1
|
Sha S, Ren L, Xing X, Guo W, Wang Y, Li Y, Cao Y, Qu L. Recent advances in immunotherapy targeting amyloid-beta and tauopathies in Alzheimer's disease. Neural Regen Res 2026; 21:577-587. [PMID: 39885674 DOI: 10.4103/nrr.nrr-d-24-00846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/28/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease, a devastating neurodegenerative disorder, is characterized by progressive cognitive decline, primarily due to amyloid-beta protein deposition and tau protein phosphorylation. Effectively reducing the cytotoxicity of amyloid-beta42 aggregates and tau oligomers may help slow the progression of Alzheimer's disease. Conventional drugs, such as donepezil, can only alleviate symptoms and are not able to prevent the underlying pathological processes or cognitive decline. Currently, active and passive immunotherapies targeting amyloid-beta and tau have shown some efficacy in mice with asymptomatic Alzheimer's disease and other transgenic animal models, attracting considerable attention. However, the clinical application of these immunotherapies demonstrated only limited efficacy before the discovery of lecanemab and donanemab. This review first discusses the advancements in the pathogenesis of Alzheimer's disease and active and passive immunotherapies targeting amyloid-beta and tau proteins. Furthermore, it reviews the advantages and disadvantages of various immunotherapies and considers their future prospects. Although some antibodies have shown promise in patients with mild Alzheimer's disease, substantial clinical data are still lacking to validate their effectiveness in individuals with moderate Alzheimer's disease.
Collapse
Affiliation(s)
- Sha Sha
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lina Ren
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Wanshu Guo
- Department of Neurology, People's Hospital of Liaoning Province, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ying Li
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Le Qu
- Department of Dermatology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
2
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [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: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
3
|
Goff J, Khalifa M, Short SM, van der Graaf PH, Geerts H. Interactions of Therapeutic Antibodies With Presynaptically-Released Misfolded Proteins in Neurodegenerative Diseases. A Spatial Monte-Carlo Simulation Study. CPT Pharmacometrics Syst Pharmacol 2025. [PMID: 40296445 DOI: 10.1002/psp4.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/28/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
The spatial progression hypothesis of misfolded tau and alpha-synuclein proteins in Alzheimer's and Parkinson's Disease proposes the release of proteins from a presynaptic membrane followed by diffusion over the synaptic cleft and uptake by the postsynaptic membrane in the afferent neuron. A number of antibodies aiming to reduce this neuronal uptake by capturing these proteins in the extracellular space are currently in clinical development, so far without much success. For modeling the interaction between antibodies and misfolded proteins in the extremely small synaptic volume with only a few proteins navigating a crowded environment of transsynaptic proteins, traditional assumptions of ordinary differential equations (ODEs) break down. Here we use spatial Monte Carlo calculations of individual molecule trajectories in a realistic geometrical environment using the open-source software Mcell (mcell.org). For several different densities of transsynaptic proteins, we show that due to geometric constraints, less than 0.5% of the antibody in the brain interstitial fluid (ISF) can enter the crowded synaptic cleft. As a consequence, uptake of the seed-competent proteins is reduced by less than 10%, even at the highest concentration and for selective antibodies. Only the seed-competent protein that escapes the synaptic cleft (between 15% and 30%) is captured by the antibody. Given the extremely low penetrance of the antibodies, it is close to impossible for antibodies to interfere with the uptake mechanism that takes place in the synaptic cleft. These simulations using a detailed and realistic biological environment provide a possible explanation for the clinical trial failures of anti-tau and anti-αsynuclein antibodies.
Collapse
Affiliation(s)
- Jamie Goff
- Certara Predictive Technologies, Sheffield, UK
| | | | - Shaina M Short
- Certara Predictive Technologies, Radnor, Pennsylvania, USA
| | | | - Hugo Geerts
- Certara Predictive Technologies, Radnor, Pennsylvania, USA
| |
Collapse
|
4
|
Xing H, Yue S, Qin R, Du X, Wu Y, Zhangsun D, Luo S. Recent Advances in Drug Development for Alzheimer's Disease: A Comprehensive Review. Int J Mol Sci 2025; 26:3905. [PMID: 40332804 PMCID: PMC12028297 DOI: 10.3390/ijms26083905] [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: 03/12/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by cognitive impairments such as memory loss and executive dysfunction. The primary pathological features of AD include the deposition of amyloid-beta (Aβ) plaques, the hyperphosphorylation of tau proteins leading to neurofibrillary tangles, disruptions of neuronal and synaptic functions, and chronic inflammatory responses. These multifactorial interactions drive disease progression. To date, various therapeutic agents targeting these pathological mechanisms have been developed. This article provides a comprehensive review of the pathogenesis of AD, recent advances in drug development targeting different pathways, current challenges, and future directions, aiming to offer valuable insights for clinical treatment and research.
Collapse
Affiliation(s)
- Haonan Xing
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
| | - Song Yue
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
| | - Runtian Qin
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
| | - Xiaoxue Du
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
| | - Yili Wu
- Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Health, School of Mental Health, Institute of Aging, Wenzhou Medical University, Wenzhou 325000, China;
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (H.X.); (S.Y.); (R.Q.); (X.D.)
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| |
Collapse
|
5
|
Vanderlinden G, Vandenberghe R, Vandenbulcke M, Van Laere K. The Current Role of Tau PET Imaging in Neurodegeneration. Semin Nucl Med 2025:S0001-2998(25)00031-5. [PMID: 40263023 DOI: 10.1053/j.semnuclmed.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Neurodegenerative tauopathies are characterized by the pathological hyperphosphorylation of tau proteins that subsequently form aggregates. Tau PET tracers with affinity to bind these pathological tau aggregates have been developed to measure disease progression and to support therapeutic drug development. In this review, we summarize the pathophysiology of tau throughout the range of neurodegenerative tauopathies. We outline the available first- and second-generation tau PET tracers, with a focus on new tau PET tracer developments, and discuss the quantification of tau PET images. Next, we summarize how tau PET relates to cerebrospinal fluid and plasma tau biomarkers. Finally, we review the current recommendations on the clinical use of tau PET versus fluid tau biomarkers in diagnosis, prognosis and treatment development.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Research Group Psychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Soni P, Sharma SM, Pieper AA, Paul BD, Thomas B. Nrf2/Bach1 signaling axis: A promising multifaceted therapeutic strategy for Alzheimer's disease. Neurotherapeutics 2025; 22:e00586. [PMID: 40199685 PMCID: PMC12047509 DOI: 10.1016/j.neurot.2025.e00586] [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: 10/31/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, which continues to elude effective treatment despite decades of research and numerous clinical trials. While existing therapeutic strategies have primarily targeted neuropathological hallmarks such as amyloid plaques and tau tangles, they have failed to halt disease progression, leaving patients with limited options. This persistent failure reveals a critical gap in our understanding of AD and calls for a fresh perspective - one that goes beyond the traditional targets and dives deeper into the fundamental cellular processes that drive neurodegeneration. Recent advances in molecular biology underscore the significance of nuclear factor E2-related factor 2 (Nrf2), often termed the "guardian of redox homeostasis," in the pathophysiology of AD. Nrf2 orchestrates cellular responses to oxidative stress and neuroinflammation - two interlinked pathological features of AD. In the brains of AD patients, Nrf2 activity is diminished, weakening the brain's ability to counteract oxidative damage. Additionally, the BTB and CNC homology 1 (Bach1) protein, a transcriptional repressor of Nrf2, has emerged as a potential therapeutic target. Here, we review the current landscape of clinical trials in AD and identify the limitations of the conventional approaches. We then explore the prospects of a novel approach that combines Nrf2 activation with Bach1 inhibition to achieve a multipronged defense against oxidative stress, neuroinflammation, and other molecular culprits driving AD. This innovative strategy holds promise for synergistically modulating multiple neuroprotective pathways to advance AD treatment.
Collapse
Affiliation(s)
- Priyanka Soni
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Bobby Thomas
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
7
|
Tandon R, Zhao L, Watson CM, Sarkar N, Elmor M, Heilman C, Sanders K, Hales CM, Yang H, Loring DW, Goldstein FC, Hanfelt JJ, Duong DM, Johnson ECB, Wingo AP, Wingo TS, Roberts BR, Seyfried NT, Levey AI, Lah JJ, Mitchell CS. Stratifying risk of Alzheimer's disease in healthy middle-aged individuals with machine learning. Brain Commun 2025; 7:fcaf121. [PMID: 40226382 PMCID: PMC11986205 DOI: 10.1093/braincomms/fcaf121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025] Open
Abstract
Alzheimer's disease has a prolonged asymptomatic phase during which pathological changes accumulate before clinical symptoms emerge. This study aimed to stratify the risk of clinical disease to inform future disease-modifying treatments. Cerebrospinal fluid analysis from participants in the Emory Healthy Brain Study was used to classify individuals based on amyloid beta 42 (Aβ42), total tau (tTau) and phosphorylated tau (pTau) levels. Cognitively normal (CN), biomarker-positive (CN)/BM+individuals were identified using a tTau: Aβ42 ratio > 0.24, determined by Gaussian mixture models. CN/BM+ individuals (n = 134) were classified as having asymptomatic Alzheimer's disease (AsymAD), while CN, biomarker-negative (CN/BM-) individuals served as controls (n = 134). Cognitively symptomatic, biomarker-positive individuals with an Alzheimer's disease diagnosis confirmed by the Emory Cognitive Neurology Clinic were labelled as Alzheimer's disease (n = 134). Study groups were matched for age, sex, race and education. Cerebrospinal fluid samples from these matched Emory Healthy Brain Study groups were analysed using targeted proteomics via selected reaction monitoring mass spectrometry. The targeted cerebrospinal fluid panel included 75 peptides from 58 unique proteins. Machine learning approaches identified a subset of eight peptides (ADQDTIR, AQALEQAK, ELQAAQAR, EPVAGDAVPGPK, IASNTQSR, LGADMEDVCGR, VVSSIEQK, YDNSLK) that distinguished between CN/BM- and symptomatic Alzheimer's disease samples with a binary classifier area under the curve performance of 0.98. Using these eight peptides, Emory Healthy Brain Study AsymAD cases were further stratified into 'Control-like' and 'Alzheimer's disease-like' subgroups, representing varying levels of risk for developing clinical disease. The eight peptides were evaluated in an independent dataset from the Alzheimer's Disease Neuroimaging Initiative, effectively distinguishing CN/BM- from symptomatic Alzheimer's disease cases (area under the curve = 0.89) and stratifying AsymAD individuals into control-like and Alzheimer's disease-like subgroups (area under the curve = 0.89). In the absence of matched longitudinal data, an established cross-sectional event-based disease progression model was employed to assess the generalizability of these peptides for risk stratification. In summary, results from two independent modelling methods and datasets demonstrate that the identified eight peptides effectively stratify the risk of progression from asymptomatic to symptomatic Alzheimer's disease.
Collapse
Affiliation(s)
- Raghav Tandon
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Center for Machine Learning, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - Caroline M Watson
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Neel Sarkar
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Morgan Elmor
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Craig Heilman
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Katherine Sanders
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Chadwick M Hales
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Huiying Yang
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - David W Loring
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Felicia C Goldstein
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - John J Hanfelt
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - Duc M Duong
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Erik C B Johnson
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory School of Medicine, Atlanta, GA 30322, USA
- Division of Mental Health, Atlanta VA Medical Center, Atlanta, GA 30033, USA
| | - Thomas S Wingo
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Blaine R Roberts
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Nicholas T Seyfried
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Allan I Levey
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - James J Lah
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Center for Machine Learning, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
8
|
Sun Y, Liu Z, Zhang Z, Kang Y, Wang X, Zhang Y, Liu Y, Zhao P. Human induced pluripotent stem cell models for Alzheimer's disease research: a bibliometric analysis. Front Hum Neurosci 2025; 19:1548701. [PMID: 40177166 PMCID: PMC11962003 DOI: 10.3389/fnhum.2025.1548701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Alzheimer's disease (AD), the leading cause of dementia, remains without adequate treatment. Current models do not fully replicate human physiology and pathology. The advent of human induced pluripotent stem cell (hiPSC) technology offers a novel approach to studying AD. Methods Our study conducted a bibliometric analysis to assess the application and development of hiPSC technology in AD research. We retrieved 531 articles on hiPSC models of AD from the Web of Science Core Collection, published between January 2010 and June 2024. CiteSpace and VOSviewer were used to analyze authorship, geographic contributions, journal influence, and citation patterns. Results Our findings reveal a steady increase in publications over 14 years, with the United States leading in contributions, followed by China. Li-Huei Tsai from the Massachusetts Institute of Technology is a prominent researcher. PLoS One emerges as the most influential journal. Research trends have focused on inflammation, astrocytes, microglia, apolipoprotein E (ApoE), and tau. Discussion Bibliometric analysis is crucial in identifying research gaps and trends and guiding future studies to address unmet needs in understanding and modeling human physiology and pathology. Leveraging hiPSC models to investigate the molecular mechanisms of familial and sporadic AD is expected to provide a crucial foundation for developing future treatment strategies. Conclusion In summary, the bibliometric findings from this study provide a comprehensive overview of the current research landscape in hiPSC models for AD. It also highlights emerging trends and research gaps, crucial for guiding future research efforts, particularly in exploring novel therapeutic targets and improving understanding of disease mechanisms.
Collapse
Affiliation(s)
- Yuning Sun
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zhilong Liu
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zongbo Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yufeng Kang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinlian Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yiping Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Pei Zhao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| |
Collapse
|
9
|
Leuzy A, Bollack A, Pellegrino D, Teunissen CE, La Joie R, Rabinovici GD, Franzmeier N, Johnson K, Barkhof F, Shaw LM, Arkhipenko A, Schindler SE, Honig LS, Moscoso Rial A, Schöll M, Zetterberg H, Blennow K, Hansson O, Farrar G. Considerations in the clinical use of amyloid PET and CSF biomarkers for Alzheimer's disease. Alzheimers Dement 2025; 21:e14528. [PMID: 40042435 PMCID: PMC11881640 DOI: 10.1002/alz.14528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 03/09/2025]
Abstract
Amyloid-β (Aβ) positron emission tomography (PET) imaging and cerebrospinal fluid (CSF) biomarkers are now established tools in the diagnostic workup of patients with Alzheimer's disease (AD), and their use is anticipated to increase with the introduction of new disease-modifying therapies. Although these biomarkers are comparable alternatives in research settings to determine Aβ status, biomarker testing in clinical practice requires careful consideration of the strengths and limitations of each modality, as well as the specific clinical context, to identify which test is best suited for each patient. This article provides a comprehensive review of the pathologic processes reflected by Aβ-PET and CSF biomarkers, their performance, and their current and future applications and contexts of use. The primary aim is to assist clinicians in making better-informed decisions about the suitability of each biomarker in different clinical situations, thereby reducing the risk of misdiagnosis or incorrect interpretation of biomarker results. HIGHLIGHTS: Recent advances have positioned Aβ PET and CSF biomarkers as pivotal in AD diagnosis. It is crucial to understand the differences in the clinical use of these biomarkers. A team of experts reviewed the state of Aβ PET and CSF markers in clinical settings. Differential features in the clinical application of these biomarkers were reviewed. We discussed the role of Aβ PET and CSF in the context of novel plasma biomarkers.
Collapse
Grants
- AF-930351 Neurodegenerative Disease Research
- 101053962 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- R01 AG066107 NIA NIH HHS
- FO2022-0270 Bluefield Project, Olav Thon Foundation, Erling-Persson Family Foundation
- 101112145 European Union's Horizon Europe
- Alzheimer Netherlands
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- 2022-0231 Knut and Alice Wallenberg foundation
- KAW 2023.0371 Knut and Alice Wallenberg Foundation
- U19 ADNI4 Harvard Aging Brain Study
- R01 AG081394 NIA NIH HHS
- ADRC P30-AG-072979 Harvard Aging Brain Study
- 2022-1259 Regionalt Forskningsstöd
- Shanendoah Foundation
- 2020-O000028 Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, Skåne University Hospital Foundation
- The Selfridges Group Foundation
- R56 AG057195 NIA NIH HHS
- U01 NS100600 NINDS NIH HHS
- ALZ2022-0006 Hjärnfonden, Sweden
- U01 AG057195 NIA NIH HHS
- Dutch National Dementia Strategy
- ZEN24-1069572 Alzheimer's Association
- R01AG072474 Harvard Aging Brain Study
- 860197 Marie Curie International Training Network
- AF-939721 Neurodegenerative Disease Research
- R01 AG070941 NIA NIH HHS
- P01 AG036694 NIA NIH HHS
- JPND2021-00694 Neurodegenerative Disease Research
- ADSF-21-831376-C AD Strategic Fund, and Alzheimer's Association
- AF-994900 Swedish Alzheimer Foundation
- NIH
- ALFGBG-813971 County Councils, the ALF-agreement
- FO2021-0293 Swedish Brain Foundation
- U19AG063893 NINDS NIH HHS
- 2022-01018 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 201809-2016862 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 831434 Innovative Medicines Initiatives 3TR
- 101132933 European Union's Horizon Europe
- European Union Joint Programme
- Cure Alzheimer's fund, Rönström Family Foundation
- ID 390857198 Munich Cluster for Systems Neurology
- U01-AG057195 NIA NIH HHS
- Deutsche Forschungsgemeinschaft
- 2021-06545 Swedish Research Council
- Sahlgrenska Academy at the University of Gothenburg
- U19 AG024904 NIA NIH HHS
- GE Healthcare
- JPND2019-466-236 European Union Joint Program for Neurodegenerative Disorders
- P30 AG062422 NIA NIH HHS
- ADG-101096455 European Research Council
- 2022-00732 Neurodegenerative Disease Research
- 860197 Marie Skłodowska-Curie
- P01 AG019724 NIA NIH HHS
- U01NS100600 NINDS NIH HHS
- AF-980907 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- P30 AG066462 NIA NIH HHS
- 2022-00775 GHR Foundation, Swedish Research Council
- R44 AG071388 NIA NIH HHS
- FO2017-0243 Hjärnfonden, Sweden
- AF-968270 Neurodegenerative Disease Research
- KAW2014.0363 Knut and Alice Wallenberg Foundation
- SG-23-1061717 Alzheimer's Association
- 2021-02678 Swedish Research Council
- R01 AG059013 NIA NIH HHS
- R35 AG072362 NIA NIH HHS
- VGFOUREG-995510 Västra Götaland Region R&D
- American College of Radiology
- R01 AG081394-01 European Union's Horizon Europe
- R21 AG070768 NIA NIH HHS
- U19 AG063893 NIA NIH HHS
- 2022-Projekt0080 Swedish Federal Government under the ALF agreement
- ALFGBG-965326 County Councils, the ALF-agreement
- Alzheimer Drug Discovery Foundation
- Rainwater Charitable Foundation
- Research of the European Commission
- R01AG083740 National Institute of Aging
- ADSF-21-831381-C AD Strategic Fund, and Alzheimer's Association
- SG-23-1038904 Alzheimer's Association 2022-2025
- RS-2023-00263612 National Research Foundation of Korea
- P30-AG062422 NIA NIH HHS
- R21AG070768 Harvard Aging Brain Study
- 2017-02869 Swedish Research Council
- 101034344 Joint Undertaking
- ALFGBG-715986 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- ERAPERMED2021-184 ERA PerMed
- U19AG024904 Harvard Aging Brain Study
- R01 AG072474 NIA NIH HHS
- UKDRI-1003 Neurodegenerative Disease Research
- 10510032120003 Health Holland, the Dutch Research Council
- 2019-02397 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- EXC 2145 SyNergy Munich Cluster for Systems Neurology
- 1412/22 Parkinson foundation of Sweden
- R01 AG046396 NIA NIH HHS
- ALFGBG-71320 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- P01-AG019724 NIA NIH HHS
- ALFGBG-965240 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- Deutsche Parkinson Gesellschaft
- ADSF-21-831377-C AD Strategic Fund, and Alzheimer's Association
- National MS Society
- R01 AG083740 NIA NIH HHS
- 2017-00915 Neurodegenerative Disease Research
- 2023-06188 Swedish Research Council
- Alzheimer Association
- National MS Society
- Alzheimer Netherlands
- NIH
- NIA
- National Institute of Neurological Disorders and Stroke
- American College of Radiology
- Rainwater Charitable Foundation
- Deutsche Forschungsgemeinschaft
- NINDS
- Knut and Alice Wallenberg Foundation
- Swedish Research Council
- National Research Foundation of Korea
- Swedish Brain Foundation
- European Research Council
- Alzheimer's Association
- GE Healthcare
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
| | - Ariane Bollack
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
- Department of Medical Physics and BioengineeringCentre for Medical Image Computing (CMIC)University College LondonLondonUK
| | | | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam NeuroscienceNeurodegenerationAmsterdam UMC Vrije UniversiteitAmsterdamThe Netherlands
| | - Renaud La Joie
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Nicolai Franzmeier
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Institute for Stroke and Dementia Research (ISD)University HospitalLMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Keith Johnson
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentBrigham and Women's HospitalBostonMassachusettsUSA
| | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineVrije Universiteit AmsterdamAmsterdam University Medical CenterAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain imagingAmsterdamThe Netherlands
- UCL Queen Square Institute of Neurology and Center for Medical Image ComputingUniversity College LondonLondonUK
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Suzanne E. Schindler
- Department of NeurologyKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Lawrence S. Honig
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Alexis Moscoso Rial
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Nuclear Medicine Department and Molecular Imaging GroupInstituto de Investigación Sanitaria de Santiago de CompostelaSantiago de CompostelaSpain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
- Dementia Research CentreInstitute of NeurologyUniversity College LondonLondonUK
| | - Henrik Zetterberg
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesScience ParkHong KongChina
- Wisconsin Alzheimer's Disease Research CenterSchool of Medicine and Public HealthUniversity of WisconsinUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiChina
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Gill Farrar
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
| |
Collapse
|
10
|
Calvin-Dunn KN, Mcneela A, Leisgang Osse A, Bhasin G, Ridenour M, Kinney JW, Hyman JM. Electrophysiological insights into Alzheimer's disease: A review of human and animal studies. Neurosci Biobehav Rev 2025; 169:105987. [PMID: 39732222 DOI: 10.1016/j.neubiorev.2024.105987] [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: 01/22/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
This review highlights the crucial role of neuroelectrophysiology in illuminating the mechanisms underlying Alzheimer's disease (AD) pathogenesis and progression, emphasizing its potential to inform the development of effective treatments. Electrophysiological techniques provide unparalleled precision in exploring the intricate networks affected by AD, offering insights into the synaptic dysfunction, network alterations, and oscillatory abnormalities that characterize the disease. We discuss a range of electrophysiological methods, from non-invasive clinical techniques like electroencephalography and magnetoencephalography to invasive recordings in animal models. By drawing on findings from these studies, we demonstrate how electrophysiological research has deepened our understanding of AD-related network disruptions, paving the way for targeted therapeutic interventions. Moreover, we underscore the potential of electrophysiological modalities to play a pivotal role in evaluating treatment efficacy. Integrating electrophysiological data with clinical neuroimaging and longitudinal studies holds promise for a more comprehensive understanding of AD, enabling early detection and the development of personalized treatment strategies. This expanded research landscape offers new avenues for unraveling the complexities of AD and advancing therapeutic approaches.
Collapse
Affiliation(s)
- Kirsten N Calvin-Dunn
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, United States.
| | - Adam Mcneela
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States
| | - A Leisgang Osse
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - G Bhasin
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| | - M Ridenour
- Department of Psychology, University of Nevada, Las Vegas, United States
| | - J W Kinney
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - J M Hyman
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| |
Collapse
|
11
|
By S, Kahl A, Cogswell PM. Alzheimer's Disease Clinical Trials: What Have We Learned From Magnetic Resonance Imaging. J Magn Reson Imaging 2025; 61:579-594. [PMID: 39031716 DOI: 10.1002/jmri.29462] [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: 03/04/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of cognitive impairment and dementia worldwide with rising prevalence, incidence and mortality. Despite many decades of research, there remains an unmet need for disease-modifying treatment that can significantly alter the progression of disease. Recently, with United States Food and Drug Administration (FDA) drug approvals, there have been tremendous advances in this area, with agents demonstrating effects on cognition and biomarkers. Magnetic resonance imaging (MRI) plays an instrumental role in these trials. This review article aims to outline how MRI is used for screening eligibility, monitoring safety and measuring efficacy in clinical trials, leaning on the landscape of past and recent AD clinical trials that have used MRI as examples; further, insight on promising MRI biomarkers for future trials is provided. LEVEL OF EVIDENCE: 1. TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Samantha By
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Anja Kahl
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | |
Collapse
|
12
|
la Torre A, Lo Vecchio F, Angelillis VS, Gravina C, D’Onofrio G, Greco A. Reinforcing Nrf2 Signaling: Help in the Alzheimer's Disease Context. Int J Mol Sci 2025; 26:1130. [PMID: 39940900 PMCID: PMC11818887 DOI: 10.3390/ijms26031130] [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: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Oxidative stress plays a role in various pathophysiological diseases, including neurogenerative diseases, such as Alzheimer's disease (AD), which is the most prevalent neuro-pathology in the aging population. Oxidative stress has been reported to be one of the earliest pathological alterations in AD. Additionally, it was demonstrated that in older adults, there is a loss of free radical scavenging ability. The Nrf2 transcription factor is a key regulator in antioxidant defense systems, but, with aging, both the amount and the transcriptional activity of Nrf2 decrease. With the available treatments for AD being poorly effective, reinforcing the antioxidant defense systems via the Nrf2 pathway may be a way to prevent and treat AD. To highlight the predominant role of Nrf2 signaling in defending against oxidative stress and, therefore, against neurotoxicity, we present an overview of the natural compounds that exert their own neuroprotective roles through the activation of the Nrf2 pathway. This review is an opportunity to promote a holistic approach in the treatment of AD and to highlight the need to further refine the development of new potential Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Annamaria la Torre
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Filomena Lo Vecchio
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Valentina Soccorsa Angelillis
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| | - Carolina Gravina
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Grazia D’Onofrio
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Antonio Greco
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| |
Collapse
|
13
|
Ruiz-Uribe NE, Manser P, Butcher B, Li Y, Blendstrup M, Baker S, Sanabria Bohorquez S, Teng E. Cross-sectional and prognostic associations of baseline [ 18F]GTP1 tau PET signal and white matter lesion volumes for cognitive and functional decline in prodromal-to-mild Alzheimer's disease. J Alzheimers Dis 2025; 103:465-475. [PMID: 39801050 DOI: 10.1177/13872877241302497] [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: 02/06/2025]
Abstract
BACKGROUND In Alzheimer's disease (AD), tau and white matter lesion pathology are associated with clinical severity and subsequent decline, but their relative relationships with clinical assessments remain uncertain. OBJECTIVE To examine cross-sectional and prognostic associations between baseline [18F]GTP1 tau positron emission tomography (PET) standardized uptake value ratio (SUVRs) and T1 white matter hypointensity (WMHypo) volumes with clinical indices. METHODS We analyzed participants with biomarker-confirmed prodromal (n = 127) or mild (n = 233) AD with baseline [18F]GTP1 tau PET and MRI and longitudinal Clinical Dementia Rating-Sum of Boxes (CDR-SB), 13-item version of the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog13), Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), Mini-Mental Status Examination (MMSE), and Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) data. RESULTS Higher baseline [18F]GTP1 SUVRs were independently associated with poorer baseline performance and faster rates of subsequent decline on all five clinical outcome measures. Higher baseline WMHypo volumes were independently associated with poorer baseline performance on the CDR-SB, ADAS-Cog13, RBANS, and MMSE and faster rates of subsequent decline on the CDR-SB and ADCS-ADL. CONCLUSIONS The independent associations of tau and white matter lesion pathology with clinical decline in AD suggest future prognostic models should include both imaging modalities.
Collapse
Affiliation(s)
- Nancy E Ruiz-Uribe
- Genentech, Inc., South San Francisco, CA, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Paul Manser
- Genentech, Inc., South San Francisco, CA, USA
| | | | - Yihao Li
- Genentech, Inc., South San Francisco, CA, USA
| | | | - Suzanne Baker
- Genentech, Inc., South San Francisco, CA, USA
- Department of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | - Edmond Teng
- Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
14
|
Zhang Y, Chen J, Li Y, Jiao B, Luo S. Disease-modifying therapies for Alzheimer's disease: Clinical trial progress and opportunity. Ageing Res Rev 2025; 103:102595. [PMID: 39581354 DOI: 10.1016/j.arr.2024.102595] [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: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
The U.S. Food and Drug Administration (FDA) recently approved lecanemab and donanemab for the treatment of early symptomatic Alzheimer's disease (AD) after their phase III trials reached endpoints. These two anti-amyloid β monoclonal antibodies represent the latest promise of disease-modifying therapy (DMT) for AD, which undoubtedly reignites new hope for DMTs to combat the staggering financial and human costs of AD. However, in addition to these two successful antibodies, there have been enormous efforts to develop DMTs in various aspects to meet the therapeutic requirement of AD. In this review, we delineate the core principles and methodologies of diverse DMTs, covering the advances in clinical trials of drug candidates that either have been discontinued, completed, or are ongoing, as well as brain stimulation and lifestyle interventions. In addition, by overseeing the fate of various candidate molecules, we hope to provide references and ideas for prospective approaches and promising applications of DTMs for AD, particularly in terms of universality and clinical application economics, to optimize efficacy and maximize AD patient benefits in the future.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Xiangya School of Medicine, Central South University, Changsha 410013, PR China
| | - Jie Chen
- Department of Rehabilitation, Xiangya Boai Rehabilitation Hospital, Changsha 410100, PR China
| | - Yanru Li
- Department of Rehabilitation, Xiangya Boai Rehabilitation Hospital, Changsha 410100, PR China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410000, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha 410008, PR China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410000, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha 410008, PR China.
| |
Collapse
|
15
|
Zhuang X, Lin J, Song Y, Ban R, Zhao X, Xia Z, Wang Z, Zhang G. The Interplay Between Accumulation of Amyloid-Beta and Tau Proteins, PANoptosis, and Inflammation in Alzheimer's Disease. Neuromolecular Med 2024; 27:2. [PMID: 39751702 DOI: 10.1007/s12017-024-08815-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is a common progressive neurodegenerative disorder, and the vast majority of cases occur in elderly patients. Recently, the accumulation of Aβ and tau proteins has drawn considerable attention in AD research. This review explores the multifaceted interactions between these proteins and their contribution to the pathological landscape of AD, encompassing synaptic dysfunction, neuroinflammation, and PANoptosis. PANoptosis is a collective term for programmed cell death (PCD) modalities that encompass elements of apoptosis, pyroptosis, and necroptosis. The accumulation of Aβ peptides and tau proteins, along with the immune response in brain cells, may trigger PANoptosis, thus advancing the progression of the disease. Recent advancements in molecular imaging and genetics have provided deeper insights into the interactions between Aβ peptides, tau proteins, and the immune response. The review also discusses the role of mitochondrial dysregulation in AD. The exploration of the interplay between neurodegeneration, immune responses, and cell death offers promising avenues for the development of innovative treatments.
Collapse
Affiliation(s)
- Xianbo Zhuang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Jie Lin
- School of Basic Medicine Sciences, Shandong University, Jinan, China
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Yamin Song
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Ru Ban
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Xin Zhao
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, 250012, China.
- Department of Neurology, the Second People's Hospital of Liaocheng, Liaocheng, China.
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, China.
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
| |
Collapse
|
16
|
Niazi SK. Safety Concerns in Neurological Clinical Trials: A Challenge That the FDA Must Resolve. Biomedicines 2024; 12:2918. [PMID: 39767824 PMCID: PMC11673353 DOI: 10.3390/biomedicines12122918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Monoclonal antibodies approved by the FDA, lecanemab, donanemab, and aducanumab, are failing to meet the expected efficacy to treat early Alzheimer's disease, and aducanumab has been recalled. Methods: Recently, it was reported that the clinical trials of these antibodies may have violated patient's rights and subjected them to high, likely lethal risk. The challenge with developing antibodies to treat neurological disorders is their poor blood-brain barrier (BBB) penetration if the antibody must enter the brain, resulting in almost negligible brain bioavailability, requiring high dosing that can be toxic. Results: The reported efficacy of these drugs should also be reviewed, considering the placebo effects, since all antibodies have shown severe side effects that are not prevented by the placebo responses. In this critical and urgent advice to the FDA, I am suggesting a guideline amendment to all clinical trials requiring proof of sufficient brain bioavailability at the site of action, where it is known. Conclusions: For antibodies to cross the blood-brain barrier, there are proven options such as conjugating with transferrin protein, making clinical trials in its absence more questionable.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Pini L, Lista S, Griffa A, Allali G, Imbimbo BP. Can brain network connectivity facilitate the clinical development of disease-modifying anti-Alzheimer drugs? Brain Commun 2024; 7:fcae460. [PMID: 39741782 PMCID: PMC11686405 DOI: 10.1093/braincomms/fcae460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
The preclinical phase of Alzheimer's disease represents a crucial time window for therapeutic intervention but requires the identification of clinically relevant biomarkers that are sensitive to the effects of disease-modifying drugs. Amyloid peptide and tau proteins, the main histological hallmarks of Alzheimer's disease, have been widely used as biomarkers of anti-amyloid and anti-tau drugs. However, these biomarkers do not fully capture the multiple biological pathways of the brain. Indeed, robust amyloid-target engagement by anti-amyloid monoclonal antibodies has recently translated into modest cognitive and clinical benefits in Alzheimer's disease patients, albeit with potentially life-threatening side effects. Moreover, targeting the tau pathway has yet to result in any positive clinical outcomes. Findings from computational neuroscience have demonstrated that brain regions do not work in isolation but are interconnected within complex network structures. Brain connectivity studies suggest that misfolded proteins can spread through these connections, leading to the hypothesis that Alzheimer's disease is a pathology of network disconnectivity. Based on these assumptions, here we discuss how incorporating brain connectivity outcomes could better capture global brain functionality and, in conjunction with traditional Alzheimer's disease biomarkers, could facilitate the clinical development of new disease-modifying anti-Alzheimer's disease drugs.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience, Università degli Studi di Padova, 35121 Padova, Italy
- Padova Neuroscience Center, Università degli Studi di Padova, 35121 Padova, Italy
| | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain
| | - Alessandra Griffa
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Montpaisible 16, 1011 Lausanne, Switzerland
- Medical Image Processing Laboratory, Neuro-X Institute, École Polytechnique Fédérale De Lausanne (EPFL), Campus Biotech Chemin des Mines 9, 1202 Geneva, Switzerland
| | - Gilles Allali
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Montpaisible 16, 1011 Lausanne, Switzerland
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, 43122 Parma, Italy
| |
Collapse
|
18
|
Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, Golabi B, Aletaha R, Motlagh Asghari K, Hamidi S, Mousavi SE, Jamalkhani S, Karamzad N, Shamekh A, Mohammadinasab R, Sullman MJM, Şahin F, Kolahi AA. Alzheimer's disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024; 11:1474043. [PMID: 39736972 PMCID: PMC11682909 DOI: 10.3389/fmed.2024.1474043] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Background Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired reasoning. It is the leading cause of dementia in older adults, marked by the pathological accumulation of amyloid-beta plaques and neurofibrillary tangles. These pathological changes lead to widespread neuronal damage, significantly impacting daily functioning and quality of life. Objective This comprehensive review aims to explore various aspects of Alzheimer's disease, including its epidemiology, risk factors, clinical presentation, diagnostic advancements, management strategies, caregiving challenges, and emerging therapeutic interventions. Methods A systematic literature review was conducted across multiple electronic databases, including PubMed, MEDLINE, Cochrane Library, and Scopus, from their inception to May 2024. The search strategy incorporated a combination of keywords and Medical Subject Headings (MeSH) terms such as "Alzheimer's disease," "epidemiology," "risk factors," "symptoms," "diagnosis," "management," "caregiving," "treatment," and "novel therapies." Boolean operators (AND, OR) were used to refine the search, ensuring a comprehensive analysis of the existing literature on Alzheimer's disease. Results AD is significantly influenced by genetic predispositions, such as the apolipoprotein E (APOE) ε4 allele, along with modifiable environmental factors like diet, physical activity, and cognitive engagement. Diagnostic approaches have evolved with advances in neuroimaging techniques (MRI, PET), and biomarker analysis, allowing for earlier detection and intervention. The National Institute on Aging and the Alzheimer's Association have updated diagnostic criteria to include biomarker data, enhancing early diagnosis. Conclusion The management of AD includes pharmacological treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, which provide symptomatic relief but do not slow disease progression. Emerging therapies, including amyloid-beta and tau-targeting treatments, gene therapy, and immunotherapy, offer potential for disease modification. The critical role of caregivers is underscored, as they face considerable emotional, physical, and financial burdens. Support programs, communication strategies, and educational interventions are essential for improving caregiving outcomes. While significant advancements have been made in understanding and managing AD, ongoing research is necessary to identify new therapeutic targets and enhance diagnostic and treatment strategies. A holistic approach, integrating clinical, genetic, and environmental factors, is essential for addressing the multifaceted challenges of Alzheimer's disease and improving outcomes for both patients and caregivers.
Collapse
Affiliation(s)
- Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Morsali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Golabi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aletaha
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kimia Motlagh Asghari
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Hamidi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Jamalkhani
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional, Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J. M. Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Schauer SP, Toth B, Lee J, Honigberg LA, Ramakrishnan V, Jiang J, Kollmorgen G, Bayfield A, Wild N, Hoffman J, Ceniceros R, Dolton M, Bohórquez SMS, Hoogenraad CC, Wildsmith KR, Teng E, Monteiro C, Anania V, Yeh FL. Pharmacodynamic effects of semorinemab on plasma and CSF biomarkers of Alzheimer's disease pathophysiology. Alzheimers Dement 2024; 20:8855-8866. [PMID: 39513754 DOI: 10.1002/alz.14346] [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: 05/15/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Semorinemab, an anti-tau monoclonal antibody, was assessed in two Phase II trials for Alzheimer's disease (AD). Plasma and cerebrospinal fluid (CSF) biomarkers provided insights into the drug's potential mechanism of action. METHODS Qualified assays were used to measure biomarkers of tau, amyloidosis, glial activity, neuroinflammation, synaptic function, and neurodegeneration from participant samples in Tauriel (NCT03289143) and Lauriet (NCT03828747) Phase II trials. RESULTS Plasma phosphorylated Tau 181 (pTau181) and CSF chitinase-3-like protein 1 (YKL-40) increased following semorinemab treatment in both studies. In Lauriet, increasing plasma glial fibrillary protein (GFAP) concentrations stabilized with semorinemab, while this was not observed in Tauriel. Other AD pathophysiology biomarkers showed no consistent response to semorinemab. DISCUSSION Increases in CSF YKL-40 suggest that semorinemab may stimulate microglia activation in the presence of AD-associated Tau pathology, but not in healthy controls. Stabilization of plasma GFAP in Lauriet indicates a possible impact on reactive gliosis in mild-to-moderate AD. TRIAL REGISTRATION Tauriel ClinicalTrials.gov Identifier: NCT03289143. Lauriet ClinicalTrials.gov Identifier: NCT03828747. Phase 1 ClinicalTrials.gov Identifier: NCT02820896. HIGHLIGHTS AD pathophysiology biomarkers were measured to assess the mechanism of action. Semorinemab increased CSF YKL-40 in participants with AD but not in healthy controls. Semorinemab possibly stabilized plasma GFAP in the Lauriet trial. Semorinemab treatment may activate microglia and moderate reactive gliosis.
Collapse
Affiliation(s)
- Stephen P Schauer
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Balazs Toth
- Department of Data and Statistical Sciences, Immunology, Infectious Diseases, and Ophthalmology, Genentech Inc., South San Francisco, California, USA
| | - Julie Lee
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Lee A Honigberg
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Vidya Ramakrishnan
- Department of Clinical Pharmacology, Modeling and Simulation, Development Sciences, Genentech Inc., South San Francisco, California, USA
| | - Jenny Jiang
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | | | | | | | - Jennifer Hoffman
- Department of Translational Medicine, Operations Diagnostics Technology Innovation, Genentech Inc., South San Francisco, California, USA
| | - Ryan Ceniceros
- Department of Early Clinical Development, Clinical Operations, Genentech Inc., South San Francisco, California, USA
| | - Michael Dolton
- Department of Clinical Pharmacology, Modeling and Simulation, Development Sciences, Genentech Inc., South San Francisco, California, USA
| | - Sandra M Sanabria Bohórquez
- Department of Early Clinical Development, Clinical Imaging Group, Genentech Inc., South San Francisco, California, USA
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Kristin R Wildsmith
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Edmond Teng
- Department of Early Clinical Development, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Cecilia Monteiro
- Department of Early Clinical Development, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Veronica Anania
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| | - Felix L Yeh
- Department of Translational Medicine, OMNI, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
20
|
Kovacech B, Cullen NC, Novak P, Hanes J, Kontsekova E, Katina S, Parrak V, Fresser M, Vanbrabant J, Feldman HH, Winblad B, Stoops E, Vanmechelen E, Zilka N. Post hoc analysis of ADAMANT, a phase 2 clinical trial of active tau immunotherapy with AADvac1 in patients with Alzheimer's disease, positive for plasma p-tau217. Alzheimers Res Ther 2024; 16:254. [PMID: 39580468 PMCID: PMC11585249 DOI: 10.1186/s13195-024-01620-7] [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: 09/13/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The spread of tau pathology closely correlates with the disease course and cognitive decline in Alzheimer's disease (AD). Tau-targeting immunotherapies are being developed to stop the spread of tau pathology and thus halt disease progression. In this post hoc analysis of the ADAMANT clinical trial, we examined the performance of AADvac1, an active immunotherapy targeting the microtubule-binding region (MTBR) of tau, in a subgroup of participants with elevated plasma p-tau217, indicating AD-related neuropathological changes. METHODS ADAMANT was a 24-month, randomized, placebo-controlled, parallel-group, double-blinded, multicenter, phase 2 clinical trial in subjects with mild AD. The trial participants were randomized 3:2 to receive six doses of AADvac1 or placebo at 4-week intervals, followed by five booster doses at 14-week intervals. The primary outcome was safety. The secondary outcomes were the Clinical Dementia Rating-Sum of Boxes (CDR-SB), the Alzheimer's Disease Cooperative Study - Activities of Daily Living score for Mild Cognitive Impairment 18-item version (ADCS-ADL-MCI-18), and immunogenicity. Volumetric MRI, plasma neurofilament light (NfL), and glial fibrillary acidic protein (GFAP) were exploratory outcomes. The inclusion criterion for this post-hoc analysis was a baseline plasma p-tau217 level above the cutoff for AD. RESULTS Among 196 ADAMANT participants, 137 were positive for plasma p-tau217 (mean age 71.4 years, 59% women). AADvac1 was safe and well tolerated in this subgroup. AADvac1 reduced the rate of accumulation of log-plasma NfL by 56% and that of GFAP by 73%. The treatment differences in the CDR-SB and ADCS-ADL-MCI-18 scores favored AADvac1 but were not statistically significant. AADvac1 had no effect on whole-brain volume but nonsignificantly reduced the loss of brain cortical tissue in several regions. Importantly, the impact on the study outcomes was more pronounced in participants with higher anti-tau antibody levels. CONCLUSIONS These results suggest that AADvac1 tau immunotherapy can reduce plasma biomarkers of neurodegeneration and neuroinflammation. These findings and possible observations on brain atrophy and cognition are hypothesis-generating and warrant further evaluation in a larger clinical trial. TRIAL REGISTRATION EudraCT 2015-000630-30 (primary) and NCT02579252.
Collapse
Affiliation(s)
- Branislav Kovacech
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Clinical Research Centre, Jan Waldenströms Gata 35, 202 13, Malmö, Sweden
| | - Petr Novak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Jozef Hanes
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Eva Kontsekova
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102 Bratislava, Slovakia and Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84510, Slovakia
| | - Stanislav Katina
- Department of Mathematics and Statistics, Axon Neuroscience R&D Services SE, Bratislava, Slovakia, and (current) Masaryk University, Kotlářská 267/2, Brno, 611 37, Czech Republic
| | - Vojtech Parrak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Michal Fresser
- Axon Neuroscience SE, 4 Arch. Makariou & Kalogreon, 6016, Larnaca, Cyprus
| | | | - Howard H Feldman
- Department of Neurosciences, Alzheimer's Disease Cooperative Study, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 86, Stockholm, Sweden
| | - Erik Stoops
- ADx NeuroSciences NV, Technologiepark 6, 9052, Ghent, Belgium
| | | | - Norbert Zilka
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| |
Collapse
|
21
|
Ou CM, Xue WW, Liu D, Ma L, Xie HT, Ning K. Stem cell therapy in Alzheimer's disease: current status and perspectives. Front Neurosci 2024; 18:1440334. [PMID: 39640295 PMCID: PMC11618239 DOI: 10.3389/fnins.2024.1440334] [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: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
An incurable neurogenerative illness, Alzheimer's disease, is the cause of most global health, medical, and social disasters. The two main symptoms are cognitive impairment and neuronal loss. Current medications that target tau protein tangles and Aβ plaques are not very effective because they only slow the symptoms of AD and do not repair damaged cells. Stem cell-based treatments, however, present an alternative strategy in the treatment of AD. They have the capacity to divide into specialized adult cells, have self-renewal abilities, and multiplication. Stem cells can now be employed as a donor source for cell therapy due to developments in stem cell technology. This review covers preclinical and clinical updates on studies based on targeting the tau protein tangles and Aβ plaque, as well as four types of stem cells employed in AD treatment. The review also outlines the two basic pathologic aspects, tau protein tangles and Aβ plaques, of AD.
Collapse
Affiliation(s)
- Chu-Min Ou
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Wei-Wei Xue
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Dong Liu
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Liya Ma
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Hai-Tao Xie
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Ke Ning
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
22
|
Qi X, Nizamutdinov D, Yi SS, Wu E, Huang JH. Disease Modifying Monoclonal Antibodies and Symptomatic Pharmacological Treatment for Alzheimer's Disease. Biomedicines 2024; 12:2636. [PMID: 39595200 PMCID: PMC11592475 DOI: 10.3390/biomedicines12112636] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's Disease (AD) is an irreversible, progressive syndrome characterized by neurocognitive impairment. Two neuropathological features seen in AD are extracellular amyloid plaques consisting of amyloid beta1-40 and 1-42, and intracellular neurofibrillary tangles (NFTs). For decades, neuroscience research has heavily focused on seeking to understand the primary mechanism of AD and searching for pharmacological approaches for the treatment of dementia. Three monoclonal antibodies that act against amyloid beta-aducanumab, lecanemab, and donanemab-have been approved by the Food and Drug Administration (FDA) for the treatment of mild cognitive impairment and mild AD, in addition to medications for cognitive symptom management such as acetylcholinesterase inhibitors and the N-methyl-D-aspartate (NMDA) antagonist. Further trials should focus on the combination of therapies targeting amyloid plaques and tau pathology.
Collapse
Affiliation(s)
- Xiaoming Qi
- Department of Neurology, Baylor Scott & White Health, Temple, TX 78508, USA;
| | - Damir Nizamutdinov
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Song Stephen Yi
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Erxi Wu
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
- College of Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- LIVESTRONG Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jason H. Huang
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
23
|
Armstrong C, Luo D, Gretzinger A, Pandey D, Lipchik A, Todi SV, Dutta AK. Novel Piperazine Based Compounds Target Alzheimer's Disease Relevant Amyloid β42 and Tau Derived Peptide AcPHF6, and the Lead Molecule Increases Viability in the Flies Expressing Human Tau Protein. ACS Chem Neurosci 2024; 15:3901-3914. [PMID: 39501783 DOI: 10.1021/acschemneuro.4c00220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025] Open
Abstract
Alzheimer's disease (AD) is the leading form of dementia in the United States and the world. The pathophysiology of AD is complex and multifaceted. Accumulation of senile plaques and neurofibrillary tangles (NFTs) are hallmarks of AD. The aggregation of amyloid β (senile plaques) and tau tangles (NFTs) results in the death of neurons in the cortex and hippocampus, which manifests itself in cognitive decline and memory loss. Current therapies rely on conventional approaches that have only treated the underlying symptoms without disease modification. Data from clinical studies point to a complex role of amyloid β (Aβ) in a way that enhances the tau phenotype throughout the disease process. To address the co-pathogenic role of Aβ and tau, we undertook development of multitarget compounds aiming at both tau and Aβ to slow or stop disease progression and provide neuroprotection. Here, we demonstrate a dose-dependent effect of the novel test compounds that inhibit aggregation of AcPHF6 (a shorter version of tau protein) and Aβ1-42 peptides in thioflavin T fluorescent assays. The compounds were also shown to disaggregate preformed aggregates dose dependently. To further validate these findings, circular dichroism experiments were carried out to examine the nature of inhibition. Additionally, transmission electron microscopy experiments were carried out to gain insights into the morphologies of aggregates obtained from dose-dependent inhibition of AcPHF6 and Aβ1-42 as well as dissociation of preformed aggregates from these peptides. Compounds D-687 and D-688 reversed Aβ1-42 induced toxicity in SH-SH5Y cells, significantly demonstrating neuroprotective properties. Finally, in a study with Drosophila melanogaster expressing human tau protein isoform (2N4R) in all the neurons, compound D-688 significantly increased the survival of flies compared to vehicle treated controls. Future studies will further examine the neuroprotective properties of these lead compounds in various animal models.
Collapse
Affiliation(s)
- Christopher Armstrong
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Dan Luo
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Anna Gretzinger
- Department of Pharmacology, Wayne State University, Detroit, Michigan 48201, United States
| | - Deepti Pandey
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Andrew Lipchik
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University, Detroit, Michigan 48201, United States
- Department of Neurology, Wayne State University, Detroit, Michigan 48201, United States
| | - Aloke K Dutta
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
24
|
Feldman HH, Cummings JL, Boxer AL, Staffaroni AM, Knopman DS, Sukoff Rizzo SJ, Territo PR, Arnold SE, Ballard C, Beher D, Boeve BF, Dacks PA, Diaz K, Ewen C, Fiske B, Gonzalez MI, Harris GA, Hoffman BJ, Martinez TN, McDade E, Nisenbaum LK, Palma J, Quintana M, Rabinovici GD, Rohrer JD, Rosen HJ, Troyer MD, Kim DY, Tanzi RE, Zetterberg H, Ziogas NK, May PC, Rommel A. A framework for translating tauopathy therapeutics: Drug discovery to clinical trials. Alzheimers Dement 2024; 20:8129-8152. [PMID: 39316411 PMCID: PMC11567863 DOI: 10.1002/alz.14250] [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: 05/07/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/25/2024]
Abstract
The tauopathies are defined by pathological tau protein aggregates within a spectrum of clinically heterogeneous neurodegenerative diseases. The primary tauopathies meet the definition of rare diseases in the United States. There is no approved treatment for primary tauopathies. In this context, designing the most efficient development programs to translate promising targets and treatments from preclinical studies to early-phase clinical trials is vital. In September 2022, the Rainwater Charitable Foundation convened an international expert workshop focused on the translation of tauopathy therapeutics through early-phase trials. Our report on the workshop recommends a framework for principled drug development and a companion lexicon to facilitate communication focusing on reproducibility and achieving common elements. Topics include the selection of targets, drugs, biomarkers, participants, and study designs. The maturation of pharmacodynamic biomarkers to demonstrate target engagement and surrogate disease biomarkers is a crucial unmet need. HIGHLIGHTS: Experts provided a framework to translate therapeutics (discovery to clinical trials). Experts focused on the "5 Rights" (target, drug, biomarker, participants, trial). Current research on frontotemporal degeneration, progressive supranuclear palsy, and corticobasal syndrome therapeutics includes 32 trials (37% on biologics) Tau therapeutics are being tested in Alzheimer's disease; primary tauopathies have a large unmet need.
Collapse
Affiliation(s)
- Howard H. Feldman
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jeffrey L. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada at Las VegasLas VegasNevadaUSA
| | - Adam L. Boxer
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Adam M. Staffaroni
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | | | - Paul R. Territo
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Steven E. Arnold
- Department of NeurologyHarvard Medical SchoolMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Clive Ballard
- College of Medicine and HealthUniversity of ExeterExeterUK
| | | | | | - Penny A. Dacks
- The Association for Frontotemporal DegenerationKing of PrussiaPennsylvaniaUSA
| | | | | | - Brian Fiske
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | | | | | | | | | - Eric McDade
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Jose‐Alberto Palma
- Novartis Institutes for Biomedical ResearchCambridgeMassachusettsUSA
- Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | | | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jonathan D. Rohrer
- Department of Neurodegenerative DiseaseDementia Research CentreQueen Square Institute of NeurologyUniversity College of LondonLondonUK
| | - Howard J. Rosen
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Doo Yeon Kim
- Department of NeurologyGenetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Rudolph E. Tanzi
- Department of NeurologyGenetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistrySahlgrenska Academy at the University of GothenburgMölndalSweden
| | | | - Patrick C. May
- ADvantage Neuroscience Consulting LLCFort WayneIndianaUSA
| | - Amy Rommel
- Rainwater Charitable FoundationFort WorthTexasUSA
| |
Collapse
|
25
|
Sandoval C, Lee J, Toth B, Nagaraj R, Schauer SP, Hoffman J, Calderon E, Kollmorgen G, Sanabria Bohórquez SM, Monteiro C, Teng E, Hanson JE, Yeh FL, Gutierrez J, Biever A. CSF complement proteins are elevated in prodromal to moderate Alzheimer's disease patients and are not altered by the anti-tau antibody semorinemab. Alzheimers Dement 2024; 20:7940-7953. [PMID: 39369294 PMCID: PMC11567840 DOI: 10.1002/alz.14271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/14/2024] [Accepted: 08/27/2024] [Indexed: 10/07/2024]
Abstract
INTRODUCTION Growing evidence suggests a role for neuroinflammation in Alzheimer's disease (AD). We investigated complement pathway activity in AD patient cerebrospinal fluid (CSF) and evaluated its modulation by the anti-tau antibody semorinemab. METHODS Immunoassays were applied to measure CSF complement proteins C4, factor B (FB), C3 and their cleavage fragments C4a, C3a, and factor Bb (Bb) in AD patients and a separate cognitively unimpaired (CU) cohort. RESULTS All measured CSF complement proteins were increased in AD versus CU subjects, with C4a displaying the most robust increase. Finally, semorinemab did not have a significant pharmacodynamic effect on CSF complement proteins. DISCUSSION Elevated levels of CSF C4a, C4, C3a, C3, Bb, and FB are consistent with complement activation in AD brains. Despite showing a reduction in CSF soluble tau species, semorinemab did not impact complement protein levels or activity. Further studies are needed to determine the value of complement proteins as neuroinflammation biomarkers in AD. HIGHLIGHTS Cerebrospinal fluid (CSF) complement proteins C4a, C3a, Bb, C4, C3, and factor B levels were increased in Alzheimer's disease (AD) patients compared to a separate cognitively unimpaired (CU) cohort. Baseline CSF complement protein levels were correlated with neuro-axonal degeneration and glial activation biomarkers in AD patients. The investigational anti-tau antibody semorinemab did not impact CSF complement protein levels or activity relative to the placebo arm.
Collapse
Affiliation(s)
- Cosme Sandoval
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Julie Lee
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Balazs Toth
- Department of Data and Statistical SciencesGenentech IncImmunology, Infectious Diseases and OpthalmologySouth San FranciscoCaliforniaUSA
| | - Rajini Nagaraj
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Stephen P. Schauer
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Jennifer Hoffman
- Department of Translational MedicineGenentech IncOperations Diagnostics Technology InnovationSouth San FranciscoCaliforniaUSA
| | - Emilia Calderon
- Department of Translational MedicineGenentech IncOperations Diagnostics Technology InnovationSouth San FranciscoCaliforniaUSA
| | | | | | - Cecilia Monteiro
- Department of Early Clinical DevelopmentGenentech IncSouth San FranciscoCaliforniaUSA
| | - Edmond Teng
- Department of Early Clinical DevelopmentGenentech IncSouth San FranciscoCaliforniaUSA
| | - Jesse E. Hanson
- Department of NeuroscienceGenentech IncSouth San FranciscoCaliforniaUSA
| | - Felix L. Yeh
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Johnny Gutierrez
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Anne Biever
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| |
Collapse
|
26
|
Zhou F, Zhao Y, Sun Y, Chen W. Molecular Insights into Tau Pathology and its Therapeutic Strategies in Alzheimer's Disease. J Integr Neurosci 2024; 23:197. [PMID: 39613463 DOI: 10.31083/j.jin2311197] [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: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 12/01/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The two major hallmarks of this disease are extracellular amyloid plaques and intracellular neurofibrillary tangles in the brain, accompanied by loss of neurons and synapses. The plaques and tangles mainly consist of amyloid-β (Aβ) and tau protein, respectively. Most of the therapeutic strategies for AD to date have focused on Aβ. However, there is still no effective therapy available. In recent years, the clinical therapeutic failure of targeting Aβ pathology has resulted in increased interest towards tau-based therapeutics. In the current review, we focus on the research progress regarding the pathological mechanisms of tau protein in this disease and discuss tau-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Futao Zhou
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yushi Zhao
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yangyan Sun
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Wanjiao Chen
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| |
Collapse
|
27
|
Koper MJ, Moonen S, Ronisz A, Ospitalieri S, Callaerts-Vegh Z, T'Syen D, Rabe S, Staufenbiel M, De Strooper B, Balusu S, Thal DR. Inhibition of an Alzheimer's disease-associated form of necroptosis rescues neuronal death in mouse models. Sci Transl Med 2024; 16:eadf5128. [PMID: 39475569 DOI: 10.1126/scitranslmed.adf5128] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/07/2023] [Accepted: 10/10/2024] [Indexed: 05/23/2025]
Abstract
Necroptosis is a regulated form of cell death that has been observed in Alzheimer's disease (AD) along with the classical pathological hallmark lesions of amyloid plaques and Tau neurofibrillary tangles. To understand the neurodegenerative process in AD, we studied the role of necroptosis in mouse models and primary mouse neurons. Using immunohistochemistry, we demonstrated activated necroptosis-related proteins in transgenic mice developing Tau pathology and in primary neurons from amyloid precursor protein (APP)-Tau double transgenic mice treated with phosphorylated Tau seeds derived from a patient with AD but not in APP transgenic mice that only exhibited β-amyloid deposits. Necroptosis proteins in granulovacuolar degeneration (GVD) bodies were associated with neuronal loss in mouse brain regions also known to be vulnerable to GVD in the human AD brain. Necroptosis inhibitors lowered the percentage of neurons showing GVD and reduced neuronal loss, both in transgenic mice and in primary mouse neurons. This suggests that a GVD-associated form of necroptosis that we refer to as "GVD-necroptosis" may represent a delayed form of necroptosis in AD. We propose that inhibition of necroptosis could rescue this type of neuronal death in AD.
Collapse
Affiliation(s)
- Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Sebastiaan Moonen
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Alicja Ronisz
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Simona Ospitalieri
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
| | | | - Dries T'Syen
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Sabine Rabe
- Novartis Institutes for Biomedical Sciences, Basel, Switzerland
| | - Matthias Staufenbiel
- Novartis Institutes for Biomedical Sciences, Basel, Switzerland
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
- UK Dementia Research Institute, University College London, London, UK
| | - Sriram Balusu
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
29
|
Pascoal TA, Aguzzoli CS, Lussier FZ, Crivelli L, Suemoto CK, Fortea J, Rosa-Neto P, Zimmer ER, Ferreira PCL, Bellaver B. Insights into the use of biomarkers in clinical trials in Alzheimer's disease. EBioMedicine 2024; 108:105322. [PMID: 39366844 DOI: 10.1016/j.ebiom.2024.105322] [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: 02/26/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
Biomarkers have been instrumental in population selection and disease monitoring in clinical trials of recently FDA-approved drugs targeting amyloid-β to slow the progression of Alzheimer's disease (AD). As new therapeutic strategies and biomarker techniques emerge, the importance of biomarkers in drug development is growing exponentially. In this emerging landscape, biomarkers are expected to serve a wide range of contexts of use in clinical trials focusing on AD and related dementias. The joint FDA-NIH BEST (Biomarkers, EndpointS, and other Tools) framework provides standardised terminology to facilitate communication among stakeholders in this increasingly complex field. This review explores various applications of biomarkers relevant to AD clinical trials, using the BEST resource as a reference. For simplicity, we predominantly provide contextual characterizations of biomarkers use from the perspective of drugs targeting amyloid-β and tau proteins. However, general definitions and concepts can be extrapolated to other targets.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | | | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lucía Crivelli
- Department of Cognitive Neurology, Fleni, Buenos Aires, Argentina
| | - Claudia K Suemoto
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Eduardo R Zimmer
- Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil; Graduate Program in Biological Sciences, Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Pamela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
30
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
31
|
Sheng L, Bhalla R. Biomarkers and Target-Specific Small-Molecule Drugs in Alzheimer's Diagnostic and Therapeutic Research: From Amyloidosis to Tauopathy. Neurochem Res 2024; 49:2273-2302. [PMID: 38844706 PMCID: PMC11310295 DOI: 10.1007/s11064-024-04178-w] [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: 01/16/2024] [Revised: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 08/09/2024]
Abstract
Alzheimer's disease (AD) is the most common type of human dementia and is responsible for over 60% of diagnosed dementia cases worldwide. Abnormal deposition of β-amyloid and the accumulation of neurofibrillary tangles have been recognised as the two pathological hallmarks targeted by AD diagnostic imaging as well as therapeutics. With the progression of pathological studies, the two hallmarks and their related pathways have remained the focus of researchers who seek for AD diagnostic and therapeutic strategies in the past decades. In this work, we reviewed the development of the AD biomarkers and their corresponding target-specific small molecule drugs for both diagnostic and therapeutic applications, underlining their success, failure, and future possibilities.
Collapse
Affiliation(s)
- Li Sheng
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
32
|
Lansdall CJ, Teng E, Chague J, Palanganda R, Delmar P, Smith J, Cummings JL, McDougall F. Care partner-informed meaningful change thresholds for the Clinical Dementia Rating-Sum of Boxes for trials of early Alzheimer's disease. Alzheimers Dement 2024; 20:5889-5900. [PMID: 39015065 PMCID: PMC11497679 DOI: 10.1002/alz.14050] [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: 02/08/2024] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Consensus definitions of meaningful within-patient change (MWPC) on the Clinical Dementia Rating-Sum of Boxes (CDR-SB) are needed. Existing estimates use clinician-rated anchors in clinically diagnosed Alzheimer's disease (AD) populations. Incorporating the care partner perspective offers important insights, and evaluating biomarker-confirmed cohorts aligns estimates with ongoing trials. METHODS Anchor-based analyses were conducted to evaluate MWPC on the CDR-SB in early AD (Tauriel; NCT03289143) using Caregiver Global Impression of Change in memory or daily activities. RESULTS Across time points and anchors, mean CDR-SB changes associated with the "somewhat worse" category ranged from 1.50 to 2.12 in early AD, 1.07 to 2.06 in mild cognitive impairment-AD, and 1.79 to 2.25 in mild AD. DISCUSSION The proposed ranges are appropriate to define meaningful progression on the CDR-SB in similar cohorts and support the interpretation of treatment benefit through MWPC analyses. Thresholds should be calibrated to the context of use; lower/higher thresholds may be applicable in studies of earlier/later disease over shorter/longer durations. HIGHLIGHTS Within-patient CDR-SB change thresholds are provided using caregiver-rated anchors. 1.5 to 2.5 points may be an appropriate range in early AD trials of similar durations. Cumulative distribution function plots illustrate the benefit of a given treatment. When selecting thresholds, the target population and study design should be considered.
Collapse
Affiliation(s)
- Claire J. Lansdall
- Product Development, Patient‐Centered Outcomes ResearchF. Hoffmann‐La Roche LtdBaselSwitzerland
| | - Edmond Teng
- Early Clinical DevelopmentGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Jerome Chague
- Product Development, Data ScienceF. Hoffmann‐La Roche LtdBaselSwitzerland
| | - Rohan Palanganda
- Product Development, Data ScienceRoche Products LtdWelwyn Garden CityUK
| | - Paul Delmar
- Product Development, Data ScienceF. Hoffmann‐La Roche LtdBaselSwitzerland
| | - Janice Smith
- Product Development, NeuroscienceRoche Products LtdWelwyn Garden CityUK
| | - Jeffrey L. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceUniversity of NevadaLas VegasNevadaUSA
| | - Fiona McDougall
- Product Development, Patient‐Centered Outcomes ResearchGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
33
|
Iwatsubo T. Development of disease-modifying therapies against Alzheimer's disease. Psychiatry Clin Neurosci 2024; 78:491-494. [PMID: 38842037 PMCID: PMC11488598 DOI: 10.1111/pcn.13681] [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: 01/04/2024] [Revised: 04/04/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024]
Abstract
To successfully develop disease-modifying therapies (DMT) against Alzheimer's disease (AD), it is important to target the mild stage of the disease, before the pathological changes progress and dementia symptoms are fully manifested. To this end, the AD Neuroimaging Initiative (ADNI), a large-scale observational study, was initiated in the U.S. with the goal of development of DMT that are effective in the early stages of mild cognitive impairment (MCI) by utilizing imaging and biomarkers. In Japan, J-ADNI enrolled and followed up 537 patients, mainly with MCI, and established a platform for evaluation including amyloid PET, and demonstrated a high similarity in the clinical course of amyloid-positive MCI (prodromal AD) in Japan and the U.S. In 2023, the anti-Aβ antibody lecanemab successfully completed a Phase III clinical trial for early AD (prodromal AD + mild AD dementia) and was granted regulatory approval and made available both in the US and Japan. Also, phase III trial of donanemab was completed successful. The J-TRC study was initiated in Japan as a "trial ready cohort (TRC)" consisting of participants who met the eligibility criteria for participation in preclinical and prodromal AD trials. Based on such a platform, the development of DMT for AD will progress more rapidly in the future.
Collapse
Affiliation(s)
- Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of MedicineThe University of TokyoBunkyo‐kuJapan
- National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| |
Collapse
|
34
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [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: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
35
|
Bougea A, Gourzis P. Biomarker-Based Precision Therapy for Alzheimer's Disease: Multidimensional Evidence Leading a New Breakthrough in Personalized Medicine. J Clin Med 2024; 13:4661. [PMID: 39200803 PMCID: PMC11355840 DOI: 10.3390/jcm13164661] [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: 06/22/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
(1) Background: Alzheimer's disease (AD) is a worldwide neurodegenerative disorder characterized by the buildup of abnormal proteins in the central nervous system and cognitive decline. Since no radical therapy exists, only symptomatic treatments alleviate symptoms temporarily. In this review, we will explore the latest advancements in precision medicine and biomarkers for AD, including their potential to revolutionize the way we diagnose and treat this devastating condition. (2) Methods: A literature search was performed combining the following Medical Subject Heading (MeSH) terms on PubMed: "Alzheimer's disease", "biomarkers", "APOE", "APP", "GWAS", "cerebrospinal fluid", "polygenic risk score", "Aβ42", "τP-181", " p-tau217", "ptau231", "proteomics", "total tau protein", and "precision medicine" using Boolean operators. (3) Results: Genome-wide association studies (GWAS) have identified numerous genetic variants associated with AD risk, while a transcriptomic analysis has revealed dysregulated gene expression patterns in the brains of individuals with AD. The proteomic and metabolomic profiling of biological fluids, such as blood, urine, and CSF, and neuroimaging biomarkers have also yielded potential biomarkers of AD that could be used for the early diagnosis and monitoring of disease progression. (4) Conclusion: By leveraging a combination of the above biomarkers, novel ultrasensitive immunoassays, mass spectrometry methods, and metabolomics, researchers are making significant strides towards personalized healthcare for individuals with AD.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Philippos Gourzis
- 1st Department of Psychiatry, University of Patras, 26504 Rio, Greece;
| |
Collapse
|
36
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
37
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
38
|
Ye J, Wan H, Chen S, Liu GP. Targeting tau in Alzheimer's disease: from mechanisms to clinical therapy. Neural Regen Res 2024; 19:1489-1498. [PMID: 38051891 PMCID: PMC10883484 DOI: 10.4103/1673-5374.385847] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/16/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Alzheimer's disease is the most prevalent neurodegenerative disease affecting older adults. Primary features of Alzheimer's disease include extracellular aggregation of amyloid-β plaques and the accumulation of neurofibrillary tangles, formed by tau protein, in the cells. While there are amyloid-β-targeting therapies for the treatment of Alzheimer's disease, these therapies are costly and exhibit potential negative side effects. Mounting evidence suggests significant involvement of tau protein in Alzheimer's disease-related neurodegeneration. As an important microtubule-associated protein, tau plays an important role in maintaining the stability of neuronal microtubules and promoting axonal growth. In fact, clinical studies have shown that abnormal phosphorylation of tau protein occurs before accumulation of amyloid-β in the brain. Various therapeutic strategies targeting tau protein have begun to emerge, and are considered possible methods to prevent and treat Alzheimer's disease. Specifically, abnormalities in post-translational modifications of the tau protein, including aberrant phosphorylation, ubiquitination, small ubiquitin-like modifier (SUMO)ylation, acetylation, and truncation, contribute to its microtubule dissociation, misfolding, and subcellular missorting. This causes mitochondrial damage, synaptic impairments, gliosis, and neuroinflammation, eventually leading to neurodegeneration and cognitive deficits. This review summarizes the recent findings on the underlying mechanisms of tau protein in the onset and progression of Alzheimer's disease and discusses tau-targeted treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jinwang Ye
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Huali Wan
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Sihua Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Gong-Ping Liu
- Co-innovation Center of Neurodegeneration, Nantong University, Nantong, Jiangsu Province, China
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 PMCID: PMC11467920 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
40
|
Tondo G, De Marchi F, Bonardi F, Menegon F, Verrini G, Aprile D, Anselmi M, Mazzini L, Comi C. Novel Therapeutic Strategies in Alzheimer's Disease: Pitfalls and Challenges of Anti-Amyloid Therapies and Beyond. J Clin Med 2024; 13:3098. [PMID: 38892809 PMCID: PMC11172489 DOI: 10.3390/jcm13113098] [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: 04/20/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) causes a significant challenge to global healthcare systems, with limited effective treatments available. This review examines the landscape of novel therapeutic strategies for AD, focusing on the shortcomings of traditional therapies against amyloid-beta (Aβ) and exploring emerging alternatives. Despite decades of research emphasizing the role of Aβ accumulation in AD pathogenesis, clinical trials targeting Aβ have obtained disappointing results, highlighting the complexity of AD pathophysiology and the need for investigating other therapeutic approaches. In this manuscript, we first discuss the challenges associated with anti-Aβ therapies, including limited efficacy and potential adverse effects, underscoring the necessity of exploring alternative mechanisms and targets. Thereafter, we review promising non-Aβ-based strategies, such as tau-targeted therapies, neuroinflammation modulation, and gene and stem cell therapy. These approaches offer new avenues for AD treatment by addressing additional pathological hallmarks and downstream effects beyond Aβ deposition.
Collapse
Affiliation(s)
- Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Fabiola De Marchi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Francesca Bonardi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Gaia Verrini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Davide Aprile
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Matteo Anselmi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Letizia Mazzini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, Sant’Andrea Hospital, University of Piemonte Orientale, Corso Abbiate 21, 13100 Vercelli, Italy;
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
41
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
42
|
Penny LK, Lofthouse R, Arastoo M, Porter A, Palliyil S, Harrington CR, Wischik CM. Considerations for biomarker strategies in clinical trials investigating tau-targeting therapeutics for Alzheimer's disease. Transl Neurodegener 2024; 13:25. [PMID: 38773569 PMCID: PMC11107038 DOI: 10.1186/s40035-024-00417-w] [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: 11/08/2023] [Accepted: 04/24/2024] [Indexed: 05/24/2024] Open
Abstract
The use of biomarker-led clinical trial designs has been transformative for investigating amyloid-targeting therapies for Alzheimer's disease (AD). The designs have ensured the correct selection of patients on these trials, supported target engagement and have been used to support claims of disease modification and clinical efficacy. Ultimately, this has recently led to approval of disease-modifying, amyloid-targeting therapies for AD; something that should be noted for clinical trials investigating tau-targeting therapies for AD. There is a clear overlap of the purpose of biomarker use at each stage of clinical development between amyloid-targeting and tau-targeting clinical trials. However, there are differences within the potential context of use and interpretation for some biomarkers in particular measurements of amyloid and utility of soluble, phosphorylated tau biomarkers. Given the complexities of tau in health and disease, it is paramount that therapies target disease-relevant tau and, in parallel, appropriate assays of target engagement are developed. Tau positron emission tomography, fluid biomarkers reflecting tau pathology and downstream measures of neurodegeneration will be important both for participant recruitment and for monitoring disease-modification in tau-targeting clinical trials. Bespoke design of biomarker strategies and interpretations for different modalities and tau-based targets should also be considered.
Collapse
Affiliation(s)
- Lewis K Penny
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Richard Lofthouse
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Mohammad Arastoo
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Andy Porter
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Soumya Palliyil
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- GT Diagnostics (UK) Ltd, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
- GT Diagnostics (UK) Ltd, Aberdeen, UK.
- TauRx Therapeutics Ltd, Aberdeen, UK.
| |
Collapse
|
43
|
Yi LX, Tan EK, Zhou ZD. Passive immunotherapy for Alzheimer's disease: challenges & future directions. J Transl Med 2024; 22:430. [PMID: 38715084 PMCID: PMC11075320 DOI: 10.1186/s12967-024-05248-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Passive immunotherapy with specific antibodies targeting Amyloid β (Aβ) peptide or tubulin-associated unit (tau) protein has emerged as a promising therapeutic approach in Alzheimer's disease (AD). However, in a recent phase III clinical study, Sperling et al. (N Engl J Med 10.1056/NEJMoa2305032, 2023) reported that solanezumab, a monoclonal antibody targeting Aβ peptide, failed to slow cognitive decline in AD patients. Previously, three other anti-Aβ antibodies, bapineuzumab, crenezumab, and gantenerumab, have also failed to show similar beneficial effects. In addition, three humanized antibodies targeting tau protein failed in their phase II trials. However, other anti-Aβ antibodies, such as lecanemab (a humanized mAb binds to soluble Aβ protofibrils), donanemab (a humanized mAb binds to insoluble, N-terminal truncated form of Aβ peptides) and aducanumab (a human mAb binds to the aggregated form of Aβ), have been shown to slow the decline of cognitive functions in early stage AD patients. The specific targets used in passive immunotherapy in these clinical trials may explain the divergent clinical outcomes. There are several challenges and limitations of passive immunotherapy using anti-Aβ antibodies and long term longitudinal studies are needed to assess their efficacy, side effects and cost effectiveness in a wider spectrum of subjects, from pre-dementia to more advanced dementia. A combination therapeutic approach using both anti-Aβ antibodies and other pharmaceutical agents should also be explored.
Collapse
Affiliation(s)
- Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore.
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| | - Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
44
|
Oosthoek M, Vermunt L, de Wilde A, Bongers B, Antwi-Berko D, Scheltens P, van Bokhoven P, Vijverberg EGB, Teunissen CE. Utilization of fluid-based biomarkers as endpoints in disease-modifying clinical trials for Alzheimer's disease: a systematic review. Alzheimers Res Ther 2024; 16:93. [PMID: 38678292 PMCID: PMC11055304 DOI: 10.1186/s13195-024-01456-1] [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: 10/27/2023] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Clinical trials in Alzheimer's disease (AD) had high failure rates for several reasons, including the lack of biological endpoints. Fluid-based biomarkers may present a solution to measure biologically relevant endpoints. It is currently unclear to what extent fluid-based biomarkers are applied to support drug development. METHODS We systematically reviewed 272 trials (clinicaltrials.gov) with disease-modifying therapies starting between 01-01-2017 and 01-01-2024 and identified which CSF and/or blood-based biomarker endpoints were used per purpose and trial type. RESULTS We found that 44% (N = 121) of the trials employed fluid-based biomarker endpoints among which the CSF ATN biomarkers (Aβ (42/40), p/tTau) were used most frequently. In blood, inflammatory cytokines, NFL, and pTau were most frequently employed. Blood- and CSF-based biomarkers were used approximately equally. Target engagement biomarkers were used in 26% (N = 72) of the trials, mainly in drugs targeting inflammation and amyloid. Lack of target engagement markers is most prominent in synaptic plasticity/neuroprotection, neurotransmitter receptor, vasculature, epigenetic regulators, proteostasis and, gut-brain axis targeting drugs. Positive biomarker results did not always translate to cognitive effects, most commonly the small significant reductions in CSF tau isoforms that were seen following anti-Tau treatments. On the other hand, the positive anti-amyloid trials results on cognitive function were supported by clear effect in most fluid markers. CONCLUSIONS As the field moves towards primary prevention, we expect an increase in the use of fluid-based biomarkers to determine disease modification. Use of blood-based biomarkers will rapidly increase, but CSF markers remain important to determine brain-specific treatment effects. With improving techniques, new biomarkers can be found to diversify the possibilities in measuring treatment effects and target engagement. It remains important to interpret biomarker results in the context of the trial and be aware of the performance of the biomarker. Diversifying biomarkers could aid in the development of surrogacy biomarkers for different drug targets.
Collapse
Affiliation(s)
- Marlies Oosthoek
- Department of Laboratory Medicine, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Lisa Vermunt
- Department of Laboratory Medicine, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Arno de Wilde
- EQT Life Sciences, Johannes Vermeersplein 9, 1071 DV, Amsterdam, The Netherlands
| | - Bram Bongers
- Department of Laboratory Medicine, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Daniel Antwi-Berko
- Department of Laboratory Medicine, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Philip Scheltens
- EQT Life Sciences, Johannes Vermeersplein 9, 1071 DV, Amsterdam, The Netherlands
- Alzheimer Center, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | - Everard G B Vijverberg
- Alzheimer Center, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Laboratory Medicine, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
45
|
Costoya-Sánchez A, Moscoso A, Sobrino T, Ruibal Á, Grothe MJ, Schöll M, Silva-Rodríguez J, Aguiar P. Partial volume correction in longitudinal tau PET studies: is it really needed? Neuroimage 2024; 289:120537. [PMID: 38367651 DOI: 10.1016/j.neuroimage.2024.120537] [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: 12/12/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND [18F]flortaucipir (FTP) tau PET quantification is known to be affected by non-specific binding in off-target regions. Although partial volume correction (PVC) techniques partially account for this effect, their inclusion may also introduce noise and variability into the quantification process. While the impact of these effects has been studied in cross-sectional designs, the benefits and drawbacks of PVC on longitudinal FTP studies is still under scrutiny. The aim of this work was to study the performance of the most common PVC techniques for longitudinal FTP imaging. METHODS A cohort of 247 individuals from the Alzheimer's Disease Neuroimaging Initiative with concurrent baseline FTP-PET, amyloid-beta (Aβ) PET and structural MRI, as well as with follow-up FTP-PET and MRI were included in the study. FTP-PET scans were corrected for partial volume effects using Meltzer's, a simple and popular analytical PVC, and both the region-based voxel-wise (RBV) and the iterative Yang (iY) corrections. FTP SUVR values and their longitudinal rates of change were calculated for regions of interest (ROI) corresponding to Braak Areas I-VI, for a temporal meta-ROI and for regions typically displaying off-target FTP binding (caudate, putamen, pallidum, thalamus, choroid plexus, hemispheric white matter, cerebellar white matter, and cerebrospinal fluid). The longitudinal correlation between binding in off-target and target ROIs was analysed for the different PVCs. Additionally, group differences in longitudinal FTP SUVR rates of change between Aβ-negative (A-) and Aβ-positive (A+), and between cognitively unimpaired (CU) and cognitively impaired (CI) individuals, were studied. Finally, we compared the ability of different partial-volume-corrected baseline FTP SUVRs to predict longitudinal brain atrophy and cognitive decline. RESULTS Among off-target ROIs, hemispheric white matter showed the highest correlation with longitudinal FTP SUVR rates from cortical target ROIs (R2=0.28-0.82), with CSF coming in second (R2=0.28-0.42). Application of voxel-wise PVC techniques minimized this correlation, with RBV performing best (R2=0.00-0.07 for hemispheric white matter). PVC also increased group differences between CU and CI individuals in FTP SUVR rates of change across all target regions, with RBV again performing best (No PVC: Cohen's d = 0.26-0.66; RBV: Cohen's d = 0.43-0.74). These improvements were not observed for differentiating A- from A+ groups. Additionally, voxel-wise PVC techniques strengthened the correlation between baseline FTP SUVR and longitudinal grey matter atrophy and cognitive decline. CONCLUSION Quantification of longitudinal FTP SUVR rates of change is affected by signal from off-target regions, especially the hemispheric white matter and the CSF. Voxel-wise PVC techniques significantly reduce this effect. PVC provided a significant but modest benefit for tasks involving the measurement of group-level longitudinal differences. These findings are particularly relevant for the estimations of sample sizes and analysis methodologies of longitudinal group studies.
Collapse
Affiliation(s)
- Alejandro Costoya-Sánchez
- Molecular Imaging Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Av. Barcelona SN, 15782, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Travesía da Choupana s/n, Santiago de Compostela, Spain
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Tomás Sobrino
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Álvaro Ruibal
- Molecular Imaging Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Av. Barcelona SN, 15782, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Travesía da Choupana s/n, Santiago de Compostela, Spain
| | - Michel J Grothe
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; Reina Sofía Alzheimer's Centre, CIEN Foundation, ISCIII, Madrid, 28031, Spain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden; Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Jesús Silva-Rodríguez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; Reina Sofía Alzheimer's Centre, CIEN Foundation, ISCIII, Madrid, 28031, Spain.
| | - Pablo Aguiar
- Molecular Imaging Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Av. Barcelona SN, 15782, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Travesía da Choupana s/n, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
46
|
Williams ZAP, Lang L, Nicolas S, Clarke G, Cryan J, Vauzour D, Nolan YM. Do microbes play a role in Alzheimer's disease? Microb Biotechnol 2024; 17:e14462. [PMID: 38593310 PMCID: PMC11003713 DOI: 10.1111/1751-7915.14462] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024] Open
Abstract
Alzheimer's disease is a complex and progressive condition that affects essential neurological functions such as memory and reasoning. In the brain, neuronal loss, synaptic dysfunction, proteinopathy, neurofibrillary tangles, and neuroinflammation are the hallmarks of Alzheimer's disease pathophysiology. In addition, recent evidence has highlighted that microbes, whether commensal or pathogenic, also have the ability to interact with their host and to regulate its immune system, therefore participating in the exchanges that lead to peripheral inflammation and neuropathology. Because of this intimate relationship, bacteria, viruses, fungi, and protozoa have been implicated in the development of Alzheimer's disease. Here, we bring together current and most recent evidence of the role of microbes in Alzheimer's disease, raising burning questions that need to be addressed to guide therapeutic approaches and potential prophylactic strategies.
Collapse
Affiliation(s)
- Zoë A. P. Williams
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Leonie Lang
- Norwich Medical School, Faculty of Medicine and Health SciencesUniversity of East AngliaNorwichUK
| | - Sarah Nicolas
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry and Neurobehavioural ScienceUniversity College CorkCorkIreland
| | - John Cryan
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - David Vauzour
- Norwich Medical School, Faculty of Medicine and Health SciencesUniversity of East AngliaNorwichUK
| | - Yvonne M. Nolan
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| |
Collapse
|
47
|
Thangwaritorn S, Lee C, Metchikoff E, Razdan V, Ghafary S, Rivera D, Pinto A, Pemminati S. A Review of Recent Advances in the Management of Alzheimer's Disease. Cureus 2024; 16:e58416. [PMID: 38756263 PMCID: PMC11098549 DOI: 10.7759/cureus.58416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative condition and a form of dementia encountered in medical practice. Despite many proposed and attempted treatments, this disease remains a major puzzle in the public health systems worldwide. The initial part of this article provides an overview and illustration of the primary mechanisms responsible for neuronal damage in AD. Subsequently, it offers a critical evaluation of the most noteworthy studies on pharmacological therapy for AD and outlines recent advancements and novel approaches to managing this condition. Main properties, categorization, Food and Drug Administration (FDA) status, mechanisms of action, benefits, and common side effects of the classical and the most recently proposed pharmacological treatments for AD are described. The conventional pharmacological agents revised comprise cholinesterase inhibitors, monoclonal antibodies, and other therapies, such as memantine, valproic acid, and rosiglitazone. The innovative reviewed pharmacological agents comprise the monoclonal antibodies: donanemab, gantenerumab, solanezumab, bapineuzumab, crenezumab, and semorinemab. Nutritional supplements such as alpha-tocopherol (vitamin E) and caprylidene are also revised. Tau and amyloid-targeting treatments include methylthioninium moiety (MT), leuco-methylthioninium bis (LMTM), an oxidized form of MT, and tramiprosate, which inhibits the beta-amyloid (Aβ) monomer aggregation into toxic oligomers. Antidiabetic and anti-neuroinflammation drugs recently proposed for AD treatment are discussed. The antidiabetic drugs include NE3107, an anti-inflammatory and insulin sensitizer, and the diabetes mainstream drug metformin. The anti-neuroinflammatory AD therapies include the use of sodium oligomannate (GV-971), infusions with intravenous immunoglobulin aiming to decrease plasma levels of the constituents of Aβ plaques, and masitinib, a tyrosine kinase inhibitor that impacts mast and microglia cells. Additional anti-inflammatory agents being currently tested in phase-2 clinical trials, such as atomoxetine (selective norepinephrine reuptake inhibitor), losartan (angiotensin 2 receptor agonist), genistein (anti-inflammatory isoflavone neuroprotective agent), trans-resveratrol (polyphenol antioxidant plant estrogen), and benfotiamine (synthetic thiamine precursor), were reviewed. Lastly, drugs targeting Alzheimer's-associated symptoms, such as brexpiprazole (serotonin dopamine activity modulator) and suvorexant (orexin receptor antagonist), respectively, used for agitation and insomnia in AD patients, are reviewed. As experimental investigations and clinical research progress, there is a possibility that a combination of newly tested medications and traditional ones may emerge as a promising treatment option for AD in the future.
Collapse
Affiliation(s)
- Skylynn Thangwaritorn
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Christopher Lee
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Elena Metchikoff
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Vidushi Razdan
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Suliman Ghafary
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Dominic Rivera
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Alvaro Pinto
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Sudhakar Pemminati
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| |
Collapse
|
48
|
Sanabria Bohórquez SM, Baker S, Manser PT, Tonietto M, Galli C, Wildsmith KR, Zou Y, Kerchner GA, Weimer R, Teng E. Evaluation of partial volume correction and analysis of longitudinal [ 18F]GTP1 tau PET imaging in Alzheimer's disease using linear mixed-effects models. FRONTIERS IN NEUROIMAGING 2024; 3:1355402. [PMID: 38606196 PMCID: PMC11008283 DOI: 10.3389/fnimg.2024.1355402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 04/13/2024]
Abstract
Purpose We evaluated the impact of partial volume correction (PVC) methods on the quantification of longitudinal [18F]GTP1 tau positron-emission tomography (PET) in Alzheimer's disease and the suitability of describing the tau pathology burden temporal trajectories using linear mixed-effects models (LMEM). Methods We applied van Cittert iterative deconvolution (VC), 2-compartment, and 3-compartment, and the geometric transfer matrix plus region-based voxelwise methods to data acquired in an Alzheimer's disease natural history study over 18 months at a single imaging site. We determined the optimal PVC method by comparing the standardized uptake value ratio change (%ΔSUVR) between diagnostic and tau burden-level groups and the longitudinal repeatability derived from the LMEM. The performance of LMEM analysis for calculating %ΔSUVR was evaluated in a natural history study and in a multisite clinical trial of semorinemab in prodromal to mild Alzheimer's disease by comparing results to traditional per-visit estimates. Results The VC, 2-compartment, and 3-compartment PVC methods had similar performance, whereas region-based voxelwise overcorrected regions with a higher tau burden. The lowest within-subject variability and acceptable group separation scores were observed without PVC. The LMEM-derived %ΔSUVR values were similar to the per-visit estimates with lower variability. Conclusion The results indicate that the tested PVC methods do not offer a clear advantage or improvement over non-PVC images for the quantification of longitudinal [18F]GTP1 PET data. LMEM offers a robust framework for the longitudinal tau PET quantification with low longitudinal test-retest variability. Clinical trial registration NCT02640092 and NCT03289143.
Collapse
Affiliation(s)
| | - Suzanne Baker
- Clinical Imaging Group, Genentech, Inc., South San Francisco, CA, United States
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Paul T. Manser
- Biostatistics, Genentech, Inc., South San Francisco, CA, United States
| | - Matteo Tonietto
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Christopher Galli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Yixuan Zou
- Data and Statistical Science, Product Development, Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Geoffrey A. Kerchner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Robby Weimer
- Translational Imaging, Genentech, Inc., South San Francisco, CA, United States
| | - Edmond Teng
- Early Clinical Development, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
49
|
Gu X, Qi L, Qi Q, Zhou J, Chen S, Wang L. Monoclonal antibody therapy for Alzheimer's disease focusing on intracerebral targets. Biosci Trends 2024; 18:49-65. [PMID: 38382942 DOI: 10.5582/bst.2023.01288] [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: 02/23/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Due to the complexity of the disorder and the presence of the blood-brain barrier (BBB), its drug discovery and development are facing enormous challenges, especially after several failures of monoclonal antibody (mAb) trials. Nevertheless, the Food and Drug Administration's approval of the mAb aducanumab has ushered in a new day. As we better understand the disease's pathogenesis and identify novel intracerebral therapeutic targets, antibody-based therapies have advanced over the past few years. The mAb drugs targeting β-amyloid or hyperphosphorylated tau protein are the focus of the current research. Massive neuronal loss and glial cell-mediated inflammation are also the vital pathological hallmarks of AD, signaling a new direction for research on mAb drugs. We have elucidated the mechanisms by which AD-specific mAbs cross the BBB to bind to targets. In order to investigate therapeutic approaches to treat AD, this review focuses on the promising mAbs targeting intracerebral dysfunction and related strategies to cross the BBB.
Collapse
Affiliation(s)
- Xiaolei Gu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Long Qi
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Qing Qi
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Song Chen
- Postdoctoral Station of Xiamen University, Fujian, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
50
|
Fleisher AS, Munsie LM, Perahia DGS, Andersen SW, Higgins IA, Hauck PM, Lo AC, Sims JR, Brys M, Mintun M. Assessment of Efficacy and Safety of Zagotenemab: Results From PERISCOPE-ALZ, a Phase 2 Study in Early Symptomatic Alzheimer Disease. Neurology 2024; 102:e208061. [PMID: 38386949 PMCID: PMC11067698 DOI: 10.1212/wnl.0000000000208061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/19/2023] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Zagotenemab (LY3303560), a monoclonal antibody that preferentially targets misfolded, extracellular, aggregated tau, was assessed in the PERISCOPE-ALZ phase 2 study to determine its ability to slow cognitive and functional decline relative to placebo in early symptomatic Alzheimer disease (AD). METHODS Participants were enrolled across 56 sites in North America and Japan. Key eligibility criteria included age of 60-85 years, Mini-Mental State Examination score of 20-28, and intermediate levels of brain tau on PET imaging. In this double-blind study, participants were equally randomized to 1,400 mg or 5,600 mg of zagotenemab, or placebo (IV infusion every 4 weeks for 100 weeks). The primary outcome was change on the Integrated AD Rating Scale (iADRS) assessed by a Bayesian Disease Progression model. Secondary measures include mixed model repeated measures analysis of additional cognitive and functional endpoints as well as biomarkers of AD pathology. RESULTS A total of 360 participants (mean age = 75.4 years; female = 52.8%) were randomized, and 218 completed the treatment period. Demographics and baseline characteristics were reasonably balanced among arms. The mean disease progression ratio (proportional decline in the treated vs placebo group) with 95% credible intervals for the iADRS was 1.10 (0.959-1.265) for the zagotenemab low-dose group and 1.05 (0.907-1.209) for the high-dose, where a ratio less than 1 favors the treatment group. Secondary clinical endpoint measures failed to show a drug-placebo difference in favor of zagotenemab. No treatment effect was demonstrated by flortaucipir PET, volumetric MRI, or neurofilament light chain (NfL) analyses. A dose-related increase in plasma phosphorylated tau181 and total tau was demonstrated. Zagotenemab treatment groups reported a higher incidence of adverse events (AEs) (85.1%) compared with the placebo group (74.6%). This difference was not attributable to any specific AE or category of AEs. DISCUSSION In participants with early symptomatic AD, zagotenemab failed to achieve significant slowing of clinical disease progression compared with placebo. Imaging biomarker and plasma NfL findings did not show evidence of pharmacodynamic activity or disease modification. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov: NCT03518073. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that for patients with early symptomatic AD, zagotenemab does not slow clinical disease progression.
Collapse
Affiliation(s)
- Adam S Fleisher
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Leanne M Munsie
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - David G S Perahia
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Scott W Andersen
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Ixavier A Higgins
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Paula M Hauck
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Albert C Lo
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - John R Sims
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Miroslaw Brys
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| | - Mark Mintun
- From Eli Lilly and Company, Indianapolis, IN. Dr. A.C. Lo is currently at Kisbee Therapeutics, Cambridge, MA
| |
Collapse
|