51
|
Susmitha G, Kumar R. Role of microbial dysbiosis in the pathogenesis of Alzheimer's disease. Neuropharmacology 2023; 229:109478. [PMID: 36871788 DOI: 10.1016/j.neuropharm.2023.109478] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly and detected during the advanced stages where the chances of reversal are minimum. The gut-brain axis mediates a bidirectional communication between the gut and brain, which is dependent on bacterial products such as short chain fatty acids (SCFA) and neurotransmitters. Accumulating lines of evidence suggests that AD is associated with significant alteration in the composition of gut microbiota. Furthermore, transfer of gut microbiota from healthy individuals to patients can reshape the gut microbiota structure and thus holds the potential to be exploited for the treatment of various neurodegenerative disease. Moreover, AD-associated gut dysbiosis can be partially reversed by using probiotics, prebiotics, natural compounds and dietary modifications, but need further validations. Reversal of AD associated gut dysbiosis alleviate AD-associated pathological feature and therefore can be explored as a therapeutic approach in the future. The current review article will describe various studies suggesting that AD dysbiosis occurs with AD and highlights the causal role by focussing on the interventions that hold the potential to reverse the gut dysbiosis partially.
Collapse
Affiliation(s)
- Gudimetla Susmitha
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India.
| |
Collapse
|
52
|
Hao Y, Dong M, Sun Y, Duan X, Niu W. Effectiveness and safety of monoclonal antibodies against amyloid-beta vis-à-vis placebo in mild or moderate Alzheimer's disease. Front Neurol 2023; 14:1147757. [PMID: 37006475 PMCID: PMC10050585 DOI: 10.3389/fneur.2023.1147757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/16/2023] [Indexed: 03/17/2023] Open
Abstract
Backgrounds and objectives Currently, no consensus has been reached on the therapeutic implications of monoclonal antibodies against amyloid-beta (Aβ) in Alzheimer's disease (AD). This study aimed to examine the effectiveness and safety of monoclonal antibodies against Aβ as a whole and also to determine the superiority of individual antibodies vis-à-vis placebo in mild or moderate AD. Methods Literature retrieval, article selection, and data abstraction were performed independently and in duplicate. Cognition and function were appraised by the Mini-Mental State Examination (MMSE), Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog), Disability Assessment for Dementia (DAD), and Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB). Effect sizes are expressed as standardized mean difference (SMD) with a 95% confidence interval (CI). Results Twenty-nine articles involving 108 drug-specific trials and 21,383 participants were eligible for synthesis. Of the four assessment scales, only CDR-SB was significantly reduced after using monoclonal antibodies against Aβ relative to placebo (SMD: -0.12; 95% CI: -0.2 to -0.03; p = 0.008). Egger's tests indicated a low likelihood of publication bias. At individual levels, bapineuzumab was associated with a significant increase in MMSE (SMD: 0.588; 95% CI: 0.226-0.95) and DAD (SMD: 0.919; 95% CI: 0.105-1.943), and a significant decrease in CDR-SB (SMD: -0.15; 95% CI: -0.282-0.018). Bapineuzumab can increase the significant risk of serious adverse events (OR: 1.281; 95% CI: 1.075-1.525). Conclusion Our findings indicate that monoclonal antibodies against Aβ can effectively improve instrumental activities of daily life in mild or moderate AD. In particular, bapineuzumab can improve cognition and function, as well as activities of daily life, and meanwhile, it triggers serious adverse events.
Collapse
Affiliation(s)
- Ying Hao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Mingrui Dong
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yingtong Sun
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiaohui Duan
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Wenquan Niu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
53
|
Role of Tau in Various Tauopathies, Treatment Approaches, and Emerging Role of Nanotechnology in Neurodegenerative Disorders. Mol Neurobiol 2023; 60:1690-1720. [PMID: 36562884 DOI: 10.1007/s12035-022-03164-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A few protein kinases and phosphatases regulate tau protein phosphorylation and an imbalance in their enzyme activity results in tau hyper-phosphorylation. Aberrant tau phosphorylation causes tau to dissociate from the microtubules and clump together in the cytosol to form neurofibrillary tangles (NFTs), which lead to the progression of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Hence, targeting hyperphosphorylated tau protein is a restorative approach for treating neurodegenerative tauopathies. The cyclin-dependent kinase (Cdk5) and the glycogen synthase kinase (GSK3β) have both been implicated in aberrant tau hyperphosphorylation. The limited transport of drugs through the blood-brain barrier (BBB) for reaching the central nervous system (CNS) thus represents a significant problem in the development of drugs. Drug delivery systems based on nanocarriers help solve this problem. In this review, we discuss the tau protein, regulation of tau phosphorylation and abnormal hyperphosphorylation, drugs in use or under clinical trials, and treatment strategies for tauopathies based on the critical role of tau hyperphosphorylation in the pathogenesis of the disease. Pathology of neurodegenerative disease due to hyperphosphorylation and various therapeutic approaches including nanotechnology for its treatment.
Collapse
|
54
|
Espay AJ, Herrup K, Daly T. Finding the falsification threshold of the toxic proteinopathy hypothesis in neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:143-154. [PMID: 36796939 DOI: 10.1016/b978-0-323-85538-9.00008-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
A biomedical hypothesis is a theoretical assumption amenable to being tested in a randomized clinical trial. The main hypotheses in neurodegenerative disorders are based on the concept that proteins accumulate in an aggregated fashion and trigger toxicity. The toxic proteinopathy hypothesis posits that neurodegeneration is caused by toxicity of aggregated amyloid in Alzheimer's disease (toxic amyloid hypothesis), aggregated α-synuclein in Parkinson's disease (toxic synuclein hypothesis), and aggregated tau in progressive supranuclear palsy (toxic tau hypothesis). To date, we have accumulated 40 negative anti-amyloid randomized clinical, 2 anti-synuclein trials, and 4 anti-tau trials. These results have not prompted a major reconsideration of the toxic proteinopathy hypothesis of causality. Imperfections in trial design and execution (incorrect dosage, insensitive endpoints, too-advanced population) but not in the underlying hypotheses have prevailed as explaining the failures. We review here the evidence suggesting that the threshold of hypothesis falsifiability may be too high and advocate in favor of a minimal set of rules that facilitate the interpretation of negative clinical trials as falsifying the driving hypotheses, in particular if the desirable change in surrogate endpoints has been achieved. We propose four steps to refute a hypothesis in future-negative surrogate-backed trials and argue that for the actual rejection to take place, refutation must be accompanied by the proposal of an alternative hypothesis. The absence of alternative hypotheses may be the single greatest reason why there remains hesitancy in rejecting the toxic proteinopathy hypothesis: in the absence of alternatives, we have no clear guidance as to where to redirect or focus.
Collapse
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States.
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Timothy Daly
- Science Norms Democracy, Sorbonne University, Paris, France
| |
Collapse
|
55
|
Plascencia-Villa G, Perry G. Lessons from antiamyloid-β immunotherapies in Alzheimer's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:267-292. [PMID: 36803816 DOI: 10.1016/b978-0-323-85555-6.00019-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The amyloid hypothesis, that established amyloid-β (Aβ) peptide as the primary cause of Alzheimer's disease (AD) and related dementia, has driven the development of treatments for neurodegeneration for 30 years. During the last decades, more than 200 clinical trials testing more than 30 anti-Aβ immunotherapies have been assessed as potential treatments for AD. A vaccine against Aβ was the first immunotherapy intended to avoid aggregation of Aβ into fibrils and senile plaques, but it dramatically failed. Several other vaccines have been proposed as potential AD treatments, targeting different domains or structural motifs of Aβ aggregates, but without clear clinical benefits or effectiveness. In contrast, anti-Aβ therapeutic antibodies have focused on recognizing and removing Aβ aggregates (oligomers, fibrils, or plaques) by eliciting immune clearance. In 2021, the first anti-Aβ antibody, aducanumab (branded as Aduhelm), received FDA approval under an accelerated approval process. The effectiveness and the overall processes regarding the approval of Aduhelm have been under major criticism and scrutiny, prompting a vote of no confidence by public and private health providers, limiting the coverage only to patients enrolled in clinical trials and not for the general elderly patients. Additionally, another three therapeutic anti-Aβ antibodies are following the same path for potential FDA approval. Here, we present the current status of anti-Aβ immunotherapies under evaluation in preclinical and clinical trials for the treatment of AD and related dementia, with a discussion of the main findings and critical lessons learned from the observations from Phase III, II, and I clinical trials of anti-Aβ vaccines and antibodies.
Collapse
Affiliation(s)
- Germán Plascencia-Villa
- Department of Neurosciences, Developmental and Regenerative Biology, The University of Texas at San Antonio (UTSA), San Antonio, TX, United States
| | - George Perry
- Department of Neurosciences, Developmental and Regenerative Biology, The University of Texas at San Antonio (UTSA), San Antonio, TX, United States.
| |
Collapse
|
56
|
Duque KR, Vizcarra JA, Hill EJ, Espay AJ. Disease-modifying vs symptomatic treatments: Splitting over lumping. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:187-209. [PMID: 36803811 DOI: 10.1016/b978-0-323-85555-6.00020-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Clinical trials of putative disease-modifying therapies in neurodegeneration have obeyed the century-old principle of convergence, or lumping, whereby any feature of a clinicopathologic disease entity is considered relevant to most of those affected. While this convergent approach has resulted in important successes in trials of symptomatic therapies, largely aimed at correcting common neurotransmitter deficiencies (e.g., cholinergic deficiency in Alzheimer's disease or dopaminergic deficiency in Parkinson's disease), it has been consistently futile in trials of neuroprotective or disease-modifying interventions. As individuals affected by the same neurodegenerative disorder do not share the same biological drivers, splitting such disease into small molecular/biological subtypes, to match people to therapies most likely to benefit them, is vital in the pursuit of disease modification. We here discuss three paths toward the splitting needed for future successes in precision medicine: (1) encourage the development of aging cohorts agnostic to phenotype in order to enact a biology-to-phenotype direction of biomarker development and validate divergence biomarkers (present in some, absent in most); (2) demand bioassay-based recruitment of subjects into disease-modifying trials of putative neuroprotective interventions in order to match the right therapies to the right recipients; and (3) evaluate promising epidemiologic leads of presumed pathogenetic potential using Mendelian randomization studies before designing the corresponding clinical trials. The reconfiguration of disease-modifying efforts for patients with neurodegenerative disorders will require a paradigm shift from lumping to splitting and from proteinopathy to proteinopenia.
Collapse
Affiliation(s)
- Kevin R Duque
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Joaquin A Vizcarra
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Emily J Hill
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
57
|
Technical Review of Clinical Outcomes Assessments Across the Continuum of Alzheimer's Disease. Neurol Ther 2023; 12:571-595. [PMID: 36790638 PMCID: PMC10043075 DOI: 10.1007/s40120-023-00443-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
INTRODUCTION Insight into the relationship between concepts that matter to the people affected by Alzheimer's disease (AD) and the clinical outcome assessments (COAs) commonly used in AD clinical studies is limited. Phases 1 and 2 of the What Matters Most (WMM) study series identified and quantitatively confirmed 42 treatment-related outcomes that are important to people affected by AD. METHODS We compared WMM concepts rated as "very important" or higher to items included in COAs used commonly in AD studies. RESULTS Twenty COAs designed to assess signs, symptoms, and impacts across the spectrum of AD were selected for review. Among these 20 COAs, only 5 reflected 12 or more WMM concepts [Integrated Alzheimer's Disease Rating Scale (iADRS), Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory (ADCS-ADL), Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory-Mild Cognitive Impairment (ADCS-ADL-MCI), Alzheimer's Disease Composite Scores (ADCOMS), and Clinical Dementia Rating; Clinical Dementia Rating-Sum of Boxes (CDR/CDR-SB)]. Multiple symptoms and impacts of AD identified as important and meaningful in the WMM studies map only indirectly at best to 7 of the 20 most widely used COAs. CONCLUSION While many frequently used COAs in AD capture some concepts identified as important to AD populations and their care partners, overlap between any single measure and the concepts that matter to people affected by AD is limited. The highest singly matched COA reflects fewer than half (45%) of WMM concepts. Use of multiple COAs expands coverage of meaningful concepts. Future research should explore the content validity of AD COAs planned for AD trials based on further confirmation of the ecological validity of the WMM items. This research should inform development and use of core outcome sets that capture WMM items and selection or development of new companion tools to fully demonstrate clinically meaningful outcomes spanning WMM.
Collapse
|
58
|
Tohumeken S, Deme P, Yoo SW, Gupta S, Rais R, Slusher BS, Haughey NJ. Neuronal deletion of nSMase2 reduces the production of Aβ and directly protects neurons. Neurobiol Dis 2023; 177:105987. [PMID: 36603748 DOI: 10.1016/j.nbd.2023.105987] [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: 07/22/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023] Open
Abstract
Extracellular vesicles (EVs) have been proposed to regulate the deposition of Aβ. Multiple publications have shown that APP, amyloid processing enzymes and Aβ peptides are associated with EVs. However, very little Aβ is associated with EVs compared with the total amount Aβ present in human plasma, CSF, or supernatants from cultured neurons. The involvement of EVs has largely been inferred by pharmacological inhibition or whole body deletion of the sphingomyelin hydrolase neutral sphingomyelinase-2 (nSMase2) that is a key regulator for the biogenesis of at-least one population of EVs. Here we used a Cre-Lox system to selectively delete nSMase2 from pyramidal neurons in APP/PS1 mice (APP/PS1-SMPD3-Nex1) and found a ∼ 70% reduction in Aβ deposition at 6 months of age and ∼ 35% reduction at 12 months of age in both cortex and hippocampus. Brain ceramides were increased in APP/PS1 compared with Wt mice, but were similar to Wt in APP/PS1-SMPD3-Nex1 mice suggesting that elevated brain ceramides in this model involves neuronally expressed nSMase2. Reduced levels of PSD95 and deficits of long-term potentiation in APP/PS1 mice were normalized in APP/PS1-SMPD3-Nex1 mice. In contrast, elevated levels of IL-1β, IL-8 and TNFα in APP/PS1 mice were not normalized in APP/PS1-SMPD3-Nex1 mice compared with APP/PS1 mice. Mechanistic studies showed that the size of liquid ordered membrane microdomains was increased in APP/PS1 mice, as were the amounts of APP and BACE1 localized to these microdomains. Pharmacological inhibition of nSMase2 activity with PDDC reduced the size of the liquid ordered membrane microdomains, reduced the localization of APP with BACE1 and reduced the production of Aβ1-40 and Aβ1-42. Although inhibition of nSMase2 reduced the release and increased the size of EVs, very little Aβ was associated with EVs in all conditions tested. We also found that nSMase2 directly protected neurons from the toxic effects of oligomerized Aβ and preserved neural network connectivity despite considerable Aβ deposition. These data demonstrate that nSMase2 plays a role in the production of Aβ by stabilizing the interaction of APP with BACE1 in liquid ordered membrane microdomains, and directly protects neurons from the toxic effects of Aβ. The effects of inhibiting nSMase2 on EV biogenesis may be independent from effects on Aβ production and neuronal protection.
Collapse
Affiliation(s)
- Sehmus Tohumeken
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Pragney Deme
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Seung Wan Yoo
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Sujasha Gupta
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Rana Rais
- The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America
| | - Barbara S Slusher
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America; The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America; The Johns Hopkins University School of Medicine, Departments of Pharmacology and Molecular Sciences, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Oncology, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Neuroscience, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Medicine, Baltimore, MD, United States of America
| | - Norman J Haughey
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America.
| |
Collapse
|
59
|
Huang P, Zhang LY, Tan YY, Chen SD. Links between COVID-19 and Parkinson's disease/Alzheimer's disease: reciprocal impacts, medical care strategies and underlying mechanisms. Transl Neurodegener 2023; 12:5. [PMID: 36717892 PMCID: PMC9885419 DOI: 10.1186/s40035-023-00337-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The impact of coronavirus disease 2019 (COVID-19) pandemic on patients with neurodegenerative diseases and the specific neurological manifestations of COVID-19 have aroused great interest. However, there are still many issues of concern to be clarified. Therefore, we review the current literature on the complex relationship between COVID-19 and neurodegenerative diseases with an emphasis on Parkinson's disease (PD) and Alzheimer's disease (AD). We summarize the impact of COVID-19 infection on symptom severity, disease progression, and mortality rate of PD and AD, and discuss whether COVID-19 infection could trigger PD and AD. In addition, the susceptibility to and the prognosis of COVID-19 in PD patients and AD patients are also included. In order to achieve better management of PD and AD patients, modifications of care strategies, specific drug therapies, and vaccines during the pandemic are also listed. At last, mechanisms underlying the link of COVID-19 with PD and AD are reviewed.
Collapse
Affiliation(s)
- Pei Huang
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Lin-Yuan Zhang
- grid.412478.c0000 0004 1760 4628Department of Neurology, Shanghai General Hospital, Shanghai, 200080 China
| | - Yu-Yan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
60
|
Johnson SC, Suárez-Calvet M, Suridjan I, Minguillón C, Gispert JD, Jonaitis E, Michna A, Carboni M, Bittner T, Rabe C, Kollmorgen G, Zetterberg H, Blennow K. Identifying clinically useful biomarkers in neurodegenerative disease through a collaborative approach: the NeuroToolKit. Alzheimers Res Ther 2023; 15:25. [PMID: 36709293 PMCID: PMC9883877 DOI: 10.1186/s13195-023-01168-y] [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: 02/25/2022] [Accepted: 01/15/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex and heterogeneous disease, which requires reliable biomarkers for diagnosis and monitoring disease activity. Preanalytical protocol and technical variability associated with biomarker immunoassays makes comparability of biomarker data across multiple cohorts difficult. This study aimed to compare cerebrospinal fluid (CSF) biomarker results across independent cohorts, including participants spanning the AD continuum. METHODS Measured on the NeuroToolKit (NTK) prototype panel of immunoassays, 12 CSF biomarkers were evaluated from three cohorts (ALFA+, Wisconsin, and Abby/Blaze). A correction factor was applied to biomarkers found to be affected by preanalytical procedures (amyloid-β1-42, amyloid-β1-40, and alpha-synuclein), and results between cohorts for each disease stage were compared. The relationship between CSF biomarker concentration and cognitive scores was evaluated. RESULTS Biomarker distributions were comparable across cohorts following correction. Correlations of biomarker values were consistent across cohorts, regardless of disease stage. Disease stage differentiation was highest for neurofilament light (NfL), phosphorylated tau, and total tau, regardless of the cohort. Correlation between biomarker concentration and cognitive scores was comparable across cohorts, and strongest for NfL, chitinase-3-like protein-1 (YKL40), and glial fibrillary acidic protein. DISCUSSION The precision of the NTK enables merging of biomarker datasets, after correction for preanalytical confounders. Assessment of multiple cohorts is crucial to increase power in future studies into AD pathogenesis.
Collapse
Affiliation(s)
- Sterling C Johnson
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center of the William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain.
- Centre for Biomedical Research Network on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
- Neurology Service, Hospital del Mar, Barcelona, Spain.
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.
| | | | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain
- Centre for Biomedical Research Network on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centre for Biomedical Research in Network Bioengineering, Biomaterials and Nanomedicine, Instituto de Salud Carlos III, Madrid, Spain
| | - Erin Jonaitis
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Tobias Bittner
- F. Hoffmann-La Roche AG, Basel, Switzerland
- Genentech, A Member of the Roche Group, San Francisco, CA, USA
| | - Christina Rabe
- Genentech, A Member of the Roche Group, San Francisco, CA, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
61
|
Hillen H. Editorial: Beta-Amyloid oligomer specific treatments for Alzheimer's disease. Front Neurosci 2023; 17:1034158. [PMID: 36761412 PMCID: PMC9905805 DOI: 10.3389/fnins.2023.1034158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
|
62
|
Qiao Y, Chi Y, Zhang Q, Ma Y. Safety and efficacy of lecanemab for Alzheimer's disease: a systematic review and meta-analysis of randomized clinical trials. Front Aging Neurosci 2023; 15:1169499. [PMID: 37213538 PMCID: PMC10196238 DOI: 10.3389/fnagi.2023.1169499] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/07/2023] [Indexed: 05/23/2023] Open
Abstract
Objective We performed a systematic review and meta-analysis of the cognitive effectiveness and safety of lecanemab on subjects with Alzheimer's disease (AD). Methods We screened the literature published before February 2023 in PubMed, Embase, Web of Science, and Cochrane that were searched for randomized controlled trials testing lecanemab for the treatment of cognitive decline in patients with MCI or AD. Outcomes measured were CDR Sum of Boxes (CDR-SB), Alzheimer's Disease Composite Score (ADCOMS), AD Assessment Scale-Cognitive Subscale (ADAS-Cog), Clinical Dementia Rating (CDR), amyloid PET Standardized Uptake Volume Ratio (SUVr), amyloid burden on PET, and risks for adverse events. Results A total of four randomized controlled trials were included, involving 3,108 AD patients (1,695 lecanemab groups and 1,413 placebo groups) to synthesize evidence. Baseline characteristics of the two groups were similar in all outcomes except that ApoE 4 status and higher MMSE score were observed in the lecanemab group. It is reported that lecanemab was beneficial to stabilize or slow down the decrease in CDR-SB (WMD: -0.45; 95% CI: -0.64, -0.25; p < 0.00001), ADCOMS (WMD: -0.05; 95% CI: -0.07, -0.03; p < 0.00001), ADAS-cog (WMD: -1.11; 95% CI: -1.64, -0.57; p < 0.0001), amyloid PET SUVr (WMD: -0.15; 95% CI: -0.48, 0.19; p = 0.38), amyloid burden on PET (WMD:-35.44; 95% CI: -65.22,-5.67; p = 0.02), adverse events (subjects with any TEAE) (OR: 0.73; 95% CI: 0.25, 2.15; p = 0.57), ARIA-E (OR:8.95; 95% CI: 5.36, 14.95; p < 0.00001), and ARIA-H (OR:2.00; 95% CI: 1.53, 2.62; p < 0.00001) in early AD patients. Conclusion Our analysis found that lecanemab showed significant positive statistical efficacy with respect to cognition, function, and behavior in patients with early AD though the actual clinical significance is yet to be established. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier: CRD42023393393.
Collapse
|
63
|
Yu Y, Zhang N, Xiang B, Ding N, Liu J, Huang J, Zhao M, Zhao Y, Wang Y, Ma Z. In vivo characterization of cerebrovascular impairment induced by amyloid β peptide overload in glymphatic clearance system using swept-source optical coherence tomography. NEUROPHOTONICS 2023; 10:015005. [PMID: 36817752 PMCID: PMC9933996 DOI: 10.1117/1.nph.10.1.015005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
SIGNIFICANCE Antiamyloid β ( A β ) immunotherapy is a promising therapeutic strategy for Alzheimer's disease (AD) but generates large amounts of soluble A β peptides that could overwhelm the clearance pathway, leading to serious side effects. Direct implications of A β in glymphatic drainage transport for cerebral vasculature and tissue are not well known. Studies are needed to resolve this issue and pave the way to better monitoring abnormal vascular events that may occur in A β -modifying therapies for AD. AIM The objective is to characterize the modification of cerebral vasculature and tissue induced by soluble A β abundantly present in the glymphatic clearance system. APPROACH A β 1 - 42 peptide was injected intracerebroventricularly and swept-source optical coherence tomography (SS-OCT) was used to monitor the progression of changes in the brain microvascular network and tissue in vivo over 14 days. Parameters reflecting vascular morphology and structure as well as tissue status were quantified and compared before treatment. RESULTS Vascular perfusion density, vessel length, and branch density decreased sharply and persistently following peptide administration. In comparison, vascular average diameter and vascular tortuosity were moderately increased at the late stage of monitoring. Endpoint density gradually increased, and the global optical attenuation coefficient value decreased significantly over time. CONCLUSIONS A β burden in the glymphatic system directly contributes to cerebrovascular structural and morphological abnormalities and global brain tissue damage, suggesting severe deleterious properties of soluble cerebrospinal fluid- A β . We also show that OCT can be used as an effective tool to monitor cerebrovascular dynamics and tissue property changes in response to therapeutic treatments in drug discovery research.
Collapse
Affiliation(s)
- Yao Yu
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao, China
| | - Ning Zhang
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
| | - Ben Xiang
- Northeastern University, College of Information Science and Engineering, Shenyang, China
| | - Ning Ding
- Northeastern University, College of Information Science and Engineering, Shenyang, China
| | - Jian Liu
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao, China
| | - Jiangmei Huang
- First Hospital of Qinhuangdao, Department of Pathology, Qinhuangdao, China
| | - Min Zhao
- First Hospital of Qinhuangdao, Department of Pathology, Qinhuangdao, China
| | - Yuqian Zhao
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
| | - Yi Wang
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao, China
| | - Zhenhe Ma
- Northeastern University at Qinhuangdao, School of Control Engineering, Qinhuangdao, China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao, China
| |
Collapse
|
64
|
Sotoudeh H, Alizadeh M, Shahidi R, Shobeiri P, love N, Singhal A. Subcortical signal alteration of corticospinal tracts. A radiologic manifestation of ARIA: A case report. Radiol Case Rep 2023; 18:275-279. [DOI: 10.1016/j.radcr.2022.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 11/09/2022] Open
|
65
|
Adhikari UK, Khan R, Mikhael M, Balez R, David MA, Mahns D, Hardy J, Tayebi M. Therapeutic anti-amyloid β antibodies cause neuronal disturbances. Alzheimers Dement 2022. [PMID: 36515320 DOI: 10.1002/alz.12833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recent published clinical trial safety data showed that 41% of Alzheimer patients experienced amyloid-related imaging abnormalities (ARIA), marks of microhemorrhages and edema in the brain, following administration of Biogen's Aduhelm/aducanumab (amino acids 3-7 of the Aβ peptide). Similarly, Janssen/Pfizer's Bapineuzumab (amino acids 1-5 of the Aβ peptide) and Roche's Gantenerumab (amino acids 2-11/18-27 of the Aβ peptide) also displayed ARIA in clinical trials, including microhemorrhage and focal areas of inflammation or vasogenic edema, respectively. The molecular mechanisms underlying ARIA caused by therapeutic anti-Aβ antibodies remain largely unknown, however, recent reports demonstrated that therapeutic anti-prion antibodies activate neuronal allergenic proteomes following cross-linking cellular prion protein. METHODS Here, we report that treatment of human induced pluripotent stem cells- derived neurons (HSCN) from a non-demented donor, co-cultured with human primary microglia with anti-Aβ1-6, or anti-Aβ17-23 antibodies activate a significant number of allergenic-related proteins as assessed by mass spectrometry. RESULTS Interestingly, a large proportion of the identified proteins included cytokines such as interleukin (IL)-4, IL-12, and IL-13 suggesting a type-1 hypersensitivity response. Following flow cytometry analysis, several proinflammatory cytokines were significantly elevated following anti-Aβ1-6, or anti-Aβ17-23 antibody treatment. DISCUSSION These results justify further and more robust investigation of the molecular mechanisms of ARIA during immunotherapy study trials of AD. HIGHLIGHTS Allergenic-related proteins are often linked with Alzheimer's disease (AD). We investigated the effects of amyloid beta (Aβ) immunotherapy on stem cell derived neurons and primary neuronal cells co-cultured with microglia. Anti-Aβ antibody treatment of neurons or neurons co-cultured with microglia led to activation of a substantial number of allergenic-related genes. These allergenic-related genes are associated with endothelial dysfunction possibly responsible for ARIA.
Collapse
Affiliation(s)
- Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rizwan Khan
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Meena Mikhael
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | | | - David Mahns
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
66
|
Ganguly D, Thomas JA, Ali A, Kumar R. Mechanistic and therapeutic implications of EphA-4 receptor tyrosine kinase in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5532-5546. [PMID: 34989046 DOI: 10.1111/ejn.15591] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/14/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
Erythropoietin-producing hepatoma (Eph) receptors belong to a family of tyrosine kinase receptors that plays a pivotal role in the development of the brain. Eph can be divided broadly into two groups, namely, EphA and EphB, comprising nine and five members, respectively. In recent years, the role of EphA-4 has become increasingly apparent in the onset of Alzheimer's disease (AD). Emerging evidence suggests that EphA-4 results in synaptic dysfunction, which in turn promotes the progression of AD. Moreover, pharmacological or genetic ablation of EphA-4 in the murine model of AD can alleviate the symptoms. The current review summarizes different pathways by which EphA-4 can influence pathogenesis. Since, majority of the studies had reported the protective effect of EphA-4 inhibition during AD, designing therapeutics based on decreasing its enzymatic activity might be necessary for introducing the novel interventions. Therefore, the review described peptide and nanobodies inhibitors of EphA-4 that exhibit the potential to modulate EphA-4 and could be used as lead molecules for the targeted therapy of AD.
Collapse
Affiliation(s)
- Devargya Ganguly
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Joshua Abby Thomas
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Abid Ali
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| |
Collapse
|
67
|
Bera A, Lavanya G, Reshmi R, Dev K, Kumar R. Mechanistic and therapeutic role of Drp1 in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5516-5531. [PMID: 35078269 DOI: 10.1111/ejn.15611] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, dynamin-related protein 1 (Drp1) was recognized to contribute significantly towards the pathogenesis of AD. Drp1 is primarily located in the cytosol, from where it translocates to the mitochondrial outer membrane and drives the mitochondrial fission via GTP hydrolysis. Drp1 interacts with Aβ and phosphorylated tau, leading to excessive mitochondrial fragmentation, which in turn results in synaptic dysfunction, neuronal damage and cognitive decline. Several studies suggest an increase in the level of Drp1 in the post-mortem brain specimen collected from the AD patients and murine models of AD. Interestingly, heterozygous deletion of Drp1 in the transgenic murine model of AD ameliorates the mitochondrial dysfunction, improving learning and memory. The current review article discusses the possible mechanistic pathways by which Drp1 can influence the pathogenesis of AD. Besides, it will describe various inhibitors for Drp1 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Arpita Bera
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Gantyada Lavanya
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Ravada Reshmi
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| |
Collapse
|
68
|
Kumar R, Tiwari V, Dey S. Role of proline-rich tyrosine kinase 2 (Pyk2) in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5442-5452. [PMID: 34905657 DOI: 10.1111/ejn.15569] [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: 07/05/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, proline-rich tyrosine kinase 2 (Pyk2), which belongs to the focal adhesion kinase (FAK) non-receptor tyrosine kinase family, was recognized to contribute significantly towards the pathogenesis of AD. Pyk2 can influence the formation of amyloid plaques as well as NFTs. The kinase can directly phosphorylate tau, which is a significant component of NFTs and enhances tau pathology. Several competitive inhibitors have been developed for Pyk2, tested in several cancer models, as Pyk2 is known to be overexpressed under those conditions. The current review article discusses the possible mechanistic pathways by which Pyk2 can influence the pathogenesis of AD. Besides, it describes various inhibitors for Pyk2 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM University, Visakhapatnam, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
69
|
Ostrowitzki S, Bittner T, Sink KM, Mackey H, Rabe C, Honig LS, Cassetta E, Woodward M, Boada M, van Dyck CH, Grimmer T, Selkoe DJ, Schneider A, Blondeau K, Hu N, Quartino A, Clayton D, Dolton M, Dang Y, Ostaszewski B, Sanabria-Bohórquez SM, Rabbia M, Toth B, Eichenlaub U, Smith J, Honigberg LA, Doody RS. Evaluating the Safety and Efficacy of Crenezumab vs Placebo in Adults With Early Alzheimer Disease: Two Phase 3 Randomized Placebo-Controlled Trials. JAMA Neurol 2022; 79:1113-1121. [PMID: 36121669 PMCID: PMC9486635 DOI: 10.1001/jamaneurol.2022.2909] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/08/2022] [Indexed: 11/14/2022]
Abstract
Importance Alzheimer disease (AD), a neurodegenerative disease characterized by β-amyloid plaques and τ tangles in the brain, represents an unmet medical need with no fully approved therapeutics to modify disease progression. Objective To investigate the safety and efficacy of crenezumab, a humanized monoclonal immunoglobulin G4 antibody targeting β-amyloid oligomers, in participants with prodromal to mild (early) AD. Design, Setting, and Participants Two phase 3 multicenter randomized double-blind placebo-controlled parallel-group efficacy and safety studies of crenezumab in participants with early AD, CREAD and CREAD2, were initiated in 2016 and 2017, respectively, and were designed to evaluate the efficacy and safety of crenezumab in participants with early AD. CREAD (194 sites in 30 countries) and CREAD2 (209 sites in 27 countries) were global multicenter studies. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. Both trials enrolled individuals aged 50 to 85 years with early AD. Participants with some comorbidities and evidence of cerebral infarction or more than 4 microbleeds or areas of leptomeningeal hemosiderosis on magnetic resonance imaging were excluded. After 2923 and 2858 were excluded, respectively, 813 participants in CREAD and 806 in CREAD2 were randomly assigned in a 1:1 ratio to either placebo or crenezumab. In the final analysis, there were 409 participants in the placebo group and 404 in the crenezumab group in CREAD and 399 in the placebo group and 407 in the crenezumab group in CREAD2. Data were analyzed up until January 2019 and August 2019, respectively. Interventions Participants received placebo or 60 mg/kg crenezumab intravenously every 4 weeks for up to 100 weeks. Main Outcomes and Measures The primary outcome was change from baseline to week 105 in Clinical Dementia Rating-Sum of Boxes (CDR-SB) score. Results There were 813 participants in CREAD (mean [SD] age, 70.7 [8.2] years; 483 female and 330 male) and 806 in CREAD2 (mean [SD] age, 70.9 [7.7] years; 456 female and 350 male). Baseline characteristics were balanced between both groups. The between-group difference in mean change from baseline in CDR-SB score (placebo minus crenezumab) was -0.17 (95% CI, -0.86 to 0.53; P = .63) at week 105 in the CREAD study (88 placebo; 86 crenezumab). Compared with previous trials, no new safety signals were identified, and amyloid-related imaging abnormalities with edema were rare, mild, and transient. No meaningful changes in AD biomarkers were observed. Both studies were discontinued following a preplanned interim analysis indicating that CREAD was unlikely to meet the primary end point. Conclusions and Relevance Crenezumab was well tolerated but did not reduce clinical decline in participants with early AD. Trial Registration ClinicalTrials.gov Identifiers: CREAD, NCT02670083; CREAD2, NCT03114657.
Collapse
Affiliation(s)
| | - Tobias Bittner
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Genentech, Inc, South San Francisco, California
| | | | | | | | - Lawrence S. Honig
- Taub Institute and Department of Neurology, Columbia University Irving Medical Center, New York, New York
| | - Emanuele Cassetta
- Fatebenefratelli Foundation, Associazione Fatebenefratelli Per la Ricerca Division, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Michael Woodward
- Austin Health Continuing Care Clinical Service Unit, Heidelberg, Germany
- University of Melbourne, Melbourne, Victoria, Australia
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Dennis J. Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | - Nan Hu
- Genentech, Inc, South San Francisco, California
| | - Angelica Quartino
- Genentech, Inc, South San Francisco, California
- Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Gothenburg, Sweden
| | | | - Michael Dolton
- Roche Products Australia Pty Ltd, Sydney, New South Wales, Australia
| | - Yifan Dang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Sanofi Genzyme, Waltham, Massachusetts
| | - Beth Ostaszewski
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | - Balazs Toth
- Genentech, Inc, South San Francisco, California
| | | | - Jillian Smith
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | - Rachelle S. Doody
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Genentech, Inc, South San Francisco, California
| |
Collapse
|
70
|
Li P, Lämmerhofer M. Generation of 13C-Labeled Inositol and Inositol Phosphates by Stable Isotope Labeling Cell Culture for Quantitative Metabolomics. Anal Chem 2022; 94:15332-15340. [DOI: 10.1021/acs.analchem.2c02819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Peng Li
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of Tübingen, 72076Tübingen, Germany
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of Tübingen, 72076Tübingen, Germany
| |
Collapse
|
71
|
Zhao P, Zhang N, An Z. Engineering antibody and protein therapeutics to cross the blood-brain barrier. Antib Ther 2022; 5:311-331. [PMID: 36540309 PMCID: PMC9759110 DOI: 10.1093/abt/tbac028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Diseases in the central nervous system (CNS) are often difficult to treat. Antibody- and protein-based therapeutics hold huge promises in CNS disease treatment. However, proteins are restricted from entering the CNS by the blood-brain barrier (BBB). To achieve enhanced BBB crossing, antibody-based carriers have been developed by utilizing the endogenous macromolecule transportation pathway, known as receptor-mediated transcytosis. In this report, we first provided an overall review on key CNS diseases and the most promising antibody- or protein-based therapeutics approved or in clinical trials. We then reviewed the platforms that are being explored to increase the macromolecule brain entry to combat CNS diseases. Finally, we have analyzed the lessons learned from past experiences and have provided a perspective on the future engineering of novel delivery vehicles for antibody- and protein-based therapies for CNS diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| |
Collapse
|
72
|
Zieneldien T, Kim J, Sawmiller D, Cao C. The Immune System as a Therapeutic Target for Alzheimer’s Disease. Life (Basel) 2022; 12:life12091440. [PMID: 36143476 PMCID: PMC9506058 DOI: 10.3390/life12091440] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a heterogeneous neurodegenerative disorder and is the most common cause of dementia. Furthermore, aging is considered the most critical risk factor for AD. However, despite the vast amount of research and resources allocated to the understanding and development of AD treatments, setbacks have been more prominent than successes. Recent studies have shown that there is an intricate connection between the immune and central nervous systems, which can be imbalanced and thereby mediate neuroinflammation and AD. Thus, this review examines this connection and how it can be altered with AD. Recent developments in active and passive immunotherapy for AD are also discussed as well as suggestions for improving these therapies moving forward.
Collapse
Affiliation(s)
- Tarek Zieneldien
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Janice Kim
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Darrell Sawmiller
- MegaNano BioTech, Inc., 3802 Spectrum Blvd. Suite 122, Tampa, FL 33612, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- USF-Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33613, USA
- Correspondence:
| |
Collapse
|
73
|
Valiukas Z, Ephraim R, Tangalakis K, Davidson M, Apostolopoulos V, Feehan J. Immunotherapies for Alzheimer’s Disease—A Review. Vaccines (Basel) 2022; 10:vaccines10091527. [PMID: 36146605 PMCID: PMC9503401 DOI: 10.3390/vaccines10091527] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disorder that falls under the umbrella of dementia and is characterised by the presence of highly neurotoxic amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs) of tau protein within the brain. Historically, treatments for AD have consisted of medications that can slow the progression of symptoms but not halt or reverse them. The shortcomings of conventional drugs have led to a growing need for novel, effective approaches to the treatment of AD. In recent years, immunotherapies have been at the forefront of these efforts. Briefly, immunotherapies utilise the immune system of the patient to treat a condition, with common immunotherapies for AD consisting of the use of monoclonal antibodies or vaccines. Most of these treatments target the production and deposition of Aβ due to its neurotoxicity, but treatments specifically targeting tau protein are being researched as well. These treatments have had great variance in their efficacy and safety, leading to a constant need for the research and development of new safe and effective treatments.
Collapse
Affiliation(s)
- Zachary Valiukas
- College of Health and Biomedicine, Victoria University, Melbourne, VIC 3011, Australia
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
| | - Kathy Tangalakis
- First Year College, Victoria University, Melbourne, VIC 3011, Australia
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC 3011, Australia
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
- Correspondence:
| |
Collapse
|
74
|
Yang H, Li J, Li X, Ma L, Hou M, Zhou H, Zhou R. Based on molecular structures: Amyloid-β generation, clearance, toxicity and therapeutic strategies. Front Mol Neurosci 2022; 15:927530. [PMID: 36117918 PMCID: PMC9470852 DOI: 10.3389/fnmol.2022.927530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Amyloid-β (Aβ) has long been considered as one of the most important pathogenic factors in Alzheimer’s disease (AD), but the specific pathogenic mechanism of Aβ is still not completely understood. In recent years, the development of structural biology technology has led to new understandings about Aβ molecular structures, Aβ generation and clearance from the brain and peripheral tissues, and its pathological toxicity. The purpose of the review is to discuss Aβ metabolism and toxicity, and the therapeutic strategy of AD based on the latest progress in molecular structures of Aβ. The Aβ structure at the atomic level has been analyzed, which provides a new and refined perspective to comprehend the role of Aβ in AD and to formulate therapeutic strategies of AD.
Collapse
Affiliation(s)
- Hai Yang
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Jinping Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaoxiong Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Linqiu Ma
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingliang Hou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huadong Zhou
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Rui Zhou
- Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Rui Zhou,
| |
Collapse
|
75
|
Khoury R, Gallop A, Roberts K, Grysman N, Lu J, Grossberg GT. Pharmacotherapy for Alzheimer’s disease: what’s new on the horizon? Expert Opin Pharmacother 2022; 23:1305-1323. [DOI: 10.1080/14656566.2022.2097868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rita Khoury
- Department of Psychiatry and Clinical Psychology, St. Georges Hospital University Medical Center, Beirut, Lebanon
- University of Balamand, Faculty of Medicine, Beirut, Lebanon
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Amy Gallop
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Kelsey Roberts
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Noam Grysman
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - Jiaxi Lu
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| | - George T. Grossberg
- Department of Psychiatry and Behavioral Neuroscience, St Louis University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
76
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
77
|
Xu YJ, Au NPB, Ma CHE. Functional and Phenotypic Diversity of Microglia: Implication for Microglia-Based Therapies for Alzheimer’s Disease. Front Aging Neurosci 2022; 14:896852. [PMID: 35693341 PMCID: PMC9178186 DOI: 10.3389/fnagi.2022.896852] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/05/2022] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease and is closely associated with the accumulation of β-amyloid (Aβ) and neurofibrillary tangles (NFTs). Apart from Aβ and NFT pathologies, AD patients also exhibit a widespread microglial activation in various brain regions with elevated production of pro-inflammatory cytokines, a phenomenon known as neuroinflammation. In healthy central nervous system, microglia adopt ramified, “surveying” phenotype with compact cell bodies and elongated processes. In AD, the presence of pathogenic proteins such as extracellular Aβ plaques and hyperphosphorylated tau, induce the transformation of ramified microglia into amoeboid microglia. Ameboid microglia are highly phagocytic immune cells and actively secrete a cascade of pro-inflammatory cytokines and chemokines. However, the phagocytic ability of microglia gradually declines with age, and thus the clearance of pathogenic proteins becomes highly ineffective, leading to the accumulation of Aβ plaques and hyperphosphorylated tau in the aging brain. The accumulation of pathogenic proteins further augments the neuroinflammatory responses and sustains the activation of microglia. The excessive production of pro-inflammatory cytokines induces a massive loss of functional synapses and neurons, further worsening the disease condition of AD. More recently, the identification of a subset of microglia by transcriptomic studies, namely disease-associated microglia (DAM), the progressive transition from homeostatic microglia to DAM is TREM2-dependent and the homeostatic microglia gradually acquire the state of DAM during the disease progression of AD. Recent in-depth transcriptomic analysis identifies ApoE and Trem2 from microglia as the major risk factors for AD pathogenesis. In this review, we summarize current understandings of the functional roles of age-dependent microglial activation and neuroinflammation in the pathogenesis of AD. To this end, the exponential growth in transcriptomic data provides a solid foundation for in silico drug screening and gains further insight into the development of microglia-based therapeutic interventions for AD.
Collapse
Affiliation(s)
- Yi-Jun Xu
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ngan Pan Bennett Au
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- *Correspondence: Chi Him Eddie Ma,
| |
Collapse
|
78
|
Brucki SMD, César-Freitas KG, Spera RR, Borges CR, Smid J. Are we ready to use anti-amyloid therapy in Alzheimer's disease? ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:15-23. [PMID: 35976307 PMCID: PMC9491439 DOI: 10.1590/0004-282x-anp-2022-s117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/29/2022] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Biomarkers have demonstrated that AD pathology exists over the disease continuum from a stage preceding symptoms over 15-25 years to the progressively more impaired symptomatic states, mild cognitive impairment (MCI), and dementia. Biomarkers include: amyloid (Aß), phosphorylated tau, and neurodegeneration. The plasma assays for Aß and tau show great promise for clinical and research use. This review has aimed not only to present the ATN diagnostic classification and the preclinical AD concepts in addressing some possibilities of cognitive assessment instruments, but also to briefly summarize the main anti-amyloid monoclonal antibodies studied in clinical trials. In addition, this paper presents a critical analysis by experts in cognitive neurology while addressing the question as to whether we are prepared for the anti-amyloid therapy era or not.
Collapse
Affiliation(s)
- Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Departamento de Neurologia, Unidade de Neurologia Cognitiva e Comportamental, São Paulo SP, Brazil
| | - Karolina Gouveia César-Freitas
- Universidade de São Paulo, Departamento de Neurologia, Unidade de Neurologia Cognitiva e Comportamental, São Paulo SP, Brazil
| | - Raphael Ribeiro Spera
- Universidade de São Paulo, Departamento de Neurologia, Unidade de Neurologia Cognitiva e Comportamental, São Paulo SP, Brazil
| | - Conrado Regis Borges
- Universidade de São Paulo, Departamento de Neurologia, Unidade de Neurologia Cognitiva e Comportamental, São Paulo SP, Brazil
| | - Jerusa Smid
- Universidade de São Paulo, Departamento de Neurologia, Unidade de Neurologia Cognitiva e Comportamental, São Paulo SP, Brazil
- Hospital Israelita Albert Einstein, Grupo Médico-Assistencial de Memória e Cognição, São Paulo SP, Brazil
- Instituto de Infectologia Emílio Ribas, São Paulo SP, Brazil
| |
Collapse
|
79
|
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disorder that is characterized clinically by progressive cognitive decline and pathologically by the β-sheet rich fibril plaque deposition of the amyloid-β (Aβ) peptide in the brain. While plaques are a hallmark of AD, plaque burden is not correlated with cognitive impairment. Instead, Aβ oligomers formed during the aggregation process represent the main agents of neurotoxicity, which occurs 10–20 years before patients begin to show symptoms. These oligomers are dynamic in nature and represented by a heterogeneous distribution of aggregates ranging from low- to high-molecular weight, some of which are toxic while others are not. A major difficulty in determining the pathological mechanism(s) of Aβ, developing reliable diagnostic markers for early-stage detection, as well as effective therapeutics for AD are the differentiation and characterization of oligomers formed throughout disease propagation based on their molecular features, effects on biological function, and relevance to disease propagation and pathology. Thus, it is critical to methodically identify the mechanisms of Aβ aggregation and toxicity, as well as describe the roles of different oligomers and aggregates in disease progression and molecular pathology. Here, we describe a variety of biophysical techniques used to isolate and characterize a range of Aβ oligomer populations, as well as discuss proposed mechanisms of toxicity and therapeutic interventions aimed at specific assemblies formed during the aggregation process. The approaches being used to map the misfolding and aggregation of Aβ are like what was done during the fundamental early studies, mapping protein folding pathways using combinations of biophysical techniques in concert with protein engineering. Such information is critical to the design and molecular engineering of future diagnostics and therapeutics for AD.
Collapse
|
80
|
Shi M, Chu F, Zhu F, Zhu J. Impact of Anti-amyloid-β Monoclonal Antibodies on the Pathology and Clinical Profile of Alzheimer's Disease: A Focus on Aducanumab and Lecanemab. Front Aging Neurosci 2022; 14:870517. [PMID: 35493943 PMCID: PMC9039457 DOI: 10.3389/fnagi.2022.870517] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/11/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of age-related dementia in the world, and its main pathological features consist of amyloid-β (Aβ) plaque deposits and neurofibrillary tangles formed by hyperphosphorylated tau protein. So far, only a few AD treatments approved have been applied in the clinic, but the effects of these drugs are limited only for partial symptomatic relief to patients with AD and are unable to alter AD progression. Later, all efforts for AD treatments with targeting the pathogenic factors were unsuccessful over the past decades, which suggested that the pathogenesis of AD is complex. Recently, disease-modifying therapies (DMTs) that can change the underlying pathophysiology of AD, with anti-Aβ monoclonal antibodies (mabs) (e.g., aducanumab, bapineuzumab, gantenerumab, solanezumab, and lecanemab) have been developed successively and conducted in clinical trials based on the theory that a systemic failure of cell-mediated Aβ clearance contributes to AD occurrence and progression. In the review, we summarized recent studies on the therapeutic effects and clinical trial results of these mabs in patients with AD. Specifically, we focused on the discussion of the impact of aducanumab and lecanemab on AD pathology and clinical profiles. The review provides a possible evidence for applying immunotherapy with anti-Aβ mabs in AD and analyzes lessons learned from these clinical trials in order to further study the therapeutic and adverse effects of these anti-Aβ mabs on AD.
Collapse
Affiliation(s)
- Mingchao Shi
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Fengna Chu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Division of Neurogeriatrcs, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
81
|
Argueta N, Notari E, Szigeti K. Role of Pharmacogenomics in Individualizing Treatment for Alzheimer's Disease. CNS Drugs 2022; 36:365-376. [PMID: 35352296 DOI: 10.1007/s40263-022-00915-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2022] [Indexed: 12/18/2022]
Abstract
The development of Alzheimer's disease therapeutics has been challenging, with 99% of clinical trials failing to find a significant difference between drug and placebo. While the quest continues for more effective treatments, there is emerging evidence that pharmacogenetic considerations are important factors in regard to metabolism, efficacy, and toxicity of drugs. Currently, there are five US Food and Drug Administration-approved drugs for the treatment of Alzheimer's disease; three acetylcholinesterase inhibitors, memantine, and aducanumab. Introducing a limited genetic panel consisting of APOE4, CYP2D6*10, and BChE*K would optimize acetylcholinesterase inhibitor therapy, facilitate immunotherapy risk assessment, and inform an amyloid-related imaging abnormality surveillance schedule. In view of the genetic heterogeneity of Alzheimer's disease identified in genome-wide association studies, pharmacogenetics is expected to play an increasing role in mechanism-specific treatment strategies and personalized medicine.
Collapse
Affiliation(s)
- Natalie Argueta
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Emily Notari
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| |
Collapse
|
82
|
den Boer JA, de Vries EJ, Borra RJ, Waarde AV, Lammertsma AA, Dierckx RA. Role of Brain Imaging in Drug Development for Psychiatry. Curr Rev Clin Exp Pharmacol 2022; 17:46-71. [DOI: 10.2174/1574884716666210322143458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/17/2020] [Accepted: 01/06/2021] [Indexed: 11/22/2022]
Abstract
Background:
Over the last decades, many brain imaging studies have contributed to
new insights in the pathogenesis of psychiatric disease. However, in spite of these developments,
progress in the development of novel therapeutic drugs for prevalent psychiatric health conditions
has been limited.
Objective:
In this review, we discuss translational, diagnostic and methodological issues that have
hampered drug development in CNS disorders with a particular focus on psychiatry. The role of
preclinical models is critically reviewed and opportunities for brain imaging in early stages of drug
development using PET and fMRI are discussed. The role of PET and fMRI in drug development
is reviewed emphasizing the need to engage in collaborations between industry, academia and
phase I units.
Conclusion:
Brain imaging technology has revolutionized the study of psychiatric illnesses, and
during the last decade, neuroimaging has provided valuable insights at different levels of analysis
and brain organization, such as effective connectivity (anatomical), functional connectivity patterns
and neurochemical information that may support both preclinical and clinical drug development.
Since there is no unifying pathophysiological theory of individual psychiatric syndromes and since
many symptoms cut across diagnostic boundaries, a new theoretical framework has been proposed
that may help in defining new targets for treatment and thus enhance drug development in CNS diseases.
In addition, it is argued that new proposals for data-mining and mathematical modelling as
well as freely available databanks for neural network and neurochemical models of rodents combined
with revised psychiatric classification will lead to new validated targets for drug development.
Collapse
Affiliation(s)
| | - Erik J.F. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ronald J.H. Borra
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A. Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rudi A. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
83
|
Low KJY, Venkatraman A, Mehta JS, Pervushin K. Molecular mechanisms of amyloid disaggregation. J Adv Res 2022; 36:113-132. [PMID: 35127169 PMCID: PMC8799873 DOI: 10.1016/j.jare.2021.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/13/2021] [Accepted: 05/16/2021] [Indexed: 12/17/2022] Open
Abstract
Importance of disaggregation mechanism and innate disaggregation in living systems. Different types and mechanism of disaggregation reported in literature. Structural details of the interactions and the disaggregation mechanisms. Amyloid disaggregation in protein aggregation disorders as a potential treatment. Proposed amyloid disaggregation mechanism of an ATP-independent chaperone (L-PGDS).
Introduction Protein aggregation and deposition of uniformly arranged amyloid fibrils in the form of plaques or amorphous aggregates is characteristic of amyloid diseases. The accumulation and deposition of proteins result in toxicity and cause deleterious effects on affected individuals known as amyloidosis. There are about fifty different proteins and peptides involved in amyloidosis including neurodegenerative diseases and diseases affecting vital organs. Despite the strenuous effort to find a suitable treatment option for these amyloid disorders, very few compounds had made it to unsuccessful clinical trials. It has become a compelling challenge to understand and manage amyloidosis with the increased life expectancy and ageing population. Objective While most of the currently available literature and knowledge base focus on the amyloid inhibitory mechanism as a treatment option, it is equally important to organize and understand amyloid disaggregation strategies. Disaggregation strategies are important and crucial as they are present innately functional in many living systems and dissolution of preformed amyloids may provide a direct benefit in many pathological conditions. In this review, we have compiled the known amyloid disaggregation mechanism, interactions, and possibilities of using disaggregases as a treatment option for amyloidosis. Methods We have provided the structural details using protein-ligand docking models to visualize the interaction between these disaggregases with amyloid fibrils and their respective proposed amyloid disaggregation mechanisms. Results After reviewing and comparing the different amyloid disaggregase systems and their proposed mechanisms, we presented two different hypotheses for ATP independent disaggregases using L-PGDS as a model. Conclusion Finally, we have highlighted the importance of understanding the underlying disaggregation mechanisms used by these chaperones and organic compounds before the implementation of these disaggregases as a potential treatment option for amyloidosis.
Collapse
|
84
|
Filippi M, Cecchetti G, Spinelli EG, Vezzulli P, Falini A, Agosta F. Amyloid-Related Imaging Abnormalities and β-Amyloid-Targeting Antibodies: A Systematic Review. JAMA Neurol 2022; 79:291-304. [PMID: 35099507 DOI: 10.1001/jamaneurol.2021.5205] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance After more than a decade of research and development of clinical trials testing anti-β-amyloid monoclonal antibodies (mAbs), extensive experience has been gained regarding the effects of these treatments in patients with Alzheimer disease (AD). On the verge of an expected large-scale introduction in the clinical setting after the recent US Food and Drug Administration approval of aducanumab, shared knowledge regarding amyloid-related imaging abnormalities (ARIAs) is of paramount importance. Objective To summarize available evidence on ARIAs from randomized clinical trials (RCTs) testing anti-β-amyloid mAbs in patients with AD and to provide a comprehensive update about risk factors, clinical correlates, and implications for withholding and reinitiating treatment. Evidence Review In this systematic review, a literature search of MEDLINE/PubMed, Embase, and Cochrane Library and a search of ClinicalTrials.gov were conducted through September 15, 2021. Publications describing RCTs, secondary analyses of RCT data, and case reports of ARIAs were included. Strengths of clinical data were graded according to the Oxford Centre for Evidence-Based Medicine. Findings Twenty-two RCTs, 11 secondary analyses of RCTs, and 1 case report, including in total 15 508 adult patients (8483 women [54.7%]; mean [SD] age, 69.6 [8.3] years) were selected for inclusion. Signal alterations that included parenchymal edema and sulcal effusion leading to transient hyperintensities on fluid-attenuated inversion recovery and T2-weighted sequences were termed ARIA-E, whereas those consisting of hemosiderin deposits, including parenchymal microhemorrhages and leptomeningeal superficial siderosis, were termed ARIA-H. Apolipoprotein E (ApoE) ε4 genotype was the main risk factor for both ARIA types; ARIA-E incidence was further associated with treatment dose, affecting the 55% of ApoE ε4 carriers in the high-dose aducanumab treatment group. Both ARIA types manifested early during study course, and symptomatic cases accounted for the 6.1% to 39.3% of ARIA-E cases at higher treatment doses across RCTs, whereas ARIA-H cases were generally asymptomatic. Most ARIA-E cases resolved with treatment withholding, although corticosteroid administration was required anecdotally. ARIA-E recurrence after dose reinitiation or adjustment varied from 13.8% to 25.6% across RCTs. Conclusions and Relevance Evidence suggests that ARIAs are frequent, mostly asymptomatic collateral events of amyloid-modifying therapies, highlighting the need for standardized clinical and neuroradiological management protocols in real-world clinical settings.
Collapse
Affiliation(s)
- Massimo Filippi
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy.,Neurorehabilitation Unit, IRCCS Ospedale San Raffaele, Milan, Italy.,Neurophysiology Service, IRCCS Ospedale San Raffaele, Milan, Italy.,Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Giordano Cecchetti
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy.,Neurophysiology Service, IRCCS Ospedale San Raffaele, Milan, Italy.,Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Edoardo Gioele Spinelli
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy.,Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Vezzulli
- Neuroradiology Unit and CERMAC (Centro Eccellenza Risonanza Magnetica ad Alto Campo), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Falini
- Vita-Salute San Raffaele University, Milan, Italy.,Neuroradiology Unit and CERMAC (Centro Eccellenza Risonanza Magnetica ad Alto Campo), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Agosta
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy.,Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
85
|
Lacorte E, Ancidoni A, Zaccaria V, Remoli G, Tariciotti L, Bellomo G, Sciancalepore F, Corbo M, Lombardo FL, Bacigalupo I, Canevelli M, Piscopo P, Vanacore N. Safety and Efficacy of Monoclonal Antibodies for Alzheimer's Disease: A Systematic Review and Meta-Analysis of Published and Unpublished Clinical Trials. J Alzheimers Dis 2022; 87:101-129. [PMID: 35275549 PMCID: PMC9198746 DOI: 10.3233/jad-220046] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Monoclonal antibodies (mAbs) are currently among the most investigated targets for potential disease-modifying therapies in Alzheimer's disease (AD). OBJECTIVE Our objectives were to identify all registered trials investigating mAbs in MCI due to AD or AD at any stage, retrieve available published and unpublished data from all registered trials, and analyze data on safety and efficacy outcomes. METHODS A systematic search of all registered trials on ClinicalTrials.gov and EUCT was performed. Available results were searched on both platforms and on PubMed, ISI Web of Knowledge, and The Cochrane Library. RESULTS Overall, 101 studies were identified on 27 mAbs. Results were available for 50 trials investigating 12 mAbs. For 18 trials, data were available from both published and unpublished sources, for 21 trials only from published sources, and for 11 trials only from unpublished sources. Meta-analyses of amyloid-related imaging abnormalities (ARIA) events showed overall risk ratios of 10.65 for ARIA-E and of 1.75 for ARIA-H. The meta-analysis of PET-SUVR showed an overall significant effect of mAbs in reducing amyloid (SMD -0.88), but when considering clinical efficacy, data on CDR-SB showed that treated patients had a statistically significant but clinically non-relevant lower worsening (MD -0.15). CONCLUSION Our results suggest that the risk-benefit profile of mAbs remains unclear. Research should focus on clarifying the effect of amyloid on cognitive decline, providing data on treatment response rate, and accounting for minimal clinically important difference. Research on mAbs should also investigate the possible long-term impact of ARIA events, including potential factors predicting their onset.
Collapse
Affiliation(s)
- Eleonora Lacorte
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Antonio Ancidoni
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Valerio Zaccaria
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Giulia Remoli
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Leonardo Tariciotti
- Neurosurgery Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Guido Bellomo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Francesco Sciancalepore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Flavia L. Lombardo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Ilaria Bacigalupo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Marco Canevelli
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|
86
|
Welty S, Thathiah A, Levine AS. DNA Damage Increases Secreted Aβ40 and Aβ42 in Neuronal Progenitor Cells: Relevance to Alzheimer's Disease. J Alzheimers Dis 2022; 88:177-190. [PMID: 35570488 PMCID: PMC9277680 DOI: 10.3233/jad-220030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Recent studies suggest a strong association between neuronal DNA damage, elevated levels of amyloid-β (Aβ), and regions of the brain that degenerate in Alzheimer's disease (AD). OBJECTIVE To investigate the nature of this association, we tested the hypothesis that extensive DNA damage leads to an increase in Aβ40 and Aβ42 generation. METHODS We utilized an immortalized human neuronal progenitor cell line (NPCs), ReN VM GA2. NPCs or 20 day differentiated neurons were treated with hydrogen peroxide or etoposide and allowed to recover for designated times. Sandwich ELISA was used to assess secreted Aβ40 and Aβ42. Western blotting, immunostaining, and neutral comet assay were used to evaluate the DNA damage response and processes indicative of AD pathology. RESULTS We determined that global hydrogen peroxide damage results in increased cellular Aβ40 and Aβ42 secretion 24 h after treatment in ReN GA2 NPCs. Similarly, DNA double strand break (DSB)-specific etoposide damage leads to increased Aβ40 and Aβ42 secretion 2 h and 4 h after treatment in ReN GA2 NPCs. In contrast, etoposide damage does not increase Aβ40 and Aβ42 secretion in post-mitotic ReN GA2 neurons. CONCLUSION These findings provide evidence that in our model, DNA damage is associated with an increase in Aβ secretion in neuronal progenitors, which may contribute to the early stages of neuronal pathology in AD.
Collapse
Affiliation(s)
- Starr Welty
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amantha Thathiah
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arthur Samuel Levine
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| |
Collapse
|
87
|
Verma A, Kumar Waiker D, Bhardwaj B, Saraf P, Shrivastava SK. The molecular mechanism, targets, and novel molecules in the treatment of Alzheimer's disease. Bioorg Chem 2021; 119:105562. [PMID: 34952243 DOI: 10.1016/j.bioorg.2021.105562] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/27/2021] [Accepted: 12/12/2021] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological illness that causes dementia mainly in the elderly. The challenging obstacles related to AD has freaked global healthcare system to encourage scientists in developing novel therapeutic startegies to overcome with the fatal disease. The current treatment therapy of AD provides only symptomatic relief and to some extent disease-modifying effects. The current approach for AD treatment involves designing of cholinergic inhibitors, Aβ disaggregation inducing agents, tau inhibitors and several antioxidants. Hence, extensive research on AD therapy urgently requires a deep understanding of its pathophysiology and exploration of various chemical scaffolds to design and develop a potential drug candidate for the treatment. Various issues linked between disease and therapy need to be considered such as BBB penetration capability, clinical failure and multifaceted pathophisiology requires a proper attention to develop a lead candidate. This review article covers all probable mechanisms including one of the recent areas for investigation i.e., lipid dyshomeostasis, pathogenic involvement of P. gingivalis and neurovascular dysfunction, recently reported molecules and drugs under clinical investigations and approved by FDA for AD treatment. Our summarized information on AD will attract the researchers to understand and explore current status and structural modifications of the recently reported heterocyclic derivatives in drug development for AD therapy.
Collapse
Affiliation(s)
- Akash Verma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Digambar Kumar Waiker
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Bhagwati Bhardwaj
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Poorvi Saraf
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant K Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
88
|
Forgrave LM, van der Gugten JG, Nguyen Q, DeMarco ML. Establishing pre-analytical requirements and maximizing peptide recovery in the analytical phase for mass spectrometric quantification of amyloid-β peptides 1-42 and 1-40 in CSF. Clin Chem Lab Med 2021; 60:198-206. [PMID: 34881836 DOI: 10.1515/cclm-2021-0549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Amyloid-β (Aβ) peptides in cerebrospinal fluid (CSF), including Aβ42 (residues 1-42) and Aβ40 (residues 1-40), are utilized as biomarkers in the diagnostic workup of Alzheimer's disease. Careful consideration has been given to the pre-analytical and analytical factors associated with measurement of these peptides via immunoassays; however, far less information is available for mass spectrometric methods. As such, we performed a comprehensive evaluation of pre-analytical and analytical factors specific to Aβ quantification using mass spectrometry. METHODS Using our quantitative mass spectrometry assay for Aβ42 and Aβ40 in CSF, we investigated the potential for interference from hemolysate, bilirubin, lipids, and anti-Aβ-antibodies. We also optimized the composition of the calibrator surrogate matrix and Aβ recovery during and after solid phase extraction (SPE). RESULTS There was no interreference observed with total protein up to 12 g/L, hemolysate up to 10% (v/v), bilirubin up to 0.5% (v/v), intralipid up to 1% (v/v), or anti-Aβ-antibodies at expected therapeutic concentrations. For hemolysate, bilirubin and lipids, visual CSF contamination thresholds were established. In the analytical phase, Aβ recovery was increased by ∼50% via SPE solvent modifications and by over 150% via modification of the SPE collection plate, which also extended analyte stability in the autosampler. CONCLUSIONS Attention to mass spectrometric-specific pre-analytical and analytical considerations improved analytical sensitivity and reproducibility, as well as, established CSF specimen acceptance and rejection criteria for use by the clinical laboratory.
Collapse
Affiliation(s)
- Lauren M Forgrave
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - J Grace van der Gugten
- Department of Pathology and Laboratory Medicine, St. Paul's Hospital, Providence Health Care, Vancouver, Canada
| | - Quyen Nguyen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, St. Paul's Hospital, Providence Health Care, Vancouver, Canada
| |
Collapse
|
89
|
Chen J, Li Q, Zhu J, Yuan Z, Wang T, Song J. GPR40 Agonist Ameliorate Pathological Neuroinflammation of Alzheimer's Disease via the Modulation of Gut Microbiota and Immune System, a Mini-Review. Neurotox Res 2021; 39:2175-2185. [PMID: 34505972 DOI: 10.1007/s12640-021-00408-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a central disease with high incidence, and its pathological process is closely associated with changes of some biological indicators in the periphery. Among them, the intestinal flora mainly causes a series of pathological changes such as inflammation through the immune system, which may contribute to the pathological process of AD. In this paper, we mainly focused the relationship between gut microbiota and immune system disorder in the neuropathology of AD, underlining the significance of the advanced mechanism of inflammatory response and providing a new direction for the treatment of AD.
Collapse
Affiliation(s)
- Jianheng Chen
- Department of Anesthesiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Zhu
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zijing Yuan
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Wang
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Song
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
90
|
Salloway S, Chalkias S, Barkhof F, Burkett P, Barakos J, Purcell D, Suhy J, Forrestal F, Tian Y, Umans K, Wang G, Singhal P, Budd Haeberlein S, Smirnakis K. Amyloid-Related Imaging Abnormalities in 2 Phase 3 Studies Evaluating Aducanumab in Patients With Early Alzheimer Disease. JAMA Neurol 2021; 79:13-21. [PMID: 34807243 PMCID: PMC8609465 DOI: 10.1001/jamaneurol.2021.4161] [Citation(s) in RCA: 364] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Question What are the characteristics of amyloid-related imaging abnormalities (ARIA) during aducanumab treatment in individuals with early Alzheimer disease? Findings In an integrated safety data set of 2 phase 3 clinical trials (EMERGE and ENGAGE) including 3285 participants, 425 patients (41.3%) in the combined 10 mg/kg aducanumab group (n = 1029) experienced ARIA; ARIA-edema occurred in 362 patients (35.2%), and 94 of these patients (26.0%) experienced associated symptoms (eg, headache, confusion, dizziness, and nausea). ARIA-microhemorrhage and ARIA–superficial siderosis occurred in 197 patients (19.1%) and 151 patients (14.7%), respectively. Meaning Amyloid-related imaging abnormalities occurred in approximately 40% of participants in the phase 3 studies of aducanumab, and approximately one-quarter of these patients experienced symptoms. Importance The EMERGE and ENGAGE phase 3 randomized clinical trials of aducanumab provide a robust data set to characterize amyloid-related imaging abnormalities (ARIA) that occur with treatment with aducanumab, an amyloid-β (Aβ)–targeting monoclonal antibody, in patients with mild cognitive impairment due to Alzheimer disease or mild Alzheimer disease dementia. Objective To describe the radiographic and clinical characteristics of ARIA that occurred in EMERGE and ENGAGE. Design, Setting, and Participants Secondary analysis of data from the EMERGE and ENGAGE trials, which were 2 double-blind, placebo-controlled, parallel-group, phase 3 randomized clinical trials that compared low-dose and high-dose aducanumab treatment with placebo among participants at 348 sites across 20 countries. Enrollment occurred from August 2015 to July 2018, and the trials were terminated early (March 21, 2019) based on a futility analysis. The combined studies consisted of a total of 3285 participants with Alzheimer disease who received 1 or more doses of placebo (n = 1087) or aducanumab (n = 2198; 2752 total person-years of exposure) during the placebo-controlled period. Primary data analyses were performed from November 2019 to July 2020, with additional analyses performed through July 2021. Interventions Participants were randomly assigned 1:1:1 to high-dose or low-dose intravenous aducanumab or placebo once every 4 weeks. Dose titration was used as a risk-minimization strategy. Main Outcomes and Measures Brain magnetic resonance imaging was used to monitor patients for ARIA; associated symptoms were reported as adverse events. Results Of 3285 included participants, the mean (SD) age was 70.4 (7.45) years; 1706 participants (52%) were female, 2661 (81%) had mild cognitive impairment due to Alzheimer disease, and 1777 (54%) used symptomatic medications for Alzheimer disease. A total of 764 participants from EMERGE and 709 participants from ENGAGE were categorized as withdrawn before study completion, most often owing to early termination of the study by the sponsor. Unless otherwise specified, all results represent analyses from the 10-mg/kg group. During the placebo-controlled period, 425 of 1029 patients (41.3%) experienced ARIA, with serious cases occurring in 14 patients (1.4%). ARIA-edema (ARIA-E) was the most common adverse event (362 of 1029 [35.2%]), and 263 initial events (72.7%) occurred within the first 8 doses of aducanumab; 94 participants (26.0%) with an event exhibited symptoms. Common associated symptoms among 103 patients with symptomatic ARIA-E or ARIA-H were headache (48 [46.6%]), confusion (15 [14.6%]), dizziness (11 [10.7%]), and nausea (8 [7.8%]). Incidence of ARIA-E was highest in aducanumab-treated participants who were apolipoprotein E ε4 allele carriers. Most events (479 of 488 [98.2%]) among those with ARIA-E resolved radiographically; 404 of 488 (82.8%) resolved within 16 weeks. In the placebo group, 29 of 1076 participants (2.7%) had ARIA-E (apolipoprotein E ε4 carriers: 16 of 742 [2.2%]; noncarriers, 13 of 334 [3.9%]). ARIA-microhemorrhage and ARIA–superficial siderosis occurred in 197 participants (19.1%) and 151 participants (14.7%), respectively. Conclusions and Relevance In this integrated safety data set from EMERGE and ENGAGE, the most common adverse event in the 10-mg/kg group was ARIA-E, which occurred in 362 of the 1029 patients (35.2%) in the 10-mg/kg group with at least 1 postbaseline MRI scan, with 94 patients (26.0%) experiencing associated symptoms. The most common associated symptom was headache. Trial Registrations ClinicalTrials.gov Identifiers: NCT02484547, NCT02477800
Collapse
Affiliation(s)
- Stephen Salloway
- Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | | | - Frederik Barkhof
- Institutes of Healthcare Engineering and Neurology, University College London, London, United Kingdom.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Jerome Barakos
- California Pacific Medical Center, San Francisco.,Bioclinica, Newark, California
| | - Derk Purcell
- California Pacific Medical Center, San Francisco.,Bioclinica, Newark, California
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Brod SA. Anti-Inflammatory Agents: An Approach to Prevent Cognitive Decline in Alzheimer's Disease. J Alzheimers Dis 2021; 85:457-472. [PMID: 34842189 DOI: 10.3233/jad-215125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Systemic inflammation is an organism's response to an assault by the non-self. However, that inflammation may predispose humans to illnesses targeted to organs, including Alzheimer's disease (AD). Lesions in AD have pro-inflammatory cytokines and activated microglial/monocyte/macrophage cells. Up to this point, clinical trials using anti-amyloid monoclonal antibodies have not shown success. Maybe it is time to look elsewhere by combating inflammation. Neuroinflammation with CNS cellular activation and excessive expression of immune cytokines is suspected as the "principal culprit" in the higher risk for sporadic AD. Microglia, the resident immune cell of the CNS, perivascular myeloid cells, and activated macrophages produce IL-1, IL-6 at higher levels in patients with AD. Anti-inflammatory measures that target cellular/cytokine-mediated damage provide a rational therapeutic strategy. We propose a clinical trial using oral type 1 IFNs to act as such an agent; one that decreases IL-1 and IL-6 secretion by activating lamina propria lymphocytes in the gut associated lymphoid tissue with subsequent migration to the brain undergoing inflammatory responses. A clinical trial would be double-blind, parallel 1-year clinical trial randomized 1 : 1 oral active type 1 IFN versus best medical therapy to determine whether ingested type I IFN would decrease the rate of cognitive decline in mild cognitive impairment or mild AD. Using cognitive psychometrics, imaging, and fluid biomarkers (MxA for effective type I IFN activity beyond the gut), we can determine if oral type I IFN can prevent cognitive decline in AD.
Collapse
Affiliation(s)
- Staley A Brod
- Department of Neurology, Medical College of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
92
|
Alzheimer's disease clinical trial update 2019-2021. J Neurol 2021; 269:1038-1051. [PMID: 34609602 DOI: 10.1007/s00415-021-10790-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022]
Abstract
The current clinical trial landscape targeting Alzheimer's disease (AD) is reviewed in the context of studies completed from 2019 to 2021. This review focuses on available data for observational and phase II/III clinical trial results, which will have the most impact on the field. ClinicalTrials.gov, the United States (US) comprehensive federal registry, was queried to identify completed trials. There are currently 226 interventional clinical trials and 51 observational studies completed, suspended, terminated, or withdrawn within our selected time frame. This review reveals that the role of biomarkers is expanding and although many lessons have been learned, many challenges remain when targeting disease modification of AD through amyloid and tau. In addition, to halt or slow clinical progression of AD, new clinical and observational trials are focusing on prevention as well as the role of more diverse biological processes known to influence AD pathology.
Collapse
|
93
|
Wang J, Cao H. Zebrafish and Medaka: Important Animal Models for Human Neurodegenerative Diseases. Int J Mol Sci 2021; 22:10766. [PMID: 34639106 PMCID: PMC8509648 DOI: 10.3390/ijms221910766] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Animal models of human neurodegenerative disease have been investigated for several decades. In recent years, zebrafish (Danio rerio) and medaka (Oryzias latipes) have become popular in pathogenic and therapeutic studies about human neurodegenerative diseases due to their small size, the optical clarity of embryos, their fast development, and their suitability to large-scale therapeutic screening. Following the emergence of a new generation of molecular biological technologies such as reverse and forward genetics, morpholino, transgenesis, and gene knockout, many human neurodegenerative disease models, such as Parkinson's, Huntington's, and Alzheimer's, were constructed in zebrafish and medaka. These studies proved that zebrafish and medaka genes are functionally conserved in relation to their human homologues, so they exhibit similar neurodegenerative phenotypes to human beings. Therefore, fish are a suitable model for the investigation of pathologic mechanisms of neurodegenerative diseases and for the large-scale screening of drugs for potential therapy. In this review, we summarize the studies in modelling human neurodegenerative diseases in zebrafish and medaka in recent years.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
94
|
Nimmo JT, Kelly L, Verma A, Carare RO, Nicoll JAR, Dodart JC. Amyloid-β and α-Synuclein Immunotherapy: From Experimental Studies to Clinical Trials. Front Neurosci 2021; 15:733857. [PMID: 34539340 PMCID: PMC8441015 DOI: 10.3389/fnins.2021.733857] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022] Open
Abstract
Alzheimer’s disease and Lewy body diseases are the most common causes of neurodegeneration and dementia. Amyloid-beta (Aβ) and alpha-synuclein (αSyn) are two key proteins involved in the pathogenesis of these neurodegenerative diseases. Immunotherapy aims to reduce the harmful effects of protein accumulation by neutralising toxic species and facilitating their removal. The results of the first immunisation trial against Aβ led to a small percentage of meningoencephalitis cases which revolutionised vaccine design, causing a shift in the field of immunotherapy from active to passive immunisation. While the vast majority of immunotherapies have been developed for Aβ and tested in Alzheimer’s disease, the field has progressed to targeting other proteins including αSyn. Despite showing some remarkable results in animal models, immunotherapies have largely failed final stages of clinical trials to date, with the exception of Aducanumab recently licenced in the US by the FDA. Neuropathological findings translate quite effectively from animal models to human trials, however, cognitive and functional outcome measures do not. The apparent lack of translation of experimental studies to clinical trials suggests that we are not obtaining a full representation of the effects of immunotherapies from animal studies. Here we provide a background understanding to the key concepts and challenges involved in therapeutic design. This review further provides a comprehensive comparison between experimental and clinical studies in Aβ and αSyn immunotherapy and aims to determine the possible reasons for the disconnection in their outcomes.
Collapse
Affiliation(s)
- Jacqui Taryn Nimmo
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Louise Kelly
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ajay Verma
- Yumanity Therapeutics, Boston, MA, United States
| | - Roxana O Carare
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | |
Collapse
|
95
|
Stonebarger GA, Bimonte-Nelson HA, Urbanski HF. The Rhesus Macaque as a Translational Model for Neurodegeneration and Alzheimer's Disease. Front Aging Neurosci 2021; 13:734173. [PMID: 34539388 PMCID: PMC8446616 DOI: 10.3389/fnagi.2021.734173] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/12/2021] [Indexed: 12/01/2022] Open
Abstract
A major obstacle to progress in understanding the etiology of normative and pathological human brain aging is the availability of suitable animal models for experimentation. The present article will highlight our current knowledge regarding human brain aging and neurodegeneration, specifically in the context of Alzheimer's disease (AD). Additionally, it will examine the use of the rhesus macaque monkey as a pragmatic translational animal model in which to study underlying causal mechanisms. Specifically, the discussion will focus on behavioral and protein-level brain changes that occur within the central nervous system (CNS) of aged monkeys, and compare them to the changes observed in humans during clinically normative aging and in AD.
Collapse
Affiliation(s)
- Gail A. Stonebarger
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
96
|
Martinkova J, Quevenco FC, Karcher H, Ferrari A, Sandset EC, Szoeke C, Hort J, Schmidt R, Chadha AS, Ferretti MT. Proportion of Women and Reporting of Outcomes by Sex in Clinical Trials for Alzheimer Disease: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2124124. [PMID: 34515784 DOI: 10.1001/jamanetworkopen.2021.24124] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
IMPORTANCE Women represent two-thirds of patients with Alzheimer disease (AD), and sex differences might affect results of randomized clinical trials (RCTs). However, little information exists on differences in sex as reported in RCTs for AD. OBJECTIVE To assess the ratio of females to males and the reporting of sex-stratified data in large pharmaceutical RCTs for AD. DATA SOURCES A search for pharmaceutical RCTs for AD was conducted on September 4, 2019, using ClinicalTrials.gov with the key word Alzheimer disease, and articles related to those trials were identified using the PubMed, Scopus, and Google Scholar databases. Searches were conducted between September 4 and October 31, 2019, and between April 15 and May 31, 2020. STUDY SELECTION Controlled RCTs that had more than 100 participants and tested the efficacy of drugs or herbal extracts were included. Of 1047 RCTs identified, 409 were published and therefore screened. A total of 77 articles were included in the final analysis, including 56 primary articles on AD, 13 secondary articles on AD, and 8 articles on mild cognitive impairment. DATA EXTRACTION AND SYNTHESIS The location and date of publication; number, sex, and age of patients enrolled; disease severity; experimental or approved status of the drug; and whether the study included a sex-stratified analysis in the protocol, methods, or results were extracted by 1 reviewer for each article, and the meta-analysis followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline. Data were analyzed using a mixed-effects model. MAIN OUTCOMES AND MEASURES The mean proportion of women enrolled in the trials and the associations between prespecified variables were analyzed. The proportion of articles that included sex-stratified results and the temporal trends in the reporting of these results were also studied. RESULTS In this review of 56 RCTs for AD involving 39 575 participants, 23 348 women (59.0%) were included. The mean (SD) proportion of women in RCTs of approved drugs was 67.3% (6.9%), and in RCTs of experimental drugs was 57.9% (5.9%). The proportion of women in RCTs of experimental drugs was significantly lower than the proportion of women in the general population with AD in the US (62.1%; difference, -4.56% [95% CI, -6.29% to -2.87%]; P < .001) and Europe (68.2%; difference, -10.67% [95% CI, -12.39% to -8.97%]; P < .001). Trials of approved drugs had a higher probability of including women than trials of experimental drugs (odds ratio [OR], 1.26; 95% CI, 1.05-1.52; P = .02). Both the severity of AD at baseline and the trial location were associated with the probability of women being enrolled in trials (severity: OR, 0.98; 95% CI, 0.97-1.00; P = .02; location in Europe: OR, 1.26; 95% CI, 1.05-1.52; P = .01; location in North America: OR, 0.81; 95% CI, 0.71-0.93; P = .002). Only 7 articles (12.5%) reported sex-stratified results, with an increasing temporal trend (R, 0.30; 95% CI, 0.05-0.59; P = .03). CONCLUSIONS AND RELEVANCE In this systematic review and meta-analysis, the proportion of women in RCTs for AD, although higher than the proportion of men, was significantly lower than that in the general population. Only a small proportion of trials reported sex-stratified results. These findings support strategies to improve diversity in enrollment and data reporting in RCTs for AD.
Collapse
Affiliation(s)
- Julie Martinkova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
- Women's Brain Project, Guntershausen, Switzerland
| | - Frances-Catherine Quevenco
- Women's Brain Project, Guntershausen, Switzerland
- Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | | | | | | | - Cassandra Szoeke
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| | - Reinhold Schmidt
- Department of Neurogeriatrics, University Clinic of Neurology, Medical University Graz, Graz, Austria
| | | | | |
Collapse
|
97
|
Bomasang-Layno E, Bronsther R. Diagnosis and Treatment of Alzheimer's Disease:: An Update. Dela J Public Health 2021; 7:74-85. [PMID: 34604768 PMCID: PMC8482985 DOI: 10.32481/djph.2021.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
98
|
Chiang TI, Yu YH, Lin CH, Lane HY. Novel Biomarkers of Alzheimer's Disease: Based Upon N-methyl-D-aspartate Receptor Hypoactivation and Oxidative Stress. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:423-433. [PMID: 34294612 PMCID: PMC8316669 DOI: 10.9758/cpn.2021.19.3.423] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Early detection and prevention of Alzheimer’s disease (AD) is important. The current treatment for early AD is acetylcholine esterase inhibitors (AChEIs); however, the efficacy is poor. Besides, AChEI did not show efficacy in mild cognitive impairment (MCI). Beta-amyloid (Aβ) deposits have been regarded to be highly related to the pathogenesis of AD. However, many clinical trials aiming at the clearance of Aβ deposits failed to improve the cognitive decline of AD, even at its early phase. There should be other important mechanisms unproven in the course of AD and MCI. Feasible biomarkers for the diagnosis and treatment response of AD are lacking to date. The N-methyl-D-aspartate receptor (NMDAR) activation plays an important role in learning and memory. On the other hand, oxidative stress has been regarded to contribute to aging with the assumption that free radicals damage cell constituents and connective tissues. Our recent study found that an NMDAR enhancer, sodium benzoate (the pivotal inhibitor of D-amino acid oxidase [DAAO]), improved the cognitive and global function of patients with early-phase AD. Further, we found that peripheral DAAO levels were higher in patients with MCI and AD than healthy controls. We also found that sodium benzoate was able to change the activity of antioxidant. These pieces of evidence suggest that the NMDAR function is associated with anti-oxidation, and have potential to be biomarkers for the diagnosis and treatment response of AD.
Collapse
Affiliation(s)
- Ting-I Chiang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Hsiang Yu
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.,Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
99
|
Espay AJ, Sturchio A, Schneider LS, Ezzat K. Soluble Amyloid-β Consumption in Alzheimer's Disease. J Alzheimers Dis 2021; 82:1403-1415. [PMID: 34151810 DOI: 10.3233/jad-210415] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain proteins function in their soluble, native conformation and cease to function when transformed into insoluble aggregates, also known as amyloids. Biophysically, the soluble-to-insoluble phase transformation represents a process of polymerization, similar to crystallization, dependent on such extrinsic factors as concentration, pH, and a nucleation surface. The resulting cross-β conformation of the insoluble amyloid is markedly stable, making it an unlikely source of toxicity. The spread of brain amyloidosis can be fully explained by mechanisms of spontaneous or catalyzed polymerization and phase transformation instead of active replication, which is an enzyme- and energy-requiring process dependent on a specific nucleic acid code for the transfer of biological information with high fidelity. Early neuronal toxicity in Alzheimer's disease may therefore be mediated to a greater extent by a reduction in the pool of soluble, normal-functioning protein than its accumulation in the polymerized state. This alternative loss-of-function hypothesis of pathogenicity can be examined by assessing the clinical and neuroimaging effects of administering non-aggregating peptide analogs to replace soluble amyloid-β levels above the threshold below which neuronal toxicity may occur. Correcting the depletion of soluble amyloid-β, however, would only exemplify 'rescue medicine.' Precision medicine will necessitate identifying the pathogenic factors catalyzing the protein aggregation in each affected individual. Only then can we stratify patients for etiology-specific treatments and launch precision medicine for Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Lon S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kariem Ezzat
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
100
|
Dolton MJ, Chesterman A, Moein A, Sink KM, Waitz A, Blondeau K, Kerchner GA, Hu N, Brooks L, Wetzel-Smith MK, Roden A, Deshmukh A, Peng K, Carrasco-Triguero M, Smith J, Ostrowitzki S, Quartino A. Safety, Tolerability, and Pharmacokinetics of High-Volume Subcutaneous Crenezumab, With and Without Recombinant Human Hyaluronidase in Healthy Volunteers. Clin Pharmacol Ther 2021; 110:1337-1348. [PMID: 34347883 DOI: 10.1002/cpt.2385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/22/2021] [Indexed: 11/07/2022]
Abstract
Compared with intravenous formulations, subcutaneous (s.c.) formulations of therapeutic monoclonal antibodies may provide increased patient access and more convenient administration options, although historically high-volume s.c. administration (> 10-15 mL) has been challenging. We report results from two phase I studies in healthy participants (GP29523 and GP40201) that evaluated s.c. crenezumab, an anti-Aβ monoclonal antibody in development for individuals at risk for autosomal-dominant Alzheimer's disease. GP29523 assessed safety, tolerability, and pharmacokinetics (PK) in 68 participants (aged 50-80 years) who received single ascending doses (600-7,200 mg) of crenezumab or placebo (4-40 mL). GP40201 assessed safety, tolerability, and PK in 72 participants (aged 18-80 years) who received different combinations of dose (1,700-6,800 mg), infusion volume (10-40 mL), and flow rate (2-4 mL/minute), with/without recombinant human hyaluronidase (rHuPH20). There were no serious or dose-limiting adverse events in either study. There were no meaningful differences in pain scores among reference placebo (4 mL), test placebo (4-40 mL), or crenezumab (600-7,200 mg) in GP29523, or across treatments with varying infusion volume, flow rate, dose, or rHuPH20 co-administration or concentration in GP40201. Transient erythema was the most common infusion site reaction in both studies. In GP40201 at volumes of ≥ 20 mL, rHuPH20 co-administration appeared to reduce infusion site swelling incidence, but, in some cases, was associated with larger areas of infusion site erythema. Crenezumab exhibited approximately dose-proportional PK, and s.c. bioavailability was 66% and independent of dose or rHuPH20 co-administration. High-dose, high-concentration, high-volume s.c. crenezumab formulated with/without rHuPH20 was well-tolerated in healthy participants, with an acceptable safety profile.
Collapse
Affiliation(s)
| | | | - Anita Moein
- Genentech, Inc., South San Francisco, California, USA
| | - Kaycee M Sink
- Genentech, Inc., South San Francisco, California, USA
| | - Ariel Waitz
- Genentech, Inc., South San Francisco, California, USA
| | | | | | - Nan Hu
- Genentech, Inc., South San Francisco, California, USA
| | - Logan Brooks
- Genentech, Inc., South San Francisco, California, USA
| | | | - Amanda Roden
- Genentech, Inc., South San Francisco, California, USA
| | - Ajay Deshmukh
- Genentech, Inc., South San Francisco, California, USA
| | - Kun Peng
- Genentech, Inc., South San Francisco, California, USA
| | | | - Jill Smith
- Roche Products Limited, Welwyn Garden City, UK
| | | | | |
Collapse
|