADMET 2) with clinically significant apathy in Alzheimer's disease. MEASUREMENTS The Clinical Global Impression of Change in Apathy scale was used as the anchor measure and the MCID on the Neuropsychiatric Inventory - Apathy (NPI-A), Dementia Apathy Interview and Rating (DAIR), and Apathy Evaluation Scale-Informant (AES-I) were estimated with linear mixed models across all study visits. The estimated thresholds were evaluated with performance metrics. RESULTS Among the MCID was a decrease of four points (95% CI: -4.0 to -4.8) on the NPI-A, 0.56 points (95% CI: -0.47 to -0.65) on the DAIR, and three points on the AES-I (95% CI: -0.9 to -5.4). Distribution-based analyses were largely consistent with the anchor-based analyses. The MCID across the three measures showed ∼60% accuracy. Sensitivity analyses found that MMSE scores and apathy severity at baseline influenced the estimated MCID. CONCLUSIONS MCIDs for apathy on three scales will help evaluate treatment efficacy at the individual level. However, the modest correspondence between MCID and clinical impression of change suggests the need to consider other scales.
Collapse
Affiliation(s)
- Shankar Tumati
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario
| | - Nathan Herrmann
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario
| | - Jaime Perin
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Alan J. Lerner
- University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Jacobo Mintzer
- Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina
| | - Prasad R. Padala
- Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Olga Brawman-Mintzer
- Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina
| | | | | | - Suzanne Craft
- Wake Forest University, Winston-Salem, North Carolina
| | - Allan Levey
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, Georgia
| | - David Shade
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Krista L. Lanctôt
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Ontario
| |
Collapse
|
12
|
Ito D, Okada K. Rethinking antisense oligonucleotide therapeutics for amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2024; 11:3054-3063. [PMID: 39473221 DOI: 10.1002/acn3.52234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024] Open
Abstract
Antisense oligonucleotides, which are used to silence target genes, are gaining attention as a novel drug discovery modality for proteinopathies. However, while clinical trials for neurodegenerative diseases like amyotrophic lateral sclerosis have been conducted in recent years, the results have not always been favorable. The results from a Phase III trial of the antisense oligonucleotide, that is, tofersen, which targets SOD1 mRNA, showed decreased levels of cerebrospinal fluid SOD1 and plasma neurofilament light chain but no improvements in primary clinical endpoint. Moreover, case reports pertaining to patients with amyotrophic lateral sclerosis carrying FUS and C9orf72 mutations who received antisense oligonucleotide-based treatments have demonstrated a notable reduction in the targeted protein (thus providing the proof of mechanism) but with no discernible clinical benefits. There are several possible reasons why antisense oligonucleotides knockdown fails to achieve proof of concept, which need to be addressed: on-target adverse effects resulting from the loss of function of target gene and irreversible neuronal death cascade due to toxic protein accumulation, among other factors. This review provides an overview of the current status and discusses the prospects of antisense oligonucleotides treatment for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Daisuke Ito
- Memory Center, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Kamatham PT, Shukla R, Khatri DK, Vora LK. Pathogenesis, diagnostics, and therapeutics for Alzheimer's disease: Breaking the memory barrier. Ageing Res Rev 2024; 101:102481. [PMID: 39236855 DOI: 10.1016/j.arr.2024.102481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for 60-70 % of all cases. It affects millions of people worldwide. AD poses a substantial economic burden on societies and healthcare systems. AD is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. As the prevalence of AD continues to increase, understanding its pathogenesis, improving diagnostic methods, and developing effective therapeutics have become paramount. This comprehensive review delves into the intricate mechanisms underlying AD, explores the current state of diagnostic techniques, and examines emerging therapeutic strategies. By revealing the complexities of AD, this review aims to contribute to the growing body of knowledge surrounding this devastating disease.
Collapse
Affiliation(s)
- Pushpa Tryphena Kamatham
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rashi Shukla
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland BT9 7BL, UK.
| |
Collapse
|
14
|
Thawabteh AM, Ghanem AW, AbuMadi S, Thaher D, Jaghama W, Karaman D, Karaman R. Recent Advances in Therapeutics for the Treatment of Alzheimer's Disease. Molecules 2024; 29:5131. [PMID: 39519769 PMCID: PMC11547905 DOI: 10.3390/molecules29215131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The most prevalent chronic neurodegenerative illness in the world is Alzheimer's disease (AD). It results in mental symptoms including behavioral abnormalities and cognitive impairment, which have a substantial financial and psychological impact on the relatives of the patients. The review discusses various pathophysiological mechanisms contributing to AD, including amyloid beta, tau protein, inflammation, and other factors, while emphasizing the need for effective disease-modifying therapeutics that alter disease progression rather than merely alleviating symptoms. This review mainly covers medications that are now being studied in clinical trials or recently approved by the FDA that fall under the disease-modifying treatment (DMT) category, which alters the progression of the disease by targeting underlying biological mechanisms rather than merely alleviating symptoms. DMTs focus on improving patient outcomes by slowing cognitive decline, enhancing neuroprotection, and supporting neurogenesis. Additionally, the review covers amyloid-targeting therapies, tau-targeting therapies, neuroprotective therapies, and others. This evaluation specifically looked at studies on FDA-approved novel DMTs in Phase II or III development that were carried out between 2021 and 2024. A thorough review of the US government database identified clinical trials of biologics and small molecule drugs for 14 agents in Phase I, 34 in Phase II, and 11 in Phase III that might be completed by 2028.
Collapse
Affiliation(s)
- Amin Mahmood Thawabteh
- Department of Chemistry, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine;
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Aseel Wasel Ghanem
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Sara AbuMadi
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Dania Thaher
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Weam Jaghama
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Donia Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
15
|
Koychev I, Reid G, Nguyen M, Mentz RJ, Joyce D, Shah SH, Holman RR. Inflammatory proteins associated with Alzheimer's disease reduced by a GLP1 receptor agonist: a post hoc analysis of the EXSCEL randomized placebo controlled trial. Alzheimers Res Ther 2024; 16:212. [PMID: 39358806 PMCID: PMC11448378 DOI: 10.1186/s13195-024-01573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists are a viable option for the prevention of Alzheimer's disease (AD) but the mechanisms of this potential disease modifying action are unclear. We investigated the effects of once-weekly exenatide (EQW) on AD associated proteomic clusters. METHODS The Exenatide Study of Cardiovascular Event Lowering study compared the cardiovascular effects of EQW 2 mg with placebo in 13,752 people with type 2 diabetes mellitus. 4,979 proteins were measured (Somascan V0.4) on baseline and 1-year plasma samples of 3,973 participants. C-reactive protein (CRP), ficolin-2 (FCN2), plasminogen activator inhibitor 1 (PAI-1), soluble vascular cell adhesion protein 1 (sVCAM1) and 4 protein clusters were tested in multivariable mixed models. RESULTS EQW affected FCN2 (Cohen's d -0.019), PAI-1 (Cohen's d -0.033), sVCAM-1 (Cohen's d 0.035) and a cytokine-cytokine cluster (Cohen's d 0.037) significantly compared with placebo. These effects were sustained in individuals over the age of 65 but not in those under 65. CONCLUSIONS EQW treatment was associated with significant change in inflammatory proteins associated with AD. TRIAL REGISTRATION EXSCEL is registered on ClinicalTrials.gov: NCT01144338 on 10th of June 2010.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK.
| | - Graham Reid
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Maggie Nguyen
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
| | | | - Dan Joyce
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Svati H Shah
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Jin M, Noble JM. What's in It for Me? Contextualizing the Potential Clinical Impacts of Lecanemab, Donanemab, and Other Anti-β-amyloid Monoclonal Antibodies in Early Alzheimer's Disease. eNeuro 2024; 11:ENEURO.0088-24.2024. [PMID: 39332901 PMCID: PMC11439562 DOI: 10.1523/eneuro.0088-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024] Open
Abstract
A new era of disease-modifying therapy for Alzheimer's disease (AD) arrived in 2021 following the Food and Drug Administration's (FDA) decision to grant accelerated approval for aducanumab, an anti-β-amyloid (Aβ) monoclonal antibody designed to target Aβ aggregates, a biological component of AD. More recently, trial outcomes for lecanemab and donanemab, two additional antibodies of this drug class, have shown favorable and significant slowing of metrics for cognitive and functional decline. Lecanemab and donanemab have since received similar FDA approval to aducanumab in January 2023 and July 2024, respectively. Given that these therapies are a clearly emerging tool in the repertoire of clinicians treating AD and related dementias, a critical dialogue has been ongoing regarding the potential impacts and place for these therapies. Here, we seek to contextualize this debate by first considering factors involved in theoretically extrapolating current randomized control trial outcomes to estimate meaningful clinical impacts. In the process of this exercise, we outline a generally useful concept termed Summative Treatment-Associated Benefit measuring Long-term Efficacy/Effectiveness Area as a metric of summative benefits of treatment over the life course of an individual. Second, we consider current real-world factors, such as conditions of FDA approval and other points involved in clinical decision-making that will influence and/or temper the actual impacts of this drug class.
Collapse
Affiliation(s)
- Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, New York 10032
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, New York 10027
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and the GH Sergievsky Center, Columbia University Irving Medical Center, New York, New York 10032
| |
Collapse
|
17
|
Lerch O, Ferreira D, Stomrud E, van Westen D, Tideman P, Palmqvist S, Mattsson-Carlgren N, Hort J, Hansson O, Westman E. Predicting progression from subjective cognitive decline to mild cognitive impairment or dementia based on brain atrophy patterns. Alzheimers Res Ther 2024; 16:153. [PMID: 38970077 PMCID: PMC11225196 DOI: 10.1186/s13195-024-01517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 06/24/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder where pathophysiological changes begin decades before the onset of clinical symptoms. Analysis of brain atrophy patterns using structural MRI and multivariate data analysis are an effective tool in identifying patients with subjective cognitive decline (SCD) at higher risk of progression to AD dementia. Atrophy patterns obtained from models trained to classify advanced AD versus normal subjects, may not be optimal for subjects at an early stage, like SCD. In this study, we compared the accuracy of the SCD progression prediction using the 'severity index' generated using a standard classification model trained on patients with AD dementia versus a new model trained on β-amyloid (Aβ) positive patients with amnestic mild cognitive impairment (aMCI). METHODS We used structural MRI data of 504 patients from the Swedish BioFINDER-1 study cohort (cognitively normal (CN), Aβ-negative = 220; SCD, Aβ positive and negative = 139; aMCI, Aβ-positive = 106; AD dementia = 39). We applied multivariate data analysis to create two predictive models trained to discriminate CN individuals from either individuals with Aβ positive aMCI or AD dementia. Models were applied to individuals with SCD to classify their atrophy patterns as either high-risk "disease-like" or low-risk "CN-like". Clinical trajectory and model accuracy were evaluated using 8 years of longitudinal data. RESULTS In predicting progression from SCD to MCI or dementia, the standard, dementia-based model, reached 100% specificity but only 10.6% sensitivity, while the new, aMCI-based model, reached 72.3% sensitivity and 60.9% specificity. The aMCI-based model was superior in predicting progression from SCD to MCI or dementia, reaching a higher receiver operating characteristic area under curve (AUC = 0.72; P = 0.037) in comparison with the dementia-based model (AUC = 0.57). CONCLUSION When predicting conversion from SCD to MCI or dementia using structural MRI data, prediction models based on individuals with milder levels of atrophy (i.e. aMCI) may offer superior clinical value compared to standard dementia-based models.
Collapse
Affiliation(s)
- Ondrej Lerch
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, 15006, Czech Republic.
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, 14183, Sweden.
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, 14183, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, 55902, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, 20502, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 21428, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Institution for Clinical Sciences Lund, Lund University, Lund, 22184, Sweden
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, 20502, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 21428, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, 20502, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 21428, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, 20502, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 21428, Sweden
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, 15006, Czech Republic
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, 20502, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 21428, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, 14183, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE58AF, UK
| |
Collapse
|
18
|
Leroi I, Dolphin H, Dinh R, Foley T, Kennelly S, Kinchin I, O'Caoimh R, O'Dowd S, O'Philbin L, O'Reilly S, Trepel D, Timmons S. Navigating the future of Alzheimer's care in Ireland - a service model for disease-modifying therapies in small and medium-sized healthcare systems. BMC Health Serv Res 2024; 24:705. [PMID: 38840115 PMCID: PMC11151472 DOI: 10.1186/s12913-024-11019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/19/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND A new class of antibody-based drug therapy with the potential for disease modification is now available for Alzheimer's disease (AD). However, the complexity of drug eligibility, administration, cost, and safety of such disease modifying therapies (DMTs) necessitates adopting new treatment and care pathways. A working group was convened in Ireland to consider the implications of, and health system readiness for, DMTs for AD, and to describe a service model for the detection, diagnosis, and management of early AD in the Irish context, providing a template for similar small-medium sized healthcare systems. METHODS A series of facilitated workshops with a multidisciplinary working group, including Patient and Public Involvement (PPI) members, were undertaken. This informed a series of recommendations for the implementation of new DMTs using an evidence-based conceptual framework for health system readiness based on [1] material resources and structures and [2] human and institutional relationships, values, and norms. RESULTS We describe a hub-and-spoke model, which utilises the existing dementia care ecosystem as outlined in Ireland's Model of Care for Dementia, with Regional Specialist Memory Services (RSMS) acting as central hubs and Memory Assessment and Support Services (MASS) functioning as spokes for less central areas. We provide criteria for DMT referral, eligibility, administration, and ongoing monitoring. CONCLUSIONS Healthcare systems worldwide are acknowledging the need for advanced clinical pathways for AD, driven by better diagnostics and the emergence of DMTs. Despite facing significant challenges in integrating DMTs into existing care models, the potential for overcoming challenges exists through increased funding, resources, and the development of a structured national treatment network, as proposed in Ireland's Model of Care for Dementia. This approach offers a replicable blueprint for other healthcare systems with similar scale and complexity.
Collapse
Affiliation(s)
- Iracema Leroi
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland.
- Global Brain Health Institute, Dublin, Ireland.
- HRB-CTN Dementia Trials Ireland, Dublin, Ireland.
| | - Helena Dolphin
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Rachel Dinh
- Centre for Global Health, Trinity College Dublin, Dublin, Ireland
| | - Tony Foley
- Department of General Practice, School of Medicine, University College Cork, Cork, Ireland
| | - Sean Kennelly
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- Global Brain Health Institute, Dublin, Ireland
- HRB-CTN Dementia Trials Ireland, Dublin, Ireland
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Irina Kinchin
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- Global Brain Health Institute, Dublin, Ireland
| | - Rónán O'Caoimh
- HRB-CTN Dementia Trials Ireland, Dublin, Ireland
- Department of Geriatric Medicine, Mercy University Hospital, Cork, Ireland
| | - Sean O'Dowd
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Health Service Executive's National Dementia Office, Dublin, Ireland
| | | | | | - Dominic Trepel
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- Global Brain Health Institute, Dublin, Ireland
| | - Suzanne Timmons
- Global Brain Health Institute, School of Medicine, Trinity College Dublin, Lloyd Building, Dublin 2, Dublin, Ireland
- HRB-CTN Dementia Trials Ireland, Dublin, Ireland
- Centre for Gerontology and Rehabilitation, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Naude J, Wang M, Leon R, Smith E, Ismail Z. Tau-PET in early cortical Alzheimer brain regions in relation to mild behavioral impairment in older adults with either normal cognition or mild cognitive impairment. Neurobiol Aging 2024; 138:19-27. [PMID: 38490074 DOI: 10.1016/j.neurobiolaging.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/17/2024]
Abstract
Mild Behavioral Impairment (MBI) leverages later-life emergent and persistent neuropsychiatric symptoms (NPS) to identify a high-risk group for incident dementia. Phosphorylated tau (p-tau) is a hallmark biological manifestation of Alzheimer disease (AD). We investigated associations between MBI and tau accumulation in early-stage AD cortical regions. In 442 Alzheimer's Disease Neuroimaging Initiative participants with normal cognition or mild cognitive impairment, MBI status was determined alongside corresponding p-tau and Aβ. Two meta-regions of interest were generated to represent Braak I and III neuropathological stages. Multivariable linear regression modelled the association between MBI as independent variable and tau tracer uptake as dependent variable. Among Aβ positive individuals, MBI was associated with tau uptake in Braak I (β=0.45(0.15), p<.01) and Braak III (β=0.24(0.07), p<.01) regions. In Aβ negative individuals, MBI was not associated with tau in the Braak I region (p=0.11) with a negative association in Braak III (p=.01). These findings suggest MBI may be a sequela of neurodegeneration, and can be implemented as a cost-effective framework to help improve screening efficiency for AD.
Collapse
Affiliation(s)
- James Naude
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Meng Wang
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Rebeca Leon
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Eric Smith
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Zahinoor Ismail
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada; Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
20
|
Wahlberg K, Winblad B, Cole A, Herring WL, Ramsberg J, Torontali I, Visser PJ, Wimo A, Wollaert L, Jönsson L. People get ready! A new generation of Alzheimer's therapies may require new ways to deliver and pay for healthcare. J Intern Med 2024; 295:281-291. [PMID: 38098165 DOI: 10.1111/joim.13759] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
The development of disease-modifying therapies (DMTs) for Alzheimer's disease (AD) has progressed over the last decade, and the first-ever therapies with potential to slow the progression of disease are approved in the United States. AD DMTs could provide life-changing opportunities for people living with this disease, as well as for their caregivers. They could also ease some of the immense societal and economic burden of dementia. However, AD DMTs also come with major challenges due to the large unmet medical need, high prevalence of AD, new costs related to diagnosis, treatment and monitoring, and uncertainty in the therapies' actual clinical value. This perspective article discusses, from the broad perspective of various health systems and stakeholders, how we can overcome these challenges and improve society's readiness for AD DMTs. We propose that innovative payment models such as performance-based payments, in combination with learning healthcare systems, could be the way forward to enable timely patient access to treatments, improve accuracy of cost-effectiveness evaluations and overcome budgetary barriers. Other important considerations include the need for identification of key drivers of patient value, the relevance of different economic perspectives (i.e. healthcare vs. societal) and ethical questions in terms of treatment eligibility criteria.
Collapse
Affiliation(s)
- Karin Wahlberg
- The Swedish Institute for Health Economics, Lund, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | | | - William L Herring
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | | | | | - Pieter-Jelle Visser
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, Maastricht, The Netherlands
| | - Anders Wimo
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | | | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
21
|
Jönsson L, Ivkovic M, Atri A, Handels R, Gustavsson A, Hahn-Pedersen JH, León T, Lilja M, Gundgaard J, Raket LL. Progression analysis versus traditional methods to quantify slowing of disease progression in Alzheimer's disease. Alzheimers Res Ther 2024; 16:48. [PMID: 38424559 PMCID: PMC10903002 DOI: 10.1186/s13195-024-01413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The clinical meaningfulness of the effects of recently approved disease-modifying treatments (DMT) in Alzheimer's disease is under debate. Available evidence is limited to short-term effects on clinical rating scales which may be difficult to interpret and have limited intrinsic meaning to patients. The main value of DMTs accrues over the long term as they are expected to cause a delay or slowing of disease progression. While awaiting such evidence, the translation of short-term effects to time delays or slowing of progression could offer a powerful and readily interpretable representation of clinical outcomes. METHODS We simulated disease progression trajectories representing two arms, active and placebo, of a hypothetical clinical trial of a DMT. The placebo arm was simulated based on estimated mean trajectories of clinical dementia rating scale-sum of boxes (CDR-SB) recordings from amyloid-positive subjects with mild cognitive impairment (MCI) from Alzheimer's Disease Neuroimaging Initiative (ADNI). The active arm was simulated to show an average slowing of disease progression versus placebo of 20% at each visit. The treatment effects in the simulated trials were estimated with a progression model for repeated measures (PMRM) and a mixed model for repeated measures (MMRM) for comparison. For PMRM, the treatment effect is expressed in units of time (e.g., days) and for MMRM in units of the outcome (e.g., CDR-SB points). PMRM results were implemented in a health economics Markov model extrapolating disease progression and death over 15 years. RESULTS The PMRM model estimated a 19% delay in disease progression at 18 months and 20% (~ 7 months delay) at 36 months, while the MMRM model estimated a 25% reduction in CDR-SB (~ 0.5 points) at 36 months. The PMRM model had slightly greater power compared to MMRM. The health economic model based on the estimated time delay suggested an increase in life expectancy (10 months) without extending time in severe stages of disease. CONCLUSION PMRM methods can be used to estimate treatment effects in terms of slowing of progression which translates to time metrics that can be readily interpreted and appreciated as meaningful outcomes for patients, care partners, and health care practitioners.
Collapse
Affiliation(s)
- Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, 171 64, Sweden.
| | | | - Alireza Atri
- Banner Sun Health Research Institute and Banner Alzheimer's Institute, Banner Health, Sun City and Phoenix, AZ, USA
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ron Handels
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, 171 64, Sweden
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, Maastricht University Medical Centre+, Maastricht, MD, 6200, The Netherlands
| | - Anders Gustavsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, 171 64, Sweden
- Quantify Research, Hantverkargatan 8, Stockholm, 112 21, Sweden
| | | | | | - Mathias Lilja
- Quantify Research, Hantverkargatan 8, Stockholm, 112 21, Sweden
| | | | - Lars Lau Raket
- Novo Nordisk A/S, Søborg, Denmark
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Chudzik A, Śledzianowski A, Przybyszewski AW. Machine Learning and Digital Biomarkers Can Detect Early Stages of Neurodegenerative Diseases. SENSORS (BASEL, SWITZERLAND) 2024; 24:1572. [PMID: 38475108 PMCID: PMC10934426 DOI: 10.3390/s24051572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's Disease (AD) and Parkinson's Disease (PD) are devastating conditions that can develop without noticeable symptoms, causing irreversible damage to neurons before any signs become clinically evident. NDs are a major cause of disability and mortality worldwide. Currently, there are no cures or treatments to halt their progression. Therefore, the development of early detection methods is urgently needed to delay neuronal loss as soon as possible. Despite advancements in Medtech, the early diagnosis of NDs remains a challenge at the intersection of medical, IT, and regulatory fields. Thus, this review explores "digital biomarkers" (tools designed for remote neurocognitive data collection and AI analysis) as a potential solution. The review summarizes that recent studies combining AI with digital biomarkers suggest the possibility of identifying pre-symptomatic indicators of NDs. For instance, research utilizing convolutional neural networks for eye tracking has achieved significant diagnostic accuracies. ROC-AUC scores reached up to 0.88, indicating high model performance in differentiating between PD patients and healthy controls. Similarly, advancements in facial expression analysis through tools have demonstrated significant potential in detecting emotional changes in ND patients, with some models reaching an accuracy of 0.89 and a precision of 0.85. This review follows a structured approach to article selection, starting with a comprehensive database search and culminating in a rigorous quality assessment and meaning for NDs of the different methods. The process is visualized in 10 tables with 54 parameters describing different approaches and their consequences for understanding various mechanisms in ND changes. However, these methods also face challenges related to data accuracy and privacy concerns. To address these issues, this review proposes strategies that emphasize the need for rigorous validation and rapid integration into clinical practice. Such integration could transform ND diagnostics, making early detection tools more cost-effective and globally accessible. In conclusion, this review underscores the urgent need to incorporate validated digital health tools into mainstream medical practice. This integration could indicate a new era in the early diagnosis of neurodegenerative diseases, potentially altering the trajectory of these conditions for millions worldwide. Thus, by highlighting specific and statistically significant findings, this review demonstrates the current progress in this field and the potential impact of these advancements on the global management of NDs.
Collapse
Affiliation(s)
- Artur Chudzik
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Albert Śledzianowski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Andrzej W. Przybyszewski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
- UMass Chan Medical School, Department of Neurology, 65 Lake Avenue, Worcester, MA 01655, USA
| |
Collapse
|
23
|
Tarawneh R, Pankratz VS. The search for clarity regarding "clinically meaningful outcomes" in Alzheimer disease clinical trials: CLARITY-AD and Beyond. Alzheimers Res Ther 2024; 16:37. [PMID: 38365811 PMCID: PMC10870501 DOI: 10.1186/s13195-024-01412-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
CLARITY-AD is an 18-month, double-blinded, placebo-controlled, phase 3 trial which examined the safety and efficacy of the anti-amyloid agent, lecanemab, in mild cognitive impairment and mild dementia due to Alzheimer disease (AD). Lecanemab effectively reduced mean brain amyloid burden and was associated with statistically significant favorable effects, reflected by moderately less decline in the primary and secondary clinical outcomes, at 18 months compared to placebo. However, there is controversy within the AD community regarding the clinical significance of these results and whether they translate into clinically meaningful and tangible benefits on cognition or daily functions.We here review the primary and secondary clinical outcomes of CLARITY-AD and present our interpretation of the potential clinical meaningfulness of the group-level differences in study outcomes in the context of the 18-month study duration. We propose that the validation of stage-appropriate group-level thresholds for clinical meaningfulness of AD trial outcomes in biologically confirmed cohorts will allow objective interpretation of trial results and guide clinical decision-making. Further, in accordance with FDA guidance which emphasizes patient-focused drug development, the contextualization of AD clinical trial outcomes can be facilitated by supplementary individual-level data analyses which measure the risk of disease progression or summarize intraindividual change, using prespecified thresholds of clinically meaningful change, in each of the study groups over the trial period. The concepts of "time-saved" and "time-based" slowing in disease progression can be used to communicate clinical outcomes associated with emerging disease-modifying AD therapies to various stakeholders. We also describe several factors that need to be considered when evaluating outcomes of emerging AD therapies, including disease stage, the neuropathologic complexity of AD, time-based effects of disease-modifying therapies, and the possible influence of individual factors on treatment response and/or risk for adverse events. The consideration of these factors in the design and reporting of future trials of emerging AD therapies will guide clinicians regarding their appropriateness for use in various patient populations.Finally, we emphasize that data from clinical cohorts with longer durations of treatment and follow-up, including extension studies and patient registries, is needed to evaluate the long-term safety and efficacy of lecanemab in early symptomatic AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology and Center for Memory and Aging, University of New Mexico, Albuquerque, NM, USA.
| | - Vernon S Pankratz
- Department of Internal Medicine, Division of Epidemiology, Biostatistics, and Preventive Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
24
|
Scott IA. Monoclonal antibodies for treating early Alzheimer disease-a commentary on recent 'positive' trials. Age Ageing 2024; 53:afae023. [PMID: 38411409 DOI: 10.1093/ageing/afae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 12/30/2023] [Indexed: 02/28/2024] Open
Abstract
Recent phase 3 randomised controlled trials of amyloid-targeting monoclonal antibodies in people with pre-clinical or early Alzheimer disease have reported positive results, raising hope of finally having disease-modifying drugs. Given their far-reaching implications for clinical practice, the methods and findings of these trials, and the disease causation theory underpinning the mechanism of drug action, need to be critically appraised. Key considerations are the representativeness of trial populations; balance of prognostic factors at baseline; psychometric properties and minimal clinically important differences of the primary efficacy outcome measures; level of study fidelity; consistency of subgroup analyses; replication of findings in similar trials; sponsor role and potential conflicts of interest; consistency of results with disease causation theory; cost and resource estimates; and alternative prevention and treatment strategies. In this commentary, we show shortcomings in each of these areas and conclude that monoclonal antibody treatment for early Alzheimer disease is lacking high-quality evidence of clinically meaningful impacts at an affordable cost.
Collapse
Affiliation(s)
- Ian A Scott
- Centre for Health Services Research, University of Queensland, Brisbane, QLD, Australia
- Department of Internal Medicine and Clinical Epidemiology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
25
|
Cummings J, Osse AML, Cammann D, Powell J, Chen J. Anti-Amyloid Monoclonal Antibodies for the Treatment of Alzheimer's Disease. BioDrugs 2024; 38:5-22. [PMID: 37955845 PMCID: PMC10789674 DOI: 10.1007/s40259-023-00633-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/14/2023]
Abstract
Two monoclonal antibodies (mAbs), aducanumab and lecanemab, have received accelerated approval from the US FDA for initiation of treatment in early Alzheimer's disease patients who have proven β-amyloid pathology (Aβ). One of these, lecanemab, has subsequently received full approval and other monoclonal antibodies are poised for positive review and approval. Anti-amyloid mAbs share the feature of producing a marked reduction in total brain Aβ revealed by amyloid positron emission tomography. Trials associated with slowing of cognitive decline have achieved a reduction in measurable plaque Aβ in the range of 15-25 centiloids; trials of agents that did not reach this threshold were not associated with cognitive benefit. mAbs have differences in terms of titration schedules, MRI monitoring schedules for amyloid-related imaging abnormalities (ARIA), and continuing versus interrupted therapy. The approximate 30% slowing of decline observed with mAbs is clinically meaningful in terms of extended cognitive integrity and delay of onset of the more severe dementia phases of Alzheimer's disease. Approval of these agents initiates a new era in Alzheimer's disease therapeutics with disease-modifying properties. Further advances are needed, i.e. greater efficacy, improved safety, enhanced convenience, and better understanding of ill-understood observations such as brain volume loss.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Department of Brain Health, Chambers-Grundy Center for Transformative Neuroscience, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
- , 1380 Opal Valley Street, Henderson, NV, 89052, USA.
| | - Amanda M Leisgang Osse
- Department of Brain Health, Chambers-Grundy Center for Transformative Neuroscience, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jayde Powell
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| |
Collapse
|
26
|
Morin P, Aguilar BJ, Li X, Chen J, Berlowitz D, Zhang R, Tahami Monfared AA, Zhang Q, Xia W. Alzheimer's Disease Stage Transitions Among United States Veterans. J Alzheimers Dis 2024; 97:687-695. [PMID: 38143359 DOI: 10.3233/jad-230850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and related dementias are progressive neurological disorders with stage-specific clinical features and challenges. An important knowledge gap is the "window of time" within which patients transition from mild cognitive impairment or mild AD to moderate or severe AD. Better characterization/establishment of transition times would help clinicians initiating treatments, including anti-amyloid therapy. OBJECTIVE To describe cognitive test score-based AD stage transitions in Veterans with AD in the US Veterans Affairs Healthcare System (VAHS). METHODS This retrospective analysis (2010-2019) identified Veterans with AD from the VAHS Electronic Health Record (EHR) notes. AD stage was based on Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), or Saint Louis University Mental Status (SLUMS) Examination scores in the EHR. RESULTS We identified 296,519 Veterans with cognitive test-based AD staging. Over the 10-year study, the proportion of veterans with MMSE scores declined from 24.9% to 9.5% while those with SLUMS rose from 9.0% to 17.8%; and MoCA rose from 5.0% to 25.4%. The average forward transition times between each stage were approximately 2-4 years, whether assessed by MMSE, MoCA, or SLUMS. CONCLUSION The average transition time for cognitive test-based assessments of initial cognitive decline, early-stage AD, and moderate/severe AD in the VAHS is 2-4 years. In view of the short window for introducing disease-modifying therapy and the significant benefits of early treatment of AD, our data suggest a critical need for treatment guidelines in the management of AD.
Collapse
Affiliation(s)
- Peter Morin
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Byron J Aguilar
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
| | - Xuyang Li
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
| | - Jinying Chen
- Department of Preventive Medicine and Epidemiology, Data Science Core, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Dan Berlowitz
- Department of Public Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Raymond Zhang
- Alzheimer's Disease and Brain Health, EisaiInc., Nutley, NJ, USA
| | - Amir Abbas Tahami Monfared
- Alzheimer's Disease and Brain Health, EisaiInc., Nutley, NJ, USA
- Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Quanwu Zhang
- Alzheimer's Disease and Brain Health, EisaiInc., Nutley, NJ, USA
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biological Sciences, Kennedy College of Science, University of Massachusetts Lowell, Lowell, MA, USA
| |
Collapse
|
27
|
Pajewski NM, Donohue MC, Raman R, Espeland MA. Ascertainment and Statistical Issues for Randomized Trials of Cardiovascular Interventions for Cognitive Impairment and Dementia. Hypertension 2024; 81:45-53. [PMID: 37732473 PMCID: PMC10840823 DOI: 10.1161/hypertensionaha.123.19941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
There has been considerable progress in the prevention and treatment of cardiovascular disease, reducing the population burden of cardiovascular morbidity and mortality. Recently, some randomized trials, including the SPRINT (Systolic Blood Pressure Intervention Trial), have suggested that improvements in cardiovascular risk factors may also slow cognitive decline and reduce the eventual development of dementia. Unfortunately, the randomized trial template that has been used repeatedly to successfully demonstrate reductions in major adverse cardiac events faces several design and analytic obstacles when applied in the context of cognitive decline and dementia. Here, we review these obstacles, motivated by SPRINT and the context of selecting an appropriate cognitive end point for future preventive randomized trials. A few options are available, spanning neuropsychological test scores or composites reflecting specific domains of cognitive function, adjudicated cognitive impairment, or potentially physiological biomarkers. This choice entails considerations around statistical power, modes of ascertainment, the clinical relevance of treatment effects, a myriad of statistical issues (interval censoring, missing data, the competing risk of death, practice effects, etc), as well as ethical considerations around equipoise. Collectively, these considerations indicate that trials aiming to mitigate the cardiovascular contribution to cognitive decline and dementia will generally need to be large, inclusive of a wide age range of older adults, and with multiple years of follow-up.
Collapse
Affiliation(s)
- Nicholas M. Pajewski
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Michael C. Donohue
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA
| | - Rema Raman
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA
| | - Mark A. Espeland
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC
- Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
28
|
Dong Q, Li Z, Liu W, Chen K, Su Y, Wu J, Caselli RJ, Reiman EM, Wang Y, Shen J. Correlation studies of Hippocampal Morphometry and Plasma NFL Levels in Cognitively Unimpaired Subjects. IEEE TRANSACTIONS ON COMPUTATIONAL SOCIAL SYSTEMS 2023; 10:3602-3608. [PMID: 38084365 PMCID: PMC10713345 DOI: 10.1109/tcss.2023.3313819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Alzheimer's disease(AD) is being the burden of society and family. Applying computing-aided strategies to reveal its pathology is one of the research highlights. Plasma neurofilament light (NFL) is an emerging noninvasive and economic biomarker for AD molecular pathology. It is valuable to reveal the correlations between the plasma NFL levels and neurodegeneration, especially hippcampal deformations at the preclinical stage. The negative correlation between plasma NFL levels and hippocampal volumes has been documented. However, the relationship between the plasma NFL levels and the hippocampal morphometry details at the preclinical stage is still elusive. This study seeks to demonstrate the capacity of our proposed surface-based hippocampal morphometry system to discern the plasma NFL positive (NFL+>41.9 pg/L) level and plasma NFL negative (NFL-<41.9pg/L) level and illustrate its superiority to the hippocampal volume measurement by drawing the cohort of 154 CU middle aged and elderly adults. We also apply this morphometry measure and a proposed sparse coding based classification algorithm to classify CU individuals with NFL+ and NFL- levels. Experimental results show that the proposed hippocampal morphometry system offers stronger statistical power to discriminate CU subjects with NFL+ and NFL- levels, comparing with the hippocampal volume measure. Furthermore, this system can discriminate plasma NFL levels in CU individuals (Accuracy=0.86). Both the group level and individual level analysis results indicate that the association between plasma NFL levels and the hippocampal shapes can be mapped at the preclinical stage.
Collapse
Affiliation(s)
- Qunxi Dong
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhigang Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Weijia Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Jianfeng Wu
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State Univ., Tempe, AZ, USA
| | | | | | - Yalin Wang
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State Univ., Tempe, AZ, USA
| | - Jian Shen
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
29
|
Liu KY, Walsh S, Brayne C, Merrick R, Richard E, Howard R. Evaluation of clinical benefits of treatments for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2023; 4:e645-e651. [PMID: 37924845 DOI: 10.1016/s2666-7568(23)00193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 11/06/2023] Open
Abstract
The need for regulatory approval of new therapies for the treatment of Alzheimer's disease-a progressive neurodegenerative condition-has made the assessment of treatment efficacy an urgent priority for discussion and investigation in the field. In the first part of this Personal View, we summarise current views on what constitutes a clinically meaningful benefit from treatment for Alzheimer's disease, including the concept of a minimum treatment effect against which to compare trial outcomes and its limitations. Considering existing and divergent definitions of clinically meaningful change, we define this concept in the second part of the Personal View by proposing a new approach that consecutively considers whether a treatment benefit for Alzheimer's disease is noticeable, valuable, and worthwhile in the context of costs and risks. This approach could be a useful foundation from which the field can move forwards on this issue and address existing gaps in understanding.
Collapse
Affiliation(s)
- Kathy Y Liu
- Division of Psychiatry, University College London, London, UK.
| | - Sebastian Walsh
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Richard Merrick
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands; Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
30
|
Cummings J. Meaningful benefit and minimal clinically important difference (MCID) in Alzheimer's disease: Open peer commentary. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12411. [PMID: 37521521 PMCID: PMC10372384 DOI: 10.1002/trc2.12411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023]
Abstract
Introduction Approval of the anti-amyloid monoclonal antibodies has stimulated an important discussion of the value to be placed on the magnitude of slowing achieved by treatment compared to placebo. Methods The minimal clinically important difference (MCID) was reviewed in the context of other measures and analyses that provide perspective on the meaningfulness of treatment responses. Results TheMCID is a clinician-anchored approach to making this determination. The MCID applies best to symptomatic therapies for which the drug-placebo difference remains constant. Disease-modifying therapies produce a progressive divergence of drug and placebo trajectories; early in the course the MCID would not be achieved, later the MCID will be achieved, and with continuing therapy the MCID will be exceeded. Clinicians are not the only stakeholders involved in determining the value proposition of slowing disease progression. Patient-reported outcomes and caregiver-related measures offer important complementary insights. Analytic approaches also widen the perspective on the observed drug-placebo differences. Risk ratios, numbers needed to treat versus number needed to harm, time-to-event analyses, and predictive benefits based on biomarkers all add depth to the discussion. Discussion Multiple stakeholder perspectives are needed to determine the importance to be attributed to the therapeutic changes observed with monoclonal antibody therapies and other emerging treatments.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
31
|
Salemme S, Ancidoni A, Locuratolo N, Piscopo P, Lacorte E, Canevelli M, Vanacore N. Advances in amyloid-targeting monoclonal antibodies for Alzheimer's disease: clinical and public health issues. Expert Rev Neurother 2023; 23:1113-1129. [PMID: 37975226 DOI: 10.1080/14737175.2023.2284305] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a major global public health challenge. To date, no treatments have been shown to stop the underlying pathological processes. The cerebral accumulation of amyloid-beta (Ab) is still considered as the primum movens of AD and disease-modifying treatments targeting Ab are reaching - or have already reached - clinical practice. AREAS COVERED The authors explore the main advancements from Aβ-targeting monoclonal antibodies (mAbs) for the treatment of AD. From a public health perspective, they address ethically relevant issues such as the benevolence and non-maleficence principles. They report on the potential biological and clinical benefits of these drugs, discussing minimal clinically important differences (MCID) and other relevant outcomes. They examine the short- and long-term effects of amyloid-related imaging abnormalities (ARIA), and explore the differences between eligibility criteria in clinical trials, appropriate use recommendations, and prescribing information content. In doing so, they contextualize the discussion on the disagreements among different regulatory authorities. EXPERT OPINION Although anti-β-amyloid monoclonal antibodies may be effective in selected scenarios, non-negligible knowledge gaps and implementation limits persist. Overcoming these gaps can no longer be postponed if we are to ensure the principles of Quality of Care for patients with cognitive impairment who would be eligible for this class of drugs.
Collapse
Affiliation(s)
- Simone Salemme
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Ancidoni
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Nicoletta Locuratolo
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Eleonora Lacorte
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Marco Canevelli
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Human Neuroscience, "Sapienza" University, Rome, Italy
| | - Nicola Vanacore
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|
32
|
Rentz DM, Klinger HM, Samaroo A, Fitzpatrick C, Schneider OR, Amagai S, Peipert JD. Face Name Associative Memory Exam and biomarker status in the ARMADA study: Advancing reliable measurement in Alzheimer's disease and cognitive aging. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12473. [PMID: 37693224 PMCID: PMC10483494 DOI: 10.1002/dad2.12473] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/30/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023]
Abstract
The Face Name Associative Memory Exam (FNAME) was introduced into the NIH Toolbox as part of the ARMADA study and establishes normative data for diverse participants, ages 64 to 85+, and proposes cutoff scores between biomarker positive versus negative (+/-) groups. The FNAME was administered to 257 participants across the clinical spectrum with 122 having amyloid biomarkers. Linear regression explored the association between demographics and FNAME and between amyloid (+/-) groups. Receiver operating characteristic curves (ROC) identified performance thresholds that best discriminated between biomarker (+/-) individuals. Lower FNAME scores occurred in males, older ages, Black/African Americans, Hispanics, and biomarker-positive participants. ROC analyses demonstrated acceptable accuracy (0.73 to 0.77) but only when combined with clinical status. The diagnostic discrimination of amyloid positivity was acceptable but not excellent, suggesting the FNAME may be a better screening indicator of clinical status rather than amyloid deposition in cognitively normal individuals. Normative data are provided.
Collapse
Affiliation(s)
- Dorene M. Rentz
- Departments of NeurologyMassachusetts General HospitalBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Hannah M. Klinger
- Departments of NeurologyMassachusetts General HospitalBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Colleen Fitzpatrick
- Departments of NeurologyMassachusetts General HospitalBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Saki Amagai
- Northwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | | |
Collapse
|
33
|
Liu KY, Villain N, Ayton S, Ackley SF, Planche V, Howard R, Thambisetty M. Key questions for the evaluation of anti-amyloid immunotherapies for Alzheimer's disease. Brain Commun 2023; 5:fcad175. [PMID: 37389302 PMCID: PMC10306158 DOI: 10.1093/braincomms/fcad175] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
The clinical benefit associated with anti-amyloid immunotherapies, a new class of drugs for the treatment of Alzheimer's disease, is predicated on their ability to modify disease course by lowering brain amyloid levels. At the time of writing, two amyloid-lowering antibodies, aducanumab and lecanemab, have obtained United States Food and Drug Administration accelerated approval, with further agents of this class in the Alzheimer's disease treatment pipeline. Based on limited published clinical trial data to date, regulators, payors and physicians will need to assess their efficacy, clinical effectiveness and safety, as well as cost and accessibility. We propose that attention to three important questions related to treatment efficacy, clinical effectiveness and safety should guide evidence-based consideration of this important class of drugs. These are: (1) Were trial statistical analyses appropriate and did they convincingly support claims of efficacy? (2) Do reported treatment effects outweigh safety concerns and are they generalizable to a representative clinical population of people with Alzheimer's disease? and (3) Do the data convincingly demonstrate disease course modification, suggesting that increasing clinical benefits beyond the duration of the trials are likely? We suggest specific approaches to interpreting trial results for these drugs and highlight important areas of uncertainty where additional data and a cautious interpretation of existing results is warranted. Safe, effective and accessible treatments for Alzheimer's disease are eagerly awaited by millions of patients and their caregivers worldwide. While amyloid-targeting immunotherapies may be promising disease-modifying Alzheimer's disease treatments, rigorous and unbiased assessment of clinical trial data is critical to regulatory decision-making and subsequently determining their provision and utility in routine clinical practice. Our recommendations provide a framework for evidence-based appraisal of these drugs by regulators, payors, physicians and patients.
Collapse
Affiliation(s)
- Kathy Y Liu
- Division of Psychiatry, University College London, London W1T 7NF, UK
| | - Nicolas Villain
- AP-HP.Sorbonne Université, Institut de la Mémoire et de la Maladie d’Alzheimer, Département de Neurologie, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Sorbonne Université, Institut national de la Santé et de la Recherche Medical (INSERM) U1127, Centre National de la Recherche Scientifique (CNRS) 7225, Institut du Cerveau—ICM, 75013 Paris, France
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Sarah F Ackley
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Vincent Planche
- Univ. Bordeaux, CNRS, UMR 5293, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France
- Centre Mémoire Ressources Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Robert Howard
- Division of Psychiatry, University College London, London W1T 7NF, UK
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, USA
| |
Collapse
|
34
|
Cummings J. Anti-Amyloid Monoclonal Antibodies are Transformative Treatments that Redefine Alzheimer's Disease Therapeutics. Drugs 2023; 83:569-576. [PMID: 37060386 PMCID: PMC10195708 DOI: 10.1007/s40265-023-01858-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2023] [Indexed: 04/16/2023]
Abstract
Two anti-amyloid monoclonal antibodies (MABs)-lecanemab (Leqembi®) and aducanumab (Aduhelm®)-have been approved in the USA for the treatment of Alzheimer's disease (AD). Anti-amyloid monoclonal antibodies are the first disease-modifying therapies for AD that achieve slowing of clinical decline by intervening in the basic biological processes of the disease. These are breakthrough agents that can slow the inevitable progression of AD into more severe cognitive impairment. The results of trials of anti-amyloid MABs support the amyloid hypothesis and amyloid as a target for AD drug development. The success of MABs reflects a relentless application of neuroscience knowledge to solving major challenges facing humankind. The success of these transformative agents will foster the development of more anti-amyloid MABs, other types of anti-amyloid therapies, treatments of other targets of AD biology, and new approaches to therapies for an array of neurodegenerative disorders. Monoclonal antibodies have side effects and, during the period of treatment initiation, patients must be closely monitored for the occurrence of amyloid-related imaging abnormalities (ARIA) and infusion reactions. A successful first step in the development of disease-modifying therapy for AD defines desirable features for the next phase of therapeutic development including less frequent ARIA, more convenient administration, and greater efficacy. Unprecedented agents make new demands on patients and care partners, clinicians, payers, and health care systems. Collaboration among stakeholders is essential to take advantage of the therapeutic benefits offered by these agents and to make them widely available. Monoclonal antibodies usher in a new era in AD therapy and define a new landscape of what is possible for therapeutic development for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Joy Chambers-Grundy Professor of Brain Science, Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| |
Collapse
|
35
|
Saposnik G, Sánchez-Benavidez G, García-Arcelay E, Franco-Macías E, Bensi C, Carmelingo S, Allegri RF, Pérez-Martínez DA, Maurino J. Design of a Non-Interventional Study to Assess Neurologists' Perspectives and Pharmacological Treatment Decisions in Early Alzheimer's Disease. Neurol Ther 2023; 12:995-1006. [PMID: 36952172 DOI: 10.1007/s40120-023-00466-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023] Open
Abstract
INTRODUCTION The current therapeutic landscape of Alzheimer's disease (AD) is evolving rapidly. Our treatment options include new anti-amyloid-β protein disease-modifying therapies (DMTs) that decrease cognitive decline in patients with early AD (prodromal and mild AD dementia). Despite these advances, we have limited information on how neurologists would apply the results of recent DMT trials to make treatment decisions. Our goal is to identify factors associated with the use of new AD DMTs among neurologists applying concepts from behavioral economics. METHODS This non-interventional, cross-sectional, web-based study will assess 400 neurologists with expertise in AD from across Spain. Participants will start by completing demographic information, practice settings, and a behavioral battery to address their tolerance to uncertainty and risk preferences. Participants will then be presented with 10 simulated case scenarios or vignettes of common encounters in patients with early AD to evaluate treatment initiation with anti-amyloid-β DMTs (e.g., aducanumab, lecanemab, etc.). The primary outcomes will be therapeutic inertia and suboptimal decisions. Discrete choice experiments will be used to determine the weight of factors influencing treatment choices. RESULTS The results of this study will provide new insights into a better understanding of the most relevant factors associated with therapeutic decisions on the use of DMTs, assessing how neurologists handle uncertainty when making treatment choices, and identifying the prevalence of therapeutic inertia in the management of early AD.
Collapse
Affiliation(s)
- Gustavo Saposnik
- Clinical Outcomes and Decision Neuroscience Unit, Li Ka Shing Institute, University of Toronto, Toronto, Canada.
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, 55 Queen St E, Toronto, ON, M5C 1R6, Canada.
| | - Gonzalo Sánchez-Benavidez
- BarcelonaBeta Brain Research Center, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | | | - Emilio Franco-Macías
- Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Seville, Spain
| | - Catalina Bensi
- Medical Department, Roche Farma, Buenos Aires, Argentina
| | | | - Ricardo F Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, Fleni, Buenos Aires, Argentina
| | - David A Pérez-Martínez
- Department of Neurology, Hospital Universitario Doce de Octubre, Hospital Universitario La Luz, Madrid, Spain
| | | |
Collapse
|
36
|
Technical Review of Clinical Outcomes Assessments Across the Continuum of Alzheimer's Disease. Neurol Ther 2023; 12:571-595. [PMID: 36790638 PMCID: PMC10043075 DOI: 10.1007/s40120-023-00443-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
INTRODUCTION Insight into the relationship between concepts that matter to the people affected by Alzheimer's disease (AD) and the clinical outcome assessments (COAs) commonly used in AD clinical studies is limited. Phases 1 and 2 of the What Matters Most (WMM) study series identified and quantitatively confirmed 42 treatment-related outcomes that are important to people affected by AD. METHODS We compared WMM concepts rated as "very important" or higher to items included in COAs used commonly in AD studies. RESULTS Twenty COAs designed to assess signs, symptoms, and impacts across the spectrum of AD were selected for review. Among these 20 COAs, only 5 reflected 12 or more WMM concepts [Integrated Alzheimer's Disease Rating Scale (iADRS), Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory (ADCS-ADL), Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory-Mild Cognitive Impairment (ADCS-ADL-MCI), Alzheimer's Disease Composite Scores (ADCOMS), and Clinical Dementia Rating; Clinical Dementia Rating-Sum of Boxes (CDR/CDR-SB)]. Multiple symptoms and impacts of AD identified as important and meaningful in the WMM studies map only indirectly at best to 7 of the 20 most widely used COAs. CONCLUSION While many frequently used COAs in AD capture some concepts identified as important to AD populations and their care partners, overlap between any single measure and the concepts that matter to people affected by AD is limited. The highest singly matched COA reflects fewer than half (45%) of WMM concepts. Use of multiple COAs expands coverage of meaningful concepts. Future research should explore the content validity of AD COAs planned for AD trials based on further confirmation of the ecological validity of the WMM items. This research should inform development and use of core outcome sets that capture WMM items and selection or development of new companion tools to fully demonstrate clinically meaningful outcomes spanning WMM.
Collapse
|
37
|
Increased Hippocampal-Inferior Temporal Gyrus White Matter Connectivity following Donepezil Treatment in Patients with Early Alzheimer's Disease: A Diffusion Tensor Probabilistic Tractography Study. J Clin Med 2023; 12:jcm12030967. [PMID: 36769615 PMCID: PMC9917574 DOI: 10.3390/jcm12030967] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
The incidence of Alzheimer's disease (AD) has been increasing each year, and a defective hippocampus has been primarily associated with an early stage of AD. However, the effect of donepezil treatment on hippocampus-related networks is unknown. Thus, in the current study, we evaluated the hippocampal white matter (WM) connectivity in patients with early-stage AD before and after donepezil treatment using probabilistic tractography, and we further determined the WM integrity and changes in brain volume. Ten patients with early-stage AD (mean age = 72.4 ± 7.9 years; seven females and three males) and nine healthy controls (HC; mean age = 70.7 ± 3.5 years; six females and three males) underwent a magnetic resonance (MR) examination. After performing the first MR examination, the patients received donepezil treatment for 6 months. The brain volumes and diffusion tensor imaging scalars of 11 regions of interest (the superior/middle/inferior frontal gyrus, the superior/middle/inferior temporal gyrus, the amygdala, the caudate nucleus, the hippocampus, the putamen, and the thalamus) were measured using MR imaging and DTI, respectively. Seed-based structural connectivity analyses were focused on the hippocampus. The patients with early AD had a lower hippocampal volume and WM connectivity with the superior frontal gyrus and higher mean diffusivity (MD) and radial diffusivity (RD) in the amygdala than HC (p < 0.05, Bonferroni-corrected). However, brain areas with a higher (or lower) brain volume and WM connectivity were not observed in the HC compared with the patients with early AD. After six months of donepezil treatment, the patients with early AD showed increased hippocampal-inferior temporal gyrus (ITG) WM connectivity (p < 0.05, Bonferroni-corrected).
Collapse
|
38
|
Cummings J, Apostolova L, Rabinovici GD, Atri A, Aisen P, Greenberg S, Hendrix S, Selkoe D, Weiner M, Petersen RC, Salloway S. Lecanemab: Appropriate Use Recommendations. J Prev Alzheimers Dis 2023; 10:362-377. [PMID: 37357276 PMCID: PMC10313141 DOI: 10.14283/jpad.2023.30] [Citation(s) in RCA: 220] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Lecanemab (Leqembi®) is approved in the United States for the treatment of Alzheimer's disease (AD) to be initiated in early AD (mild cognitive impairment [MCI] due to AD or mild AD dementia) with confirmed brain amyloid pathology. Appropriate Use Recommendations (AURs) are intended to help guide the introduction of new therapies into real-world clinical practice. Community dwelling patients with AD differ from those participating in clinical trials. Administration of lecanemab at clinical trial sites by individuals experienced with monoclonal antibody therapy also differs from the community clinic-based administration of lecanemab. These AURs use clinical trial data as well as research and care information regarding AD to help clinicians administer lecanemab with optimal safety and opportunity for effectiveness. Safety and efficacy of lecanemab are known only for patients like those participating in the phase 2 and phase 3 lecanemab trials, and these AURs adhere closely to the inclusion and exclusion criteria of the trials. Adverse events may occur with lecanemab including amyloid related imaging abnormalities (ARIA) and infusion reactions. Monitoring guidelines for these events are detailed in this AUR. Most ARIA with lecanemab is asymptomatic, but a few cases are serious or, very rarely, fatal. Microhemorrhages and rare macrohemorrhages may occur in patients receiving lecanemab. Anticoagulation increases the risk of hemorrhage, and the AUR recommends that patients requiring anticoagulants not receive lecanemab until more data regarding this interaction are available. Patients who are apolipoprotein E ε4 (APOE4) gene carriers, especially APOE4 homozygotes, are at higher risk for ARIA, and the AUR recommends APOE genotyping to better inform risk discussions with patients who are lecanemab candidates. Clinician and institutional preparedness are mandatory for use of lecanemab, and protocols for management of serious events should be developed and implemented. Communication between clinicians and therapy candidates or those on therapy is a key element of good clinical practice for the use of lecanemab. Patients and their care partners must understand the potential benefits, the potential harms, and the monitoring requirements for treatment with this agent. Culture-specific communication and building of trust between clinicians and patients are the foundation for successful use of lecanemab.
Collapse
Affiliation(s)
- J Cummings
- Jeffrey Cummings, MD, ScD, 1380 Opal Valley Street, Henderson, NV 89052, USA, , T: 702-902-3939
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ishii K, Hanaoka K, Watanabe S, Morimoto-Ishikawa D, Yamada T, Kaida H, Yamakawa Y, Minagawa S, Takenouchi S, Ohtani A, Mizuta T. High-Resolution Silicon Photomultiplier Time-of-Flight Dedicated Head PET System for Clinical Brain Studies. J Nucl Med 2023; 64:153-158. [PMID: 35798557 PMCID: PMC9841263 DOI: 10.2967/jnumed.122.264080] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/28/2023] Open
Abstract
We acquired brain 18F-FDG and 18F-flutemetamol PET images using a time-of-flight system dedicated to the head (dhPET) and a conventional whole-body PET/CT (wbPET) system and evaluated the clinical superiority of dhPET over wbPET. Methods: There were 18 subjects for the 18F-FDG PET study and 17 subjects for the 18F-flutemetamol PET study. 18F-FDG PET images were first obtained using wbPET, followed by dhPET. 18F-flutemetamol PET images were first obtained using wbPET, followed by dhPET. Images acquired using dhPET and wbPET were compared by visual inspection, voxelwise analysis, and SUV ratio (SUVR). Results: All 18F-FDG and 18F-flutemetamol images acquired using dhPET were judged as visually better than those acquired using wbPET. The voxelwise analysis demonstrated that accumulations in the cerebellum, in the lateral occipital cortices, and around the central sulcus area in dhPET 18F-FDG images were lower than those in wbPET 18F-FDG images, whereas accumulations around the ventricle systems were higher in dhPET 18F-FDG images than those in wbPET 18F-FDG images. Accumulations in the cerebellar dentate nucleus, in the midbrain, in the lateral occipital cortices, and around the central sulcus area in dhPET images were lower than those in wbPET images, whereas accumulations around the ventricle systems were higher in dhPET 18F-flutemetamol images than those in wbPET 18F-flutemetamol images. The mean cortical SUVRs of 18F-FDG and 18F-flutemetamol dhPET images were significantly higher than those of 18F-FDG and 18F-flutemetamol wbPET images, respectively. Conclusion: The dhPET images had better image quality by visual inspection and higher SUVRs than wbPET images. Although there were several regional accumulation differences between dhPET and wbPET images, understanding this phenomenon will enable full use of the features of this dhPET system in clinical practice.
Collapse
Affiliation(s)
- Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Japan; .,Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | - Kohei Hanaoka
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | - Shota Watanabe
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | - Daisuke Morimoto-Ishikawa
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | - Takahiro Yamada
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | - Hayato Kaida
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Japan;,Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, Osakasayama, Japan; and
| | | | - Suzuka Minagawa
- Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | | | - Atsushi Ohtani
- Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Tetsuro Mizuta
- Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| |
Collapse
|
40
|
Gregory S, Saunders S, Ritchie CW. Science disconnected: the translational gap between basic science, clinical trials, and patient care in Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2022; 3:e797-e803. [PMID: 36356629 DOI: 10.1016/s2666-7568(22)00219-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/22/2022] [Accepted: 09/08/2022] [Indexed: 11/09/2022] Open
Abstract
Both research and clinical practice have traditionally centred on the dementia syndrome of Alzheimer's disease rather than its preclinical and prodromal stages. However, there is a strong scientific and ethical impetus to shift focus to earlier disease stages to improve brain health outcomes and help to keep affected individuals symptom-free (dementia-free) for as long as possible. We provide an overview of recent advancements in early detection, drug development, and trial methodology that should be utilised in the development of new therapies for use in brain health clinics. We propose a triad approach to Alzheimer's disease clinical trials, encompassing (1) experimental medicine studies to gather greater knowledge of disease mechanisms, (2) a more comprehensive platform of phase 2 learning trials to inform phase 3 confirmatory trials, and (3) precision medicine involving smaller subgroups of patients with shared characteristics. This triad would ensure that treatment targets are identified accurately, trial methodology focuses on at-risk populations, and sensitive outcome measures capture potential treatment effects. Clinical services around the world must embrace the brain health clinic model so that neurodegenerative diseases can be detected in their earliest phase to quicken drug development pipelines and potentially improve prognosis.
Collapse
Affiliation(s)
- Sarah Gregory
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, Outpatient Department 2, Western General Hospital, University of Edinburgh, Edinburgh, UK.
| | - Stina Saunders
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, Outpatient Department 2, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, Outpatient Department 2, Western General Hospital, University of Edinburgh, Edinburgh, UK; Brain Health Scotland, Edinburgh, UK
| |
Collapse
|
41
|
Lipton RB, Podger L, Stewart WF, Gomez-Ulloa D, Rodriguez WI, Runken MC, Barnes FB, Serrano D. Toward the optimized assessment of clinical outcomes in studies of novel treatments for Alzheimer's disease. Expert Rev Neurother 2022; 22:863-873. [PMID: 36440481 DOI: 10.1080/14737175.2022.2149324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by a progressive decline in cognition and daily function, leading to a greater need for caregiver support. Clinical disease is segmented into a preclinical stage, mild cognitive impairment, and mild, moderate, and severe stages of Alzheimer's dementia. Although AD trials enroll participants at various stages of illness, treatment efficacy is often assessed using endpoints based on measures of outcomes that are held fixed across disease stages. We hypothesize that matching the primary outcomes measured in the endpoint hierarchy to the stage of disease targeted by the trial will increase the likelihood of detecting true treatment benefits. AREAS COVERED We discuss current approaches to assessing clinical outcomes in AD trials, followed by a consideration of how effect detection can be improved by linking the stage of AD to the endpoints that most likely reflect stage-specific disease progression. EXPERT OPINION Failing to account for stage-specific relevance and sensitivity of clinical outcomes may be one factor that contributes to trial failures in AD. Given the history of failure, experts have begun to scrutinize the relevance and sensitivity of outcomes as a potentially modifiable barrier to successful trials. To this end, we present a framework for refining trial endpoint selection and evaluation.
Collapse
Affiliation(s)
- Richard B Lipton
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, New York, NY, USA
| | | | | | | | | | - M Chris Runken
- Global HEOR, Grifols SSNA - Research Triangle Park, NC, USA
| | | | | |
Collapse
|