51
|
Kile S, Au W, Parise C, Rose K, Donnel T, Hankins A, Au Y, Chan M, Ghassemi A. Five-year outcomes after IVIG for mild cognitive impairment due to alzheimer disease. BMC Neurosci 2021; 22:49. [PMID: 34362303 PMCID: PMC8349062 DOI: 10.1186/s12868-021-00651-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 07/23/2021] [Indexed: 11/10/2022] Open
Abstract
Background The purpose of this study was to assess the five-year treatment effects of a short course of intravenous immunoglobulin (IVIG) in subjects with mild cognitive impairment (MCI) due to Alzheimer disease (AD). Methods Fifty subjects 50 to 84 years of age with MCI due to AD were administered 0.4 g/kg 10% IVIG or 0.9% saline every two weeks x five doses in a randomized double-blinded design as part of a two-year study. Twenty-seven subjects completed an additional three-year extension study. MRI brain imaging, cognitive testing, and conversion to dementia were assessed annually. Participants were stratified into early MCI (E-MCI) and late MCI (L-MCI). The primary endpoint was brain atrophy measured as annualized percent change in ventricular volume (APCV) annually for five years. ANOVA was used to compare annualized percent change in ventricular volume from baseline between the groups adjusting for MCI status (E-MCI, L-MCI). Results Differences in brain atrophy between the groups, which were statistically significant after one year, were no longer significant after five years. IVIG-treated L-MCI subjects did demonstrate a delay in conversion to dementia of 21.4 weeks. Conclusion An eight-week course of IVIG totaling 2 g/kg in MCI is safe but is not sufficient to sustain an initial reduction in brain atrophy or a temporary delay in conversion to dementia at five years. Other dosing strategies of IVIG in the early stages of AD should be investigated to assess more sustainable disease-modifying effects. Trial registration ClinicalTrials.gov NCT01300728. Registered 23 February 2011.
Collapse
Affiliation(s)
- Shawn Kile
- Sutter Neuroscience Institute, 2800 L Street, Suite 500, Sacramento, CA, 95816, USA.
| | - William Au
- Sutter Neuroscience Institute, 2800 L Street, Suite 500, Sacramento, CA, 95816, USA
| | - Carol Parise
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | - Kimberley Rose
- Sutter Neuroscience Institute, 2800 L Street, Suite 500, Sacramento, CA, 95816, USA
| | - Tammy Donnel
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | - Andrea Hankins
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | - Yvonne Au
- Sutter Neuroscience Institute, 2800 L Street, Suite 500, Sacramento, CA, 95816, USA
| | | | | |
Collapse
|
52
|
Liu K, Li J, Raghunathan R, Zhao H, Li X, Wong STC. The Progress of Label-Free Optical Imaging in Alzheimer's Disease Screening and Diagnosis. Front Aging Neurosci 2021; 13:699024. [PMID: 34366828 PMCID: PMC8341907 DOI: 10.3389/fnagi.2021.699024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/02/2021] [Indexed: 01/13/2023] Open
Abstract
As the major neurodegenerative disease of dementia, Alzheimer's disease (AD) has caused an enormous social and economic burden on society. Currently, AD has neither clear pathogenesis nor effective treatments. Positron emission tomography (PET) and magnetic resonance imaging (MRI) have been verified as potential tools for diagnosing and monitoring Alzheimer's disease. However, the high costs, low spatial resolution, and long acquisition time limit their broad clinical utilization. The gold standard of AD diagnosis routinely used in research is imaging AD biomarkers with dyes or other reagents, which are unsuitable for in vivo studies owing to their potential toxicity and prolonged and costly process of the U.S. Food and Drug Administration (FDA) approval for human use. Furthermore, these exogenous reagents might bring unwarranted interference to mechanistic studies, causing unreliable results. Several label-free optical imaging techniques, such as infrared spectroscopic imaging (IRSI), Raman spectroscopic imaging (RSI), optical coherence tomography (OCT), autofluorescence imaging (AFI), optical harmonic generation imaging (OHGI), etc., have been developed to circumvent this issue and made it possible to offer an accurate and detailed analysis of AD biomarkers. In this review, we present the emerging label-free optical imaging techniques and their applications in AD, along with their potential and challenges in AD diagnosis.
Collapse
Affiliation(s)
- Kai Liu
- Translational Biophotonics Laboratory, Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, United States
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jiasong Li
- Translational Biophotonics Laboratory, Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, United States
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, United States
| | - Raksha Raghunathan
- Translational Biophotonics Laboratory, Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, United States
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, United States
| | - Hong Zhao
- Translational Biophotonics Laboratory, Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, United States
| | - Xuping Li
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, United States
| | - Stephen T. C. Wong
- Translational Biophotonics Laboratory, Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, United States
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
53
|
Zhu L, Xu L, Wu X, Deng F, Ma R, Liu Y, Huang F, Shi L. Tau-Targeted Multifunctional Nanoinhibitor for Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2021; 13:23328-23338. [PMID: 33999598 DOI: 10.1021/acsami.1c00257] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
With the failure of various amyloid-β-targeted drugs for Alzheimer's disease (AD) in clinical trials, tau protein has gained growing attention as an alternative therapeutic target in recent years. The aggregation of tau exerts neurotoxicity, and its spreading in the brain is associated with increasing severity of clinical symptoms for AD patients; thus tau-targeting therapies hold great potential against AD. Here, a tau-targeted multifunctional nanoinhibitor based on self-assembled polymeric micelles decorated with tau-binding peptide is devised for AD treatment. Through the multivalent binding effect with the aggregating protein, this nanoinhibitor is capable of efficiently inhibiting tau protein aggregation, recognizing tau aggregates, and blocking their seeding in neural cells, thus remarkably mitigating tau-mediated cytotoxicity. Moreover, the formed nanoinhibitor-tau complex after binding is more easily degraded than mature tau aggregates, which will be conducive to enhance the therapeutic effect. We believe that this multifunctional nanoinhibitor will promote the development of new antitau strategies for AD treatment.
Collapse
Affiliation(s)
- Lin Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Linlin Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Xiaohui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Fei Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Rujiang Ma
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Fan Huang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P. R. China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
54
|
Kowalska M, Wize K, Prendecki M, Lianeri M, Kozubski W, Dorszewska J. Genetic Variants and Oxidative Stress in Alzheimer's Disease. Curr Alzheimer Res 2021; 17:208-223. [PMID: 32091332 DOI: 10.2174/1567205017666200224121447] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/08/2020] [Accepted: 02/12/2020] [Indexed: 12/26/2022]
Abstract
In an aging society, the number of people suffering from Alzheimer's Disease (AD) is still growing. Currently, intensive research is being carried out on the pathogenesis of AD. The results of these studies indicated that oxidative stress plays an important role in the onset and development of this disease. Moreover, in AD oxidative stress is generated by both genetic and biochemical factors as well as the functioning of the systems responsible for their formation and removal. The genetic factors associated with the regulation of the redox system include TOMM40, APOE, LPR, MAPT, APP, PSEN1 and PSEN2 genes. The most important biochemical parameters related to the formation of oxidative species in AD are p53, Homocysteine (Hcy) and a number of others. The formation of Reactive Oxygen Species (ROS) is also related to the efficiency of the DNA repair system, the effectiveness of the apoptosis, autophagy and mitophagy processes as well as the antioxidant potential. However, these factors are responsible for the development of many disorders, often with similar clinical symptoms, especially in the early stages of the disease. The discovery of markers of the early diagnosis of AD may contribute to the introduction of pharmacotherapy and slow down the progression of this disease.
Collapse
Affiliation(s)
- Marta Kowalska
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Wize
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Prendecki
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Margarita Lianeri
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
55
|
Li QS, De Muynck L. Differentially expressed genes in Alzheimer's disease highlighting the roles of microglia genes including OLR1 and astrocyte gene CDK2AP1. Brain Behav Immun Health 2021; 13:100227. [PMID: 34589742 PMCID: PMC8474442 DOI: 10.1016/j.bbih.2021.100227] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with abnormal tau and amyloid-β accumulation in the brain, leading to neurofibrillary tangles, neuropil threads and extracellular amyloid-β plaques. Treatment is limited to symptom management, a disease-modifying therapy is not available. To advance search of therapy approaches, there is a continued need to identify targets for disease intervention both by confirming existing hypotheses and generating new hypotheses. METHODS We conducted a mRNA-seq study to identify genes associated with AD in post-mortem brain samples from the superior temporal gyrus (STG, n = 76), and inferior frontal gyrus (IFG, n = 65) brain regions. Differentially expressed genes (DEGs) were identified correcting for gender and surrogate variables to capture hidden variation not accounted for by pre-planned covariates. The results from this study were compared with the transcriptome studies from the Accelerated Medicine Partnership - Alzheimer's Disease (AMP-AD) initiative. Over-representation and gene set enrichment analysis (GSEA) was used to identify disease-associated pathways. Protein-protein interaction (PPI) and weighted gene co-expression network analysis (WGCNA) analyses were carried out and co-expressed gene modules and their hub genes were identified and associated with additional phenotypic traits of interest. RESULTS Several hundred mRNAs were differentially expressed between AD cases and cognitively normal controls in the STG, while no and few transcripts met the same criteria (adjusted p less than 0.05 and fold change greater than 1.2) in the IFG. The findings were consistent at the gene set level with two out of three cohorts from AMP-AD. PPI analysis suggested that the DEGs were enriched in protein-protein interactions than expected by random chance. Over-representation and GSEA analysis suggested genes playing roles in neuroinflammation, amyloid-β, autophagy and trafficking being important for the AD disease process. At the gene level, 10 genes from the STG that were consistently differentially expressed in this study and in the MSBB study (one of the three cohorts within the AMP-AD initiative) were enriched in microglial genes (TREM2, C3AR1, ITGAX, OLR1, CD74, and HLA-DRA), but also included genes with a broader cell type expression pattern such as CDK2AP1. Among the DEGs with supporting evidence from an independent study, CDK2AP1 (most abundantly expressed in astrocyte) was the transcript with strongest association with antemortem cognitive measure (last Mini-Mental State Examination score) and neurofibril tangle burden but also associated with amyloid plaque burden, while OLR1 was the transcript with strongest association with amyloid plaque burden. GSEA and over-representation analyses revealed gene sets related to immune processes including neutrophil degranulation, interleukin 10 signaling, and interferon gamma signaling, complement and coagulation cascades, phosphatidylinositol signaling system, phagosome and neurotransmitter receptors and postsynaptic signal transmission were enriched from this study and replicated in an independent study. CONCLUSION This study identified differential gene sets, common with two out of three AMP-AD cohorts (ROSMAP and MSBB) and highlights microglia and astrocyte as the key cell-types with DGEs associated with AD clinical diagnosis, and/or antemortem cognitive measure as well as neuropathological indices. Future meta-analysis and causal inferential analysis will be helpful in pinpointing the most relevant pathways and genes to intervene.
Collapse
Affiliation(s)
- Qingqin S. Li
- Neuroscience Department, Janssen Research & Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA
| | - Louis De Muynck
- Neuroscience Department, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| |
Collapse
|
56
|
Kapoor A, Bartha R, Black SE, Borrie M, Freedman M, Gao F, Herrmann N, Mandzia J, Ozzoude M, Ramirez J, Scott CJM, Symons S, Fischer CE, Frank A, Seitz D, Wolf MU, Verhoeff NPLG, Naglie G, Reichman W, Masellis M, Mitchell SB, Tang-Wai DF, Tartaglia MC, Kumar S, Pollock BG, Rajji TK, Finger E, Pasternak SH, Swartz RH. Structural Brain Magnetic Resonance Imaging to Rule Out Comorbid Pathology in the Assessment of Alzheimer's Disease Dementia: Findings from the Ontario Neurodegenerative Disease Research Initiative (ONDRI) Study and Clinical Trials Over the Past 10 Years. J Alzheimers Dis 2021; 74:747-757. [PMID: 32116253 PMCID: PMC7242844 DOI: 10.3233/jad-191097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/OBJECTIVE Structural brain magnetic resonance imaging (MRI) is not mandatory in Alzheimer's disease (AD) research or clinical guidelines. We aimed to explore the use of structural brain MRI in AD/mild cognitive impairment (MCI) trials over the past 10 years and determine the frequency with which inclusion of standardized structural MRI acquisitions detects comorbid vascular and non-vascular pathologies. METHODS We systematically searched ClinicalTrials.gov for AD clinical trials to determine their neuroimaging criteria and then used data from an AD/MCI cohort who underwent standardized MRI protocols, to determine type and incidence of clinically relevant comorbid pathologies. RESULTS Of 210 AD clinical trials, 105 (50%) included structural brain imaging in their eligibility criteria. Only 58 (27.6%) required MRI. 16,479 of 53,755 (30.7%) AD participants were in trials requiring MRI. In the observational AD/MCI cohort, 141 patients met clinical criteria; 22 (15.6%) had relevant MRI findings, of which 15 (10.6%) were exclusionary for the study. DISCUSSION In AD clinical trials over the last 10 years, over two-thirds of participants could have been enrolled without brain MRI and half without even a brain CT. In a study sample, relevant comorbid pathology was found in 15% of participants, despite careful screening. Standardized structural MRI should be incorporated into NIA-AA diagnostic guidelines (when available) and research frameworks routinely to reduce diagnostic heterogeneity.
Collapse
Affiliation(s)
| | - Robert Bartha
- Robarts Research Institute and the Department of Medical Biophysics, the University of Western Ontario, London, ON, Canada
| | - Sandra E Black
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Michael Borrie
- Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada
| | - Morris Freedman
- University of Toronto, Toronto, ON, Canada.,Rotman Research Institute of Baycrest Health Sciences, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - Fuqiang Gao
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Nathan Herrmann
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Jennifer Mandzia
- Western University, London, ON, Canada.,London Health Sciences Centre, London, ON, Canada
| | - Miracle Ozzoude
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Joel Ramirez
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Sean Symons
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Corinne E Fischer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michaels Hospital, Toronto, ON, Canada
| | | | - Dallas Seitz
- Department of Psychiatry and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Michael Uri Wolf
- University of Toronto, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | | | - Gary Naglie
- University of Toronto, Toronto, ON, Canada.,Rotman Research Institute of Baycrest Health Sciences, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - William Reichman
- University of Toronto, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Sara B Mitchell
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - David F Tang-Wai
- University of Toronto, Toronto, ON, Canada.,University Health Network Memory Clinic, University of Toronto, Division of Neurology & Geriatric Medicine, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Sanjeev Kumar
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Bruce G Pollock
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tarek K Rajji
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Elizabeth Finger
- Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada.,Western University, London, ON, Canada
| | - Stephen H Pasternak
- Robarts Research Institute and the Department of Medical Biophysics, the University of Western Ontario, London, ON, Canada.,Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada.,Western University, London, ON, Canada
| | | | - Richard H Swartz
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| |
Collapse
|
57
|
Wang L, Mao X. Role of Retinal Amyloid-β in Neurodegenerative Diseases: Overlapping Mechanisms and Emerging Clinical Applications. Int J Mol Sci 2021; 22:2360. [PMID: 33653000 PMCID: PMC7956232 DOI: 10.3390/ijms22052360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/03/2023] Open
Abstract
Amyloid-β (Aβ) accumulations have been identified in the retina for neurodegeneration-associated disorders like Alzheimer's disease (AD), glaucoma, and age-related macular degeneration (AMD). Elevated retinal Aβ levels were associated with progressive retinal neurodegeneration, elevated cerebral Aβ accumulation, and increased disease severity with a decline in cognition and vision. Retinal Aβ accumulation and its pathological effects were demonstrated to occur prior to irreversible neurodegeneration, which highlights its potential in early disease detection and intervention. Using the retina as a model of the brain, recent studies have focused on characterizing retinal Aβ to determine its applicability for population-based screening of AD, which warrants a further understanding of how Aβ manifests between these disorders. While current treatments directly targeting Aβ accumulations have had limited results, continued exploration of Aβ-associated pathological pathways may yield new therapeutic targets for preserving cognition and vision. Here, we provide a review on the role of retinal Aβ manifestations in these distinct neurodegeneration-associated disorders. We also discuss the recent applications of retinal Aβ for AD screening and current clinical trial outcomes for Aβ-associated treatment approaches. Lastly, we explore potential future therapeutic targets based on overlapping mechanisms of pathophysiology in AD, glaucoma, and AMD.
Collapse
Affiliation(s)
- Liang Wang
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
58
|
Brand A, De Angelis V, Vuk T, Garraud O, Lozano M, Politis D. Review of indications for immunoglobulin (IG) use: Narrowing the gap between supply and demand. Transfus Clin Biol 2021; 28:96-122. [PMID: 33321210 DOI: 10.1016/j.tracli.2020.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cellular blood components and plasma-derived medicinal products (PDMPs) are obtained from blood donated by volunteers. In a growing number of countries, in line with World Health Organization advice issued since the mid-1970s, donors are not remunerated. In recent decades, considerable efforts have been made to restrict the indications for labile blood components to those based on evidence, to ensure efficacy and safety. By contrast, the producers of PDMPs have developed pathogen reduction techniques for inactivating the microorganisms in (pooled) plasma, but little attention has been paid to the pertinence of the clinical indications for these products. The use of blood, and of erythrocytes in particular, has declined by almost 40%, but the use of immunoglobulins (IG) tripled between 2004 and 2018, making it necessary to pay donors to obtain sufficient blood to meet the market demand for these products. We analyzed the reasons for this increase to unsustainable levels of use, by investigating (practice) guidelines, recommendations, Cochrane data analyses and systematic reviews for clinical indications for IG over time. We found no new evidence explaining the huge increase up to 2018 or the predicted 5-7% yearly annual increase until 2024. For some former evidence-based indications, biologics have largely replaced IG, but the administration of IG for doubtful indications (up to 40%) has not decreased in recent decades. The main development since 2004 is that IG use in Europe has become market-driven rather than evidence-guided. As transfusion specialists and blood therapists, we must raise the alarm that this situation is likely to continue in the absence of good clinical studies determining the place of IG alongside other treatments, and for as long as market profitability remains the dominant driving force. We discuss here approaches for reversing this trend and moving towards European self-sufficiency through non-remunerated voluntary blood donation.
Collapse
Affiliation(s)
- A Brand
- Sanquin Research, Center for Clinical Transfusion Research, Leiden and Jon J van Rood Center for Clinical Transfusion Science, Leiden University Medical Center, 2333 Leiden, the Netherlands.
| | | | - T Vuk
- Croatian Institute of Transfusion Medicine, 10000 Zagreb, Croatia
| | - O Garraud
- Inserm_1059, University of Lyon-Saint-Étienne, Saint-Étienne, France; Institut National de la Transfusion Sanguine, 75015 Paris, France
| | - M Lozano
- Department of Hemotherapy and Hemostasis. ICMHO, University Clinic Hospital, IDIBAPS, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - D Politis
- Coordinating Centre for Hemovigilance and Surveillance of Transfusion (SKAEM) of the Hellenic National Public Health Centre, Agravon street 3-5, 15123 Marousi, Greece
| |
Collapse
|
59
|
Ayton S, Bush AI. β-amyloid: The known unknowns. Ageing Res Rev 2021; 65:101212. [PMID: 33188924 DOI: 10.1016/j.arr.2020.101212] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) stands out as a major disease without any form of preventative or disease modifying therapy. This is not for lack of trying. 33 phase 3 clinical trials of drugs targeting amyloid beta (Aβ) have failed to slow cognitive decline in AD. The field is at a cross-roads about whether to continue anti-Aβ therapy or more actively pursue alternative targets. With the burden of this disease to patients, families, and healthcare budgets growing yearly, the need for disease modifying AD therapies has become one of the highest priorities in all of medicine. While pathology, genetic and biochemical data offer a popular narrative for the causative role of Aβ, there are alternative explanations, and dissenting findings that, now more than ever, warrant thorough reanalysis. This review questions the major assumptions about Aβ on which therapies for AD were premised, and invites renewed interrogation into AD pathogenesis.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
60
|
Hannestad J, Koborsi K, Klutzaritz V, Chao W, Ray R, Páez A, Jackson S, Lohr S, Cummings JL, Kay G, Nikolich K, Braithwaite S. Safety and tolerability of GRF6019 in mild-to-moderate Alzheimer's disease dementia. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12115. [PMID: 33344754 PMCID: PMC7744029 DOI: 10.1002/trc2.12115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/30/2020] [Accepted: 10/26/2020] [Indexed: 11/09/2022]
Abstract
INTRODUCTION This phase 2 trial evaluated the safety, tolerability, and feasibility of repeated infusions of the plasma fraction GRF6019 in mild-to-moderate Alzheimer's disease. METHODS In this randomized, double-blind, dose-comparison trial, 47 patients were randomized 1:1 to receive daily infusions of 100 mL (n = 24) or 250 mL (n = 23) of GRF6019 for 5 consecutive days over two dosing periods separated by a treatment-free interval of 3 months. RESULTS The mean (standard deviation [SD]) age of the enrolled patients was 74.3 (6.9), and 62% were women. Most adverse events (55%) were mild, with no clinically significant differences in safety or tolerability between the two dose levels. The mean (SD) baseline Mini-Mental State Examination score was 20.6 (3.7) in the 100 mL group and 19.6 (3.7) in the 250 mL group; at 24 weeks, the within-patient mean change from baseline was -1.0 points (95% confidence interval [CI], -3.1 to 1.1) in the 100 mL group and +1.5 points (95% CI, -0.4 to 3.3) in the 250 mL group. The within-patient mean change from baseline on the Alzheimer's Disease Assessment Scale-Cognitive subscale was -0.4 points (95% CI, -2.9 to 2.2) in the 100 mL group, while in the 250 mL group it was -0.9 points (95% CI, -3.0 to 1.2). The within-patient mean change from baseline on the Alzheimer's Disease Cooperative Study-Activities of Daily Living was -0.7 points in the 100 mL group (95% CI, -4.3 to 3.0) and -1.3 points (95% CI, -3.4 to 0.7) in the 250 mL group. The mean change from baseline on the Category Fluency Test, Clinical Dementia Rating Scale-Sum of Boxes, Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change, and Neuropsychiatric Inventory Questionnaire was similar for both treatment groups and did not show any worsening. DISCUSSION GRF6019 was safe and well tolerated, and patients experienced no cognitive decline and minimal functional decline. These results support further development of GRF6019.
Collapse
Affiliation(s)
| | | | | | | | - Rebecca Ray
- Alkahest Inc.San CarlosCaliforniaUSA
- Arcus Biosciences (present affiliation)HaywardCaliforniaUSA
| | - Antonio Páez
- Bioscience DivisionGrifols, S.A., Parque Empresarial Can Sant Joan, Avinguda de la GeneralitatBarcelonaSpain
| | - Sam Jackson
- Alkahest Inc.San CarlosCaliforniaUSA
- Alector, Inc. (present affiliation)South San FranciscoCaliforniaUSA
| | | | - Jeffrey L. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las VegasLas VegasNevadaUSA
- Lou Ruvo Center for Brain HealthCleveland ClinicLas VegasNevadaUSA
| | - Gary Kay
- Cognitive Research CorporationSt. PetersburgFloridaUSA
| | | | | |
Collapse
|
61
|
Clinically approved IVIg delivered to the hippocampus with focused ultrasound promotes neurogenesis in a model of Alzheimer's disease. Proc Natl Acad Sci U S A 2020; 117:32691-32700. [PMID: 33288687 DOI: 10.1073/pnas.1908658117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Preclinical and clinical data support the use of focused ultrasound (FUS), in the presence of intravenously injected microbubbles, to safely and transiently increase the permeability of the blood-brain barrier (BBB). FUS-induced BBB permeability has been shown to enhance the bioavailability of administered intravenous therapeutics to the brain. Ideal therapeutics candidates for this mode of delivery are those capable of inducing benefits peripherally following intravenous injection and in the brain at FUS-targeted areas. In Alzheimer's disease, intravenous immunoglobulin (IVIg), a fractionated human blood product containing polyclonal antibodies, act as immunomodulator peripherally and centrally, and it can reduce amyloid pathology in the brain. Using the TgCRND8 mouse model of amyloidosis, we tested whether FUS can improve the delivery of IVIg, administered intravenously (0.4 g/kg), to the hippocampus and reach an effective dose to reduce amyloid plaque pathology and promote neurogenesis. Our results show that FUS-induced BBB permeability is required to deliver a significant amount of IVIg (489 ng/mg) to the targeted hippocampus of TgCRN8 mice. Two IVIg-FUS treatments, administered at days 1 and 8, significantly increased hippocampal neurogenesis by 4-, 3-, and 1.5-fold in comparison to saline, IVIg alone, and FUS alone, respectively. Amyloid plaque pathology was significantly reduced in all treatment groups: IVIg alone, FUS alone, and IVIg-FUS. Putative factors promoting neurogenesis in response to IVIg-FUS include the down-regulation of the proinflammatory cytokine TNF-α in the hippocampus. In summary, FUS was required to deliver an effective dose of IVIg to promote hippocampal neurogenesis and modulate the inflammatory milieu.
Collapse
|
62
|
Lu L, Zheng X, Wang S, Tang C, Zhang Y, Yao G, Zeng J, Ge S, Wen H, Xu M, Guyatt G, Xu N. Anti-Aβ agents for mild to moderate Alzheimer's disease: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2020; 91:1316-1324. [PMID: 33046560 DOI: 10.1136/jnnp-2020-323497] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/01/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To assess the efficacy and safety of Aβ-targeting agents for mild to moderate Alzheimer's disease. METHODS The MEDLINE, Embase, Cochrane Central Register of Controlled Trials, PsycINFO, ClinicalTrials.gov and the WHO's International Clinical Trials Registry Platform search portal were searched from their inception to April 2020. We generated pooled estimates using random effects meta-analyses. RESULTS Nineteen randomised controlled trials, of which 17 had a low risk of bias, included 12 903 participants. The meta-analysis showed no difference in the cognitive subscale of Alzheimer's Disease Assessment Scale (ADAS-Cog) between anti-Aβ drugs and placebo (mean difference (MD): 0.20, 95% CI -0.40 to 0.81; I 2=99.8%; minimal important difference 3.1-3.8 points, moderate-certainty evidence). For ADAS-Cog, results suggested that one drug that increases Aβ clearance may differ in effect (MD: -0.96, 95% CI -0.99 to -0.92) from drugs that reduce Aβ production (MD: 0.78, 95% CI 0.25 to 1.32) (interaction p<0.000001); this difference also existed in the outcome of MMSE and CDR-SOB. Compared with placebo, anti-Aβ drug-related adverse events were as follows: anxiety, depression, diarrhoea, fatigue, rash, syncope and vomit. DISCUSSION From current evidence, anti-Aβ interventions are unlikely to have an important impact on slowing cognitive or functional decline. Although the subgroup analysis suggested possible benefits from Aβ clearance drugs, the analysis has limited credibility, and a benefit from drugs that increase clearance, if real, is very small. TRIAL REGISTRATION NUMBER PROSPERO registration number CRD42019126272.
Collapse
Affiliation(s)
- Liming Lu
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyan Zheng
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengwen Wang
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunzhi Tang
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqing Zhang
- Department of Health Research Methods, Evidence,and Impact, McMaster University, Hamilton, Ontario, Canada.,Guang'anmen Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Gaolei Yao
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingchun Zeng
- Department of Acupuncture, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuqi Ge
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Wen
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingzhu Xu
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Gordon Guyatt
- Department of Health Research Methods, Evidence,and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Nenggui Xu
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
63
|
Elmaleh DR, Farlow MR, Conti PS, Tompkins RG, Kundakovic L, Tanzi RE. Developing Effective Alzheimer's Disease Therapies: Clinical Experience and Future Directions. J Alzheimers Dis 2020; 71:715-732. [PMID: 31476157 PMCID: PMC6839593 DOI: 10.3233/jad-190507] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) clinical trials, focused on disease modifying drugs and conducted in patients with mild to moderate AD, as well as prodromal (early) AD, have failed to reach efficacy endpoints in improving cognitive function in most cases to date or have been terminated due to adverse events. Drugs that have reached clinical stage were reviewed using web resources (such as clinicaltrials.gov, alzforum.org, company press releases, and peer reviewed literature) to identify late stage (Phase II and Phase III) efficacy clinical trials and summarize reasons for their failure. For each drug, only the latest clinical trials and ongoing trials that aimed at improving cognitive function were included in the analysis. Here we highlight the potential reasons that have hindered clinical success, including clinical trial design and choice of outcome measures, heterogeneity of patient populations, difficulties in diagnosing and staging the disease, drug design, mechanism of action, and toxicity related to the long-term use. We review and suggest approaches for AD clinical trial design aimed at improving our ability to identify novel therapies for this devastating disease.
Collapse
Affiliation(s)
- David R Elmaleh
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.,AZTherapies Inc., Boston, MA, USA
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter S Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ronald G Tompkins
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
64
|
Wadhwa R, Paudel KR, Mehta M, Shukla SD, Sunkara K, Prasher P, Panth N, Goyal R, Chellappan DK, Gupta G, Hansbro PM, Aljabali AAA, Tambuwala MM, Dua K. Beyond the Obvious: Smoking and Respiratory Infection Implications on Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:698-708. [PMID: 33109069 DOI: 10.2174/1871527319999200817112427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 11/22/2022]
Abstract
Tobacco smoke is not only a leading cause for chronic obstructive pulmonary disease, cardiovascular disorders, and lung and oral cancers, but also causes neurological disorders such as Alzheimer 's disease. Tobacco smoke consists of more than 4500 toxic chemicals, which form free radicals and can cross blood-brain barrier resulting in oxidative stress, an extracellular amyloid plaque from the aggregation of amyloid β (Aβ) peptide deposition in the brain. Further, respiratory infections such as Chlamydia pneumoniae, respiratory syncytial virus have also been involved in the induction and development of the disease. The necessary information collated on this review has been gathered from various literature published from 1995 to 2019. The review article sheds light on the role of smoking and respiratory infections in causing oxidative stress and neuroinflammation, resulting in Alzheimer's disease (AD). This review will be of interest to scientists and researchers from biological and medical science disciplines, including microbiology, pharmaceutical sciences and the translational researchers, etc. The increasing understanding of the relationship between chronic lung disease and neurological disease is two-fold. First, this would help to identify the risk factors and possible therapeutic interventions to reduce the development and progression of both diseases. Second, this would help to reduce the probable risk of development of AD in the population prone to chronic lung diseases.
Collapse
Affiliation(s)
- Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | - Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Krishna Sunkara
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Nisha Panth
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University, Bajhol, Sultanpur, Solan, Himachal Pradesh, 173 229, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- School of Phamacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | - Alaa A A Aljabali
- Yarmouk University, Irbid 21163, Jordan, Pharmaceutical Sciences, Irbid 21163, Jordan
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County, Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| |
Collapse
|
65
|
Bustos V, Pulina MV, Ledo J. Amyloidogenic and anti-amyloidogenic properties of presenilin 1. ADVANCES IN PHARMACOLOGY 2020; 90:239-251. [PMID: 33706935 DOI: 10.1016/bs.apha.2020.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Presenilin 1 (PS1) is an intramembrane protease, the active subunit of the γ-secretase complex. Its well-studied function is the amyloidogenic cleavage of the C-terminal fragment of the amyloid precursor protein, also known as C99, to produce the Abeta peptide. Recent findings from the Greengard laboratory suggest that PS1 also have anti-amyloidogenic activities, which reduce Abeta levels. First, it redirects APP-C99 toward autophagic degradation, lowering the amount that can be converted into Abeta. The protein kinase CK1γ2 phosphorylates PS1 at Ser367. Phosphorylated PS1 at this position interacts with Annexin A2, which, in turn, interacts with the lysosomal N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) Vamp8. Annexin A2 facilitates the binding of Vamp8 to the autophagosomal SNARE Syntaxin 17 to modulate the fusion of autophagosomes with lysosomes. Thus, PS1 phosphorylated at Ser367 has an anti-amyloidogenic function, promoting autophagosome-lysosome fusion and increasing C99 degradation. Second, it enhances the ability of microglia to phagocyte and degrade extracellular Abeta oligomer, through regulating the expression of the lysosomal master regulator TFEB. Thus, PS1 has a role in both the production and the clearance of Abeta. Drugs designed to activate CK1γ2 and increase the level of PS1 phosphorylated at Ser367 should be useful in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Victor Bustos
- Laboratory of Cellular and Molecular Neuroscience, Rockefeller University, New York, NY, United States.
| | - Maria V Pulina
- Laboratory of Cellular and Molecular Neuroscience, Rockefeller University, New York, NY, United States
| | - Jose Ledo
- Laboratory of Cellular and Molecular Neuroscience, Rockefeller University, New York, NY, United States
| |
Collapse
|
66
|
Cao H, Du X, Zeng R, Lv Z, Ye S, Jiang P, Wang Z, Ma L, Huang Y, Li C, Zhang R, Liu F. Effect of Different Aβ Aggregates as Antigen on the Measure of Naturally Occurring Autoantibodies against Amyloid-β40/42 in IVIG. Curr Alzheimer Res 2020; 16:1290-1299. [PMID: 31894747 DOI: 10.2174/1567205017666200102151731] [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: 02/02/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND The specific Intravenous Immunoglobulin (IVIG) for Alzheimer's Disease (AD) is developing, which contains a high level of naturally occurring autoantibodies against amyloid-β (nAbs-Aβ), and the measure of nAbs-Aβ content is greatly essential. Though Enzyme-Linked Immunosorbent Assay (ELISA) has been widely used in detecting the nAbs-Aβ content, the impact of Aβ aggregates species chosen as antigen in ELISA on this measure has not been evaluated. OBJECTIVE To clarify the influence of different Aβ40/42 aggregates as antigen during ELISA on the content of nAbs-Aβ40/42 measured in IVIG. METHOD Preparation of various Aβ40/42 aggregates was performed by different aggregation solutions and various lengths of time, and analyzed by western blot. Different Aβ40/42 aggregates as antigen were adopted to measure the nAbs-Aβ40/42 content in IVIG by ELISA, and the control was carried out to reduce interference of nonspecific binding. The Bonferroni and Dunnett's T3 were used for statistical analysis. RESULTS The duration for the formation of Aβ40/42 aggregates had more effect on detecting nAbs-Aβ40/42 content in IVIG than the aggregation solution. Higher content of nAbs-Aβ40/42 in the same IVIG was displayed when measured with Aβ40/42 aggregates at day 3, instead of at day 0.5 and day 7.0. The nAbs- Aβ40/42 contents in the same IVIG measured with Aβ40/42 aggregates prepared in different solutions were obviously different, but there was no significant regularity among them. CONCLUSION The nAbs-Aβ40/42 content in the same IVIG is significantly different when measured with Aβ40/42 aggregated under different conditions. The nAbs-Aβ40/42 content in IVIG by antigen-dependent measures, like ELISA, is uncertain.
Collapse
Affiliation(s)
- Haijun Cao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Xi Du
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Renyong Zeng
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Zhaoji Lv
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Shengliang Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Peng Jiang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Zongkui Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Li Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Yun Huang
- School of Materials Science and Engineering, Southwest Petroleum University, Chengdu, China
| | - Changqing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Rong Zhang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Fengjuan Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| |
Collapse
|
67
|
Plotkin SS, Cashman NR. Passive immunotherapies targeting Aβ and tau in Alzheimer's disease. Neurobiol Dis 2020; 144:105010. [PMID: 32682954 PMCID: PMC7365083 DOI: 10.1016/j.nbd.2020.105010] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) and tau proteins currently represent the two most promising targets to treat Alzheimer's disease. The most extensively developed method to treat the pathologic forms of these proteins is through the administration of exogenous antibodies, or passive immunotherapy. In this review, we discuss the molecular-level strategies that researchers are using to design an effective therapeutic antibody, given the challenges in treating this disease. These challenges include selectively targeting a protein that has misfolded or is pathological rather than the more abundant, healthy protein, designing strategic constructs for immunizing an animal to raise an antibody that has the appropriate conformational selectivity to achieve this end, and clearing the pathological protein species before prion-like cell-to-cell spread of misfolded protein has irreparably damaged neurons, without invoking damaging inflammatory responses in the brain that naturally arise when the innate immune system is clearing foreign agents. The various solutions to these problems in current clinical trials will be discussed.
Collapse
Affiliation(s)
- Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC V6T 1Z1, Canada.
| | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
68
|
Imbimbo BP, Lozupone M, Watling M, Panza F. Discontinued disease-modifying therapies for Alzheimer's disease: status and future perspectives. Expert Opin Investig Drugs 2020; 29:919-933. [PMID: 32657175 DOI: 10.1080/13543784.2020.1795127] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the main cause of dementia and represents a huge burden for patients, carers, and healthcare systems. Extensive efforts for over 20 years have failed to find effective disease-modifying drugs. Although amyloid-β (Aβ) accumulation in the brain predicts cognitive decline, effective reduction of plaque load by numerous drug candidates has not yielded significant clinical benefits. A similar pattern is now emerging for drugs which target hyperphosphorylated tau, and trials with anti-inflammatory drugs have been negative despite neuroinflammation appearing to have a crucial role in AD pathogenesis. AREAS COVERED This article reviews key drugs that have been discontinued while in development for AD and delineates the future landscape for present and alternative approaches. EXPERT OPINION Anti-Aβ drugs have failed to validate the Aβ cascade hypothesis of AD. Early findings suggest that the same is happening with therapeutics targeting tau and focussing future research solely on anti-tau drugs is inappropriate. Alternative targets should be pursued, including apolipoprotein E, immunomodulation, plasma exchange, protein autophagy and clearance, mitochondrial dysfunction, abnormal glucose metabolism, neurovascular unit support, epigenetic dysregulation, synaptic loss and dysfunction, microbiota dysbiosis, and combination therapies. Meanwhile, repurposing of drugs approved for other indications is justified where scientific rationale and robust preclinical evidence exist.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici , Parma, Italy
| | - Madia Lozupone
- Unit of Epidemiological Research on Aging "Greatage Study", National Institute of Gastroenterology and Research Hospital IRCCS "S. de Bellis" , Bari, Italy.,Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Partners , Reading, UK
| | - Francesco Panza
- Unit of Epidemiological Research on Aging "Greatage Study", National Institute of Gastroenterology and Research Hospital IRCCS "S. de Bellis" , Bari, Italy
| |
Collapse
|
69
|
Ferl GZ, Fuji RN, Atwal JK, Sun T, Ramanujan S, Quartino AL. Mechanistic Modeling of Soluble Aβ Dynamics and Target Engagement in the Brain by Anti-Aβ mAbs in Alzheimer’s Disease. Curr Alzheimer Res 2020; 17:393-406. [DOI: 10.2174/1567205017666200302122307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/20/2019] [Accepted: 03/01/2020] [Indexed: 02/02/2023]
Abstract
Background:
Anti-amyloid-β (Aβ) monoclonal antibodies (mAbs) are currently in development
for treating Alzheimer’s disease.
Objectives:
To address the complexity of Aβ target engagement profiles, improve the understanding of
crenezumab Pharmacokinetics (PK) and Aβ Pharmacodynamics (PD) in the brain, and facilitate comparison
of anti-Aβ therapies with different binding characteristics.
Methods:
A mechanistic mathematical model was developed describing the distribution, elimination,
and binding kinetics of anti-Aβ mAbs and Aβ (monomeric and oligomeric forms of Aβ1-40 and
Aβ1-42) in the brain, Cerebrospinal Fluid (CSF), and plasma. Physiologically meaningful values were
assigned to the model parameters based on the previous data, with remaining parameters fitted to clinical
measurements of Aβ concentrations in CSF and plasma, and PK/PD data of patients undergoing anti-Aβ
therapy. Aβ target engagement profiles were simulated using a Monte Carlo approach to explore the impact
of biological uncertainty in the model parameters.
Results:
Model-based estimates of in vivo affinity of the antibody to monomeric Aβ were qualitatively
consistent with the previous data. Simulations of Aβ target engagement profiles captured observed mean
and variance of clinical PK/PD data.
Conclusion:
This model is useful for comparing target engagement profiles of different anti-Aβ therapies
and demonstrates that 60 mg/kg crenezumab yields a significant increase in Aβ engagement compared
with lower doses of solanezumab, supporting the selection of 60 mg/kg crenezumab for phase 3
studies. The model also provides evidence that the delivery of sufficient quantities of mAb to brain interstitial
fluid is a limiting step with respect to the magnitude of soluble Aβ oligomer neutralization.
Collapse
Affiliation(s)
- Gregory Z. Ferl
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| | - Reina N. Fuji
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| | - Jasvinder K. Atwal
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| | - Tony Sun
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| | - Saroja Ramanujan
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| | - Angelica L. Quartino
- Department of Translational & Systems Pharmacology, Genentech Research & Early Development, Genentech, Inc., South San Francisco, California, CA 94048, United States
| |
Collapse
|
70
|
Loeffler DA. AMBAR, an Encouraging Alzheimer's Trial That Raises Questions. Front Neurol 2020; 11:459. [PMID: 32547478 PMCID: PMC7272580 DOI: 10.3389/fneur.2020.00459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
Grifols' recent Alzheimer Management by Albumin Replacement (“AMBAR”) study investigated the effects of plasmapheresis with albumin replacement, plus intravenous immunoglobulin (IVIG) in some subjects, in patients with mild-to-moderate Alzheimer's disease (AD). AMBAR was a phase IIb trial in the United States and a phase III trial in Europe. There were three treatment groups (plasmapheresis with albumin replacement; plasmapheresis with low dose albumin and IVIG; plasmapheresis with high dose albumin and IVIG) and sham-treated controls. Disease progression in pooled treated patients was 66% less than control subjects based on ADAS-Cog scores (p = 0.06) and 52% less based on ADCS-ADL scores (p = 0.03). Moderate AD patients had 61% less progression, based on both ADAS-Cog and ADCS-ADL scores, than their sham-treated counterparts (p-values 0.05 and 0.002), and their CDR-Sb scores declined 53% less than their sham-treated counterparts. However, ADAS-Cog and ADCS-ADL scores were not significantly different between actively-treated and sham-treated mild AD patients, although CDR-Sb scores improved vs. baseline for treated mild AD patients. Patients administered both IVIG and albumin had less reduction in brain glucose metabolism than sham-treated patients. Questions raised by these findings include: what mechanism(s) contributed to slowing of disease progression? Is this approach as effective in mild AD as in moderate AD? Must IVIG be included in the protocol? Does age, sex, or ApoE genotype influence treatment response? Does the protocol increase the risk for amyloid-related imaging abnormalities? How long does disease progression remain slowed post-treatment? A further study should allow this approach to be optimized.
Collapse
Affiliation(s)
- David A Loeffler
- Beaumont Research Institute, Department of Neurology, Beaumont Health, Royal Oak, MI, United States
| |
Collapse
|
71
|
Uddin MS, Kabir MT, Mamun AA, Barreto GE, Rashid M, Perveen A, Ashraf GM. Pharmacological approaches to mitigate neuroinflammation in Alzheimer's disease. Int Immunopharmacol 2020; 84:106479. [PMID: 32353686 DOI: 10.1016/j.intimp.2020.106479] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases characterized by the formation of extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs). Growing evidence suggested that there is an association between neuronal dysfunction and neuroinflammation (NI) in AD, coordinated by the chronic activation of astrocytes and microglial cells along with the subsequent excessive generation of the proinflammatory molecule. Therefore, a better understanding of the relationship between the nervous and immune systems is important in order to delay or avert the neurodegenerative events of AD. The inflammatory/immune pathways and the mechanisms to control these pathways may provide a novel arena to develop new drugs in order to target NI in AD. In this review, we represent the influence of cellular mediators which are involved in the NI process, with regards to the progression of AD. We also discuss the processes and the current status of multiple anti-inflammatory agents which are used in AD and have gone through or going through clinical trials. Moreover, new prospects for targeting NI in the development of AD drugs have also been highlighted.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Mamunur Rashid
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Asma Perveen
- School of Life Sciences, The Glocal University, Saharanpur, Uttar Pradesh 247121, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
72
|
Taliyan R, Chandran SK, Kakoty V. Therapeutic Approaches to Alzheimer's Type of Dementia: A Focus on FGF21 Mediated Neuroprotection. Curr Pharm Des 2020; 25:2555-2568. [PMID: 31333086 DOI: 10.2174/1381612825666190716101411] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/08/2019] [Indexed: 12/31/2022]
Abstract
Neurodegenerative disorders are the most devastating disorder of the nervous system. The pathological basis of neurodegeneration is linked with dysfunctional protein trafficking, mitochondrial stress, environmental factors and aging. With the identification of insulin and insulin receptors in some parts of the brain, it has become evident that certain metabolic conditions associated with insulin dysfunction like Type 2 diabetes mellitus (T2DM), dyslipidemia, obesity etc., are also known to contribute to neurodegeneration mainly Alzheimer's Disease (AD). Recently, a member of the fibroblast growth factor (FGF) superfamily, FGF21 has proved tremendous efficacy in diseases like diabetes mellitus, obesity and insulin resistance (IR). Increased levels of FGF21 have been reported to exert multiple beneficial effects in metabolic syndrome. FGF21 receptors are present in certain areas of the brain involved in learning and memory. However, despite extensive research, its function as a neuroprotectant in AD remains elusive. FGF21 is a circulating endocrine hormone which is mainly secreted by the liver primarily in fasting conditions. FGF21 exerts its effects after binding to FGFR1 and co-receptor, β-klotho (KLB). It is involved in regulating energy via glucose and lipid metabolism. It is believed that aberrant FGF21 signalling might account for various anomalies like neurodegeneration, cancer, metabolic dysfunction etc. Hence, this review will majorly focus on FGF21 role as a neuroprotectant and potential metabolic regulator. Moreover, we will also review its potential as an emerging candidate for combating metabolic stress induced neurodegenerative abnormalities.
Collapse
Affiliation(s)
- Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| | - Sarathlal K Chandran
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| | - Violina Kakoty
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| |
Collapse
|
73
|
Ferretti MT, Martinkova J, Biskup E, Benke T, Gialdini G, Nedelska Z, Rauen K, Mantua V, Religa D, Hort J, Santuccione Chadha A, Schmidt R. Sex and gender differences in Alzheimer's disease: current challenges and implications for clinical practice: Position paper of the Dementia and Cognitive Disorders Panel of the European Academy of Neurology. Eur J Neurol 2020; 27:928-943. [PMID: 32056347 DOI: 10.1111/ene.14174] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is characterized by high heterogeneity in disease manifestation, progression and risk factors. High phenotypic variability is currently regarded as one of the largest hurdles in early diagnosis and in the design of clinical trials; there is therefore great interest in identifying factors driving variability that can be used for patient stratification. In addition to genetic and lifestyle factors, the individual's sex and gender are emerging as crucial drivers of phenotypic variability. Evidence exists on sex and gender differences in the rate of cognitive deterioration and brain atrophy, and in the effect of risk factors as well as in the patterns of diagnostic biomarkers. Such evidence might be of high relevance and requires attention in clinical practice and clinical trials. However, sex and gender differences are currently seldom appreciated; importantly, consideration of sex and gender differences is not currently a focus in the design and analysis of clinical trials for AD. The objective of this position paper is (i) to provide an overview of known sex and gender differences that might have implications for clinical practice, (ii) to identify the most important knowledge gaps in the field (with a special regard to clinical trials) and (iii) to provide conclusions for future studies. This scientific statement is endorsed by the European Academy of Neurology.
Collapse
Affiliation(s)
- M T Ferretti
- Institute for Regenerative Medicine - IREM, University of Zurich, Zurich, Switzerland.,Women's Brain Project, Guntershausen, Switzerland
| | - J Martinkova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - E Biskup
- College of Fundamental Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Division of Internal Medicine, University Hospital of Basel, Basel, Switzerland
| | - T Benke
- Neurology Clinic, Medical University Innsbruck, Innsbruck, Austria
| | - G Gialdini
- Neurology - Private Practice, Lucca, Italy
| | - Z Nedelska
- 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
| | - K Rauen
- Institute for Regenerative Medicine - IREM, University of Zurich, Zurich, Switzerland.,Women's Brain Project, Guntershausen, Switzerland.,Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland
| | - V Mantua
- Italian Medicines Agency, Rome, Italy
| | - D Religa
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - J 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
| | - A Santuccione Chadha
- Women's Brain Project, Guntershausen, Switzerland.,Global Medical and Scientific Affairs, Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - R Schmidt
- Department of Neurogeriatrics, University Clinic of Neurology, Medical University Graz, Graz, Austria
| |
Collapse
|
74
|
Du X, Wang Z, Lv Z, Ma L, Ye S, Liu F, Zhang R, Cao H, Li C. Content of anti-β-amyloid 42 oligomers antibodies in multiple batches from different immunoglobulin preparations. Biologicals 2020; 65:25-32. [PMID: 32165080 DOI: 10.1016/j.biologicals.2020.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/19/2020] [Accepted: 02/20/2020] [Indexed: 10/24/2022] Open
Abstract
Immunoglobulin preparations are one of the promising drugs for Alzheimer's disease (AD). Anti-β-amyloid (Aβ) oligomers antibodies in immunoglobulin preparations are considered to be critical for the therapeutic effect against Alzheimer's disease. However, the antibodies content in immunoglobulin preparations varies greatly. In order to determine which factor contributes to the difference of the antibodies content, the content of anti-Aβ oligomers antibodies in multiple batches of immunoglobulin preparations from two manufacturers were measured by enzyme-linked immunosorbent assay. The results showed that no significant difference was found in the antibodies content among different bathes of normal immunoglobulin preparations prepared by the same process from the same manufacturer, whereas significant difference was found in the antibodies content between normal immunoglobulin preparations prepared by ethanol fractionation and those by chromatography process from the same manufacturer. In addition, significant variation existed in the antibodies content between normal immunoglobulin preparations and specific immunoglobulin preparations that are produced by plasma pool of immunized donors. Based on analysis of these results, the preparation process and raw plasma could be the main contributing factors affecting the content of anti-Aβ oligomers antibodies in immunoglobulin preparations. This finding might help to develop AD-specific immunoglobulin preparation containing higher content of anti-Aβ oligomers antibodies.
Collapse
Affiliation(s)
- Xi Du
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Zongkui Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Zhaoji Lv
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Li Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Shengliang Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Fengjuan Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Rong Zhang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Haijun Cao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| | - Changqing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences& Peking Union Medical College, 26 Huacai Road, Chenghua District, Chengdu, 610052, China.
| |
Collapse
|
75
|
Yiannopoulou KG, Papageorgiou SG. Current and Future Treatments in Alzheimer Disease: An Update. J Cent Nerv Syst Dis 2020; 12:1179573520907397. [PMID: 32165850 PMCID: PMC7050025 DOI: 10.1177/1179573520907397] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/24/2020] [Indexed: 01/08/2023] Open
Abstract
Disease-modifying treatment strategies for Alzheimer disease (AD) are still under extensive research. Nowadays, only symptomatic treatments exist for this disease, all trying to counterbalance the neurotransmitter disturbance: 3 cholinesterase inhibitors and memantine. To block the progression of the disease, therapeutic agents are supposed to interfere with the pathogenic steps responsible for the clinical symptoms, classically including the deposition of extracellular amyloid β plaques and intracellular neurofibrillary tangle formation. Other underlying mechanisms are targeted by neuroprotective, anti-inflammatory, growth factor promotive, metabolic efficacious agents and stem cell therapies. Recent therapies have integrated multiple new features such as novel biomarkers, new neuropsychological outcomes, enrollment of earlier populations in the course of the disease, and innovative trial designs. In the near future different specific agents for every patient might be used in a “precision medicine” context, where aberrant biomarkers accompanied with a particular pattern of neuropsychological and neuroimaging findings could determine a specific treatment regimen within a customized therapeutic framework. In this review, we discuss potential disease-modifying therapies that are currently being studied and potential individualized therapeutic frameworks that can be proved beneficial for patients with AD.
Collapse
Affiliation(s)
| | - Sokratis G Papageorgiou
- Cognitive Disorders/Dementia Unit, 2nd Neurological Department, National and Kapodistrian University of Athens, Attikon General University Hospital, Athens, Greece
| |
Collapse
|
76
|
Frontzek K, Carta M, Losa M, Epskamp M, Meisl G, Anane A, Brandel JP, Camenisch U, Castilla J, Haïk S, Knowles T, Lindner E, Lutterotti A, Minikel EV, Roiter I, Safar JG, Sanchez-Valle R, Žáková D, Hornemann S, Aguzzi A. Autoantibodies against the prion protein in individuals with PRNP mutations. Neurology 2020; 95:e2028-e2037. [PMID: 32098855 DOI: 10.1212/wnl.0000000000009183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine whether naturally occurring autoantibodies against the prion protein are present in individuals with genetic prion disease mutations and controls, and if so, whether they are protective against prion disease. METHODS In this case-control study, we collected 124 blood samples from individuals with a variety of pathogenic PRNP mutations and 78 control individuals with a positive family history of genetic prion disease but lacking disease-associated PRNP mutations. Antibody reactivity was measured using an indirect ELISA for the detection of human immunoglobulin G1-4 antibodies against wild-type human prion protein. Multivariate linear regression models were constructed to analyze differences in autoantibody reactivity between (1) PRNP mutation carriers vs controls and (2) asymptomatic vs symptomatic PRNP mutation carriers. Robustness of results was examined in matched cohorts. RESULTS We found that antibody reactivity was present in a subset of both PRNP mutation carriers and controls. Autoantibody levels were not influenced by PRNP mutation status or clinical manifestation of prion disease. Post hoc analyses showed anti-PrPC autoantibody titers to be independent of personal history of autoimmune disease and other immunologic disorders, as well as PRNP codon 129 polymorphism. CONCLUSIONS Pathogenic PRNP variants do not notably stimulate antibody-mediated anti-PrPC immunity. Anti-PrPC immunoglobulin G autoantibodies are not associated with the onset of prion disease. The presence of anti-PrPC autoantibodies in the general population without any disease-specific association suggests that relatively high titers of naturally occurring antibodies are well-tolerated. CLINICALTRIALSGOV IDENTIFIER NCT02837705.
Collapse
Affiliation(s)
- Karl Frontzek
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | - Manfredi Carta
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Marco Losa
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Mirka Epskamp
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Georg Meisl
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Alice Anane
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jean-Philippe Brandel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ulrike Camenisch
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Joaquín Castilla
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Stéphane Haïk
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Tuomas Knowles
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ewald Lindner
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Andreas Lutterotti
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Eric Vallabh Minikel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ignazio Roiter
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jiri G Safar
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Raquel Sanchez-Valle
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Dana Žáková
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Simone Hornemann
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Adriano Aguzzi
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | | |
Collapse
|
77
|
Similarity of therapeutic networks induced by a multi-component herbal remedy, Ukgansan, in neurovascular unit cells. Sci Rep 2020; 10:2658. [PMID: 32060346 PMCID: PMC7021700 DOI: 10.1038/s41598-020-59537-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/27/2020] [Indexed: 12/17/2022] Open
Abstract
The neurovascular unit, which includes neurons, glial cells, and vascular cells, plays crucial roles in the onset and progression of Alzheimer’s disease (AD). Therefore, effective drugs against AD should be able to target the multi-cellular neurovascular unit and the therapeutic relationships among neurovascular cells should be defined. Here, we examined the therapeutic effects of Ukgansan (UGS), an herbal remedy with multi-targeting capabilities, using in vitro neurovascular unit models and an in vivo model of AD. In addition, we compared the therapeutic networks induced by UGS and its components in different neurovascular cell types. We found that UGS and its components protected neurovascular cells against diverse damaging agents and improved the behavioral patterns of AD model mice. A comparison of UGS- or its components-induced therapeutic networks, constructed from high-throughput data on gene expression, pathway activity, and protein phosphorylation, revealed similarities among neurovascular cell types, especially between BV-2 microglia and HBVP (human brain vascular pericytes). These findings, together with the functional connections between neurovascular cells, can explain the therapeutic effects of UGS. Furthermore, they suggest underlying similarities in the therapeutic mechanisms in different neurovascular cell types.
Collapse
|
78
|
Negi N, Das BK. Decoding intrathecal immunoglobulins and B cells in the CNS: their synthesis, function, and regulation. Int Rev Immunol 2020; 39:67-79. [PMID: 31928379 DOI: 10.1080/08830185.2019.1711073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The discovery of an active lymphatic system in the meninges (dura mater) has opened up a wide range of possibilities for the role of CNS immunoglobulins in brain development in early fetal life or during infancy. The antibody-dependent and -independent functions of B cells in the immunopathogenesis of multiple sclerosis are not new to immunologists, yet their role in other neurodegenerative disorders such as Alzheimer's and Parkinson's disease is incompletely understood. Deep cervical lymph nodes have emerged as a candidate site for autosensitization against CNS antigens and have been shown to provide the right kind of milieu for the dynamic interaction of antigen-presenting cells, B cells, and T cells. The presence of different B cells in the lymph nodes and the production of natural autoantibodies by B-1 cells have definitely unlocked another piece of the puzzle. At a time when CD19 and CD20 monoclonal antibodies have shown remarkable results in ameliorating the relapse and progression of multiple sclerosis, it is imperative to dissect out the diversity in B cell populations inside the CNS to identify new targets to improve current treatment regimens for neurodegenerative diseases. This review highlights the origin, migration, function, and regulation of B cells and the production of intrathecal immunoglobulins considering the previous and current findings and taking into account the differences between a healthy state and the changes that occur during an inflammatory or autoimmune response.
Collapse
Affiliation(s)
- Neema Negi
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland, Galway, Ireland
| | - Bimal K Das
- HIV Immunology Section, Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
79
|
Kile S, Au W, Parise C, Sohi J, Yarbrough T, Czeszynski A, Johnson K, Redline D, Donnel T, Hankins A, Rose K. Reduction of Amyloid in the Brain and Retina After Treatment With IVIG for Mild Cognitive Impairment. Am J Alzheimers Dis Other Demen 2020; 35:1533317519899800. [PMID: 32048858 PMCID: PMC10624008 DOI: 10.1177/1533317519899800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To assess whether intravenous immunoglobulin (IVIG) in subjects with mild cognitive impairment (MCI) results in a reduction in amyloid in the central nervous system (CNS). METHODS Five subjects with MCI underwent baseline Florbetapir positron emission tomography and retinal autofluorescent imaging. All were administered IVIG (Octagam 10%) at 0.4 g/kg every 14 days for a total of 5 infusions. After 3 months, standard uptake value ratio (SUVR) and amyloid retinal deposits were reassessed. RESULTS Three subjects had a reduction in amyloid SUVR and all 5 subjects had a reduction in amyloid retinal deposits in at least 1 eye. CONCLUSIONS A short course of IVIG over 2 months removes a measurable amount of amyloid from the CNS in persons with MCI.
Collapse
Affiliation(s)
- Shawn Kile
- Sutter Neuroscience Institute, Sacramento, CA, USA
| | - William Au
- Sutter Neuroscience Institute, Sacramento, CA, USA
| | - Carol Parise
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | - Jaideep Sohi
- Northern California PET Imaging Center, Sacramento, CA, USA
| | | | | | | | | | - Tammy Donnel
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | - Andrea Hankins
- Sutter Institute for Medical Research (SIMR), Sacramento, CA, USA
| | | |
Collapse
|
80
|
Yiannopoulou KG, Anastasiou AI, Zachariou V, Pelidou SH. Reasons for Failed Trials of Disease-Modifying Treatments for Alzheimer Disease and Their Contribution in Recent Research. Biomedicines 2019; 7:biomedicines7040097. [PMID: 31835422 PMCID: PMC6966425 DOI: 10.3390/biomedicines7040097] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Despite all scientific efforts and many protracted and expensive clinical trials, no new drug has been approved by FDA for treatment of Alzheimer disease (AD) since 2003. Indeed, more than 200 investigational programs have failed or have been abandoned in the last decade. The most probable explanations for failures of disease-modifying treatments (DMTs) for AD may include late initiation of treatments during the course of AD development, inappropriate drug dosages, erroneous selection of treatment targets, and mainly an inadequate understanding of the complex pathophysiology of AD, which may necessitate combination treatments rather than monotherapy. Clinical trials’ methodological issues have also been criticized. Drug-development research for AD is aimed to overcome these drawbacks. Preclinical and prodromal AD populations, as well as traditionally investigated populations representing all the clinical stages of AD, are included in recent trials. Systematic use of biomarkers in staging preclinical and prodromal AD and of a single primary outcome in trials of prodromal AD are regularly integrated. The application of amyloid, tau, and neurodegeneration biomarkers, including new biomarkers—such as Tau positron emission tomography, neurofilament light chain (blood and Cerebrospinal fluid (CSF) biomarker of axonal degeneration) and neurogranin (CSF biomarker of synaptic functioning)—to clinical trials allows more precise staging of AD. Additionally, use of Bayesian statistics, modifiable clinical trial designs, and clinical trial simulators enrich the trial methodology. Besides, combination therapy regimens are assessed in clinical trials. The above-mentioned diagnostic and statistical advances, which have been recently integrated in clinical trials, are relevant to the recent failures of studies of disease-modifying treatments. Their experiential rather than theoretical origins may better equip potentially successful drug-development strategies.
Collapse
Affiliation(s)
- Konstantina G. Yiannopoulou
- Memory Center, Neurological Department, Henry Dunant Hospital Center, 107 Mesogeion Avenue, 11526 Athens, Greece
- Correspondence:
| | | | - Venetia Zachariou
- Icahn School of Medicine at Mount Sinai, Nash family Department of Neurosciences, Department of Pharmacological Sciences, and Friedman Brain Institute, New York, NY 11004, USA;
| | - Sygkliti-Henrietta Pelidou
- Department of Neurology, University of Ioannina, University Hospital of Ioannina, 45500 Ioannina, Greece;
| |
Collapse
|
81
|
B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci 2019; 20:728-745. [PMID: 31712781 DOI: 10.1038/s41583-019-0233-2] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
|
82
|
Schneider LS, Thomas RG, Hendrix S, Rissman RA, Brewer JB, Salmon DP, Oltersdorf T, Okuda T, Feldman HH. Safety and Efficacy of Edonerpic Maleate for Patients With Mild to Moderate Alzheimer Disease: A Phase 2 Randomized Clinical Trial. JAMA Neurol 2019; 76:1330-1339. [PMID: 31282954 DOI: 10.1001/jamaneurol.2019.1868] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Edonerpic maleate (T-817MA) protects against Aβ40-induced neurotoxic effects and memory deficits, promotes neurite outgrowth, and preserves hippocampal synapses and spatial memory in tau transgenic mice. These effects may be mediated via sigma-1 receptor activation, delivery of synaptic AMPA receptors, or modulation of microglial function and may benefit patients with Alzheimer disease. Objective To assess the efficacy, safety, and tolerability of edonerpic for patients with mild to moderate Alzheimer disease. Design, Setting, and Participants Randomized, double-blind, placebo-controlled, parallel-group, phase 2 clinical trial conducted over 52 weeks from June 2, 2014, to December 14, 2016, at 52 US clinical and academic centers. Of 822 outpatients screened, 484 met the following criteria and were randomly assigned to treatment: 55 to 85 years of age, probable Alzheimer disease, Mini-Mental State Examination scores from 12 to 22, and taking stable doses of donepezil or rivastigmine with or without memantine. Interventions Random assignment (1:1:1 allocation) to placebo or 224 mg or 448 mg of edonerpic maleate, once per day. Main Outcomes and Measures Coprimary outcomes were scores on the Alzheimer Disease Assessment Scale-Cognitive Subscale (ADAS-cog) and Alzheimer's Disease Cooperative Study-Clinical Impression of Change (ADCS-CGIC) at week 52. Biomarkers were brain, lateral ventricular, and hippocampal volumes, as determined on magnetic resonance imaging, and cerebrospinal fluid Aβ40, Aβ42, total tau, and phospho-tau181. The primary efficacy analysis was performed on the coprimary end points for the modified intention-to-treat population. Results Of 482 participants in the safety population, 140 of 158 participants (88.6%) assigned to placebo, 117 of 166 participants (70.5%) to 224 mg of edonerpic maleate, and 120 of 158 participants (76.0%) to 448 mg of edonerpic maleate completed the trial. The mean ADAS-cog score change at week 52 was 7.91 for the placebo group, 7.45 for the 224-mg group, and 7.08 for the 448-mg group. Mean differences from placebo were -0.47 (95% CI, -2.36 to 1.43; P = .63) for the 224-mg group and -0.84 (95% CI, -2.75 to 1.08; P = .39) for the 448-mg group. Mean ADCS-CGIC scores were 5.22 for the placebo group, 5.24 for the 224-mg group, and 5.25 for the 448-mg group, with mean differences from placebo of 0.03 (95% CI, -0.20 to 0.25; P = .81) for the 224-mg group and 0.04 (95% CI, -0.19 to 0.26; P = .76) for the 448-mg group. In the safety population, a total of 7 of 158 participants (4.4%) in the placebo group, 23 of 166 participants (13.9%) in the 224-mg group, and 23 of 158 participants (14.6%) in the 448-mg group discontinued because of adverse events. The most frequent adverse events were diarrhea and vomiting. Conclusions and Relevance Edonerpic maleate appeared to be safe and tolerable, with expected gastrointestinal symptoms occurring early but without evidence for a clinical effect among patients with mild to moderate Alzheimer disease. Trial Registration ClinicalTrials.gov identifier: NCT02079909.
Collapse
Affiliation(s)
- Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles
| | - Ronald G Thomas
- Department of Neurosciences, University of California San Diego School of Medicine
| | | | - Robert A Rissman
- Department of Neurosciences, University of California San Diego School of Medicine
| | - James B Brewer
- Department of Neurosciences, University of California San Diego School of Medicine
| | - David P Salmon
- Department of Neurosciences, University of California San Diego School of Medicine
| | - Tilman Oltersdorf
- Department of Neurosciences, University of California San Diego School of Medicine
| | - Tomohiro Okuda
- Development Division, FUJIFILM Toyama Chemical Co, Ltd, Tokyo, Japan
| | - Howard H Feldman
- Department of Neurosciences, University of California San Diego School of Medicine
| | | |
Collapse
|
83
|
Long JM, Holtzman DM. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019; 179:312-339. [PMID: 31564456 PMCID: PMC6778042 DOI: 10.1016/j.cell.2019.09.001] [Citation(s) in RCA: 1849] [Impact Index Per Article: 308.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer disease (AD) is a heterogeneous disease with a complex pathobiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyperphosphorylated tau as neurofibrillary tangles remains the primary neuropathologic criteria for AD diagnosis. However, a number of recent fundamental discoveries highlight important pathological roles for other critical cellular and molecular processes. Despite this, no disease-modifying treatment currently exists, and numerous phase 3 clinical trials have failed to demonstrate benefits. Here, we review recent advances in our understanding of AD pathobiology and discuss current treatment strategies, highlighting recent clinical trials and opportunities for developing future disease-modifying therapies.
Collapse
Affiliation(s)
- Justin M Long
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
84
|
Loera-Valencia R, Cedazo-Minguez A, Kenigsberg PA, Page G, Duarte AI, Giusti P, Zusso M, Robert P, Frisoni GB, Cattaneo A, Zille M, Boltze J, Cartier N, Buee L, Johansson G, Winblad B. Current and emerging avenues for Alzheimer's disease drug targets. J Intern Med 2019; 286:398-437. [PMID: 31286586 DOI: 10.1111/joim.12959] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD), the most frequent cause of dementia, is escalating as a global epidemic, and so far, there is neither cure nor treatment to alter its progression. The most important feature of the disease is neuronal death and loss of cognitive functions, caused probably from several pathological processes in the brain. The main neuropathological features of AD are widely described as amyloid beta (Aβ) plaques and neurofibrillary tangles of the aggregated protein tau, which contribute to the disease. Nevertheless, AD brains suffer from a variety of alterations in function, such as energy metabolism, inflammation and synaptic activity. The latest decades have seen an explosion of genes and molecules that can be employed as targets aiming to improve brain physiology, which can result in preventive strategies for AD. Moreover, therapeutics using these targets can help AD brains to sustain function during the development of AD pathology. Here, we review broadly recent information for potential targets that can modify AD through diverse pharmacological and nonpharmacological approaches including gene therapy. We propose that AD could be tackled not only using combination therapies including Aβ and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood-brain barrier targets that have been studied recently. We also make a case for the role of gut microbiota in AD. Our hope is to promote the continuing research of diverse targets affecting AD and promote diverse targeting as a near-future strategy.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - A Cedazo-Minguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | | | - G Page
- Neurovascular Unit and Cognitive impairments - EA3808, University of Poitiers, Poitiers, France
| | - A I Duarte
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - P Giusti
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - M Zusso
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - P Robert
- CoBTeK - lab, CHU Nice University Côte d'Azur, Nice, France
| | - G B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - A Cattaneo
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - M Zille
- Institute of Experimental and Clinical Pharmacology and Toxicology, Lübeck, Germany
| | - J Boltze
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - N Cartier
- Preclinical research platform, INSERM U1169/MIRCen Commissariat à l'énergie atomique, Fontenay aux Roses, France.,Université Paris-Sud, Orsay, France
| | - L Buee
- Alzheimer & Tauopathies, LabEx DISTALZ, CHU-Lille, Inserm, Univ. Lille, Lille, France
| | - G Johansson
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
85
|
Kisby B, Jarrell JT, Agar ME, Cohen DS, Rosin ER, Cahill CM, Rogers JT, Huang X. Alzheimer's Disease and Its Potential Alternative Therapeutics. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2019; 9. [PMID: 31588368 PMCID: PMC6777730 DOI: 10.4172/2161-0460.1000477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer’s Disease (AD) is a chronic neurodegenerative disease that affects over 5 million individuals in the United States alone. Currently, there are only two kinds of pharmacological interventions available for symptomatic relief of AD; Acetyl Cholinesterase Inhibitors (AChEI) and N-methyl-D-aspartic Acid (NMDA) receptor antagonists and these drugs do not slow down or stop the progression of the disease. Several molecular targets have been implicated in the pathophysiology of AD, such as the tau (τ) protein, Amyloid-beta (Aβ), the Amyloid Precursor Protein (APP) and more and several responses have also been observed in the advancement of the disease, such as reduced neurogenesis, neuroinflammation, oxidative stress and iron overload. In this review, we discuss general features of AD and several small molecules across different experimental AD drug classes that have been studied for their effects in the context of the molecular targets and responses associated with the AD progression. These drugs include: Paroxetine, Desferrioxamine (DFO), N-acetylcysteine (NAC), Posiphen/-(−)Phenserine, JTR-009, Carvedilol, LY450139, Intravenous immunoglobulin G 10%, Indomethacin and Lithium Carbonate (Li2CO3).
Collapse
Affiliation(s)
- Brent Kisby
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Juliet T Jarrell
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - M Enes Agar
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - David S Cohen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eric R Rosin
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Catherine M Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
86
|
Chen Y, Wang C, Xu F, Ming F, Zhang H. Efficacy and Tolerability of Intravenous Immunoglobulin and Subcutaneous Immunoglobulin in Neurologic Diseases. Clin Ther 2019; 41:2112-2136. [PMID: 31445679 DOI: 10.1016/j.clinthera.2019.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/01/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE IV immunoglobulin (Ig) therapy has been widely used for the treatment of neurologic disorders, autoimmune diseases, immunodeficiency-related diseases, blood system diseases, and cancers. In this review, we summarize the efficacy and tolerability of IVIg and SCIg therapy in neurologic diseases. METHODS We summarized and analyzed the efficacy and tolerability of IVIg and SCIg in neurologic diseases, by analyzing the literature pertaining to the use of IVIg and SCIg to treat nervous system diseases. FINDINGS In clinical neurology practice, IVIg has been shown to be useful for the treatment of new-onset or recurrent immune diseases and for long-term maintenance treatment of chronic diseases. Moreover, IVIg may have applications in the management of intractable autoimmune epilepsy, paraneoplastic syndrome, autoimmune encephalitis, and neuromyelitis optica. SCIg is emerging as an alternative to IVIg treatment. Although SCIg has a composition similar to that of IVIg, the applications of this therapy are different. Notably, the bioavailability of SCIg is lower than that of IVIg, but the homeostasis level is more stable. Current studies have shown that these 2 therapies have pharmacodynamic equivalence. IMPLICATIONS In this review, we explored the efficacy of IVIg in the treatment of various neurologic disorders. IVIg administration still faces many challenges. Thus, it will be necessary to standardize the use of IVIg in the clinical setting. SCIg administration is a novel and feasible treatment option for neurologic and immune-related diseases, such as chronic inflammatory demyelinating polyradiculoneuropathy and idiopathic inflammatory myopathies. As our understanding of the mechanisms of action of IVIg improve, potential next-generation biologics can being developed.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunyu Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fanxi Xu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fengyu Ming
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hainan Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
87
|
Tau-Reactive Endogenous Antibodies: Origin, Functionality, and Implications for the Pathophysiology of Alzheimer's Disease. J Immunol Res 2019; 2019:7406810. [PMID: 31687413 PMCID: PMC6811779 DOI: 10.1155/2019/7406810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/19/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
In Alzheimer's disease (AD), tau pathology manifested by the accumulation of intraneuronal tangles and soluble toxic oligomers emerges as a promising therapeutic target. Multiple anti-tau antibodies inhibiting the formation and propagation of cytotoxic tau or promoting its clearance and degradation have been tested in clinical trials, albeit with the inconclusive outcome. Antibodies against tau protein have been documented both in the brain circulatory system and at the periphery, but their origin and role under normal conditions and in AD remain unclear. While it is tempting to assign them a protective role in regulating tau level and removal of toxic variants, the supportive evidence remains sporadic, requiring systematic analysis and critical evaluation. Herein, we review recent data showing the occurrence of tau-reactive antibodies in the brain and peripheral circulation and discuss their origin and significance in tau clearance. Based on the emerging evidence, we cautiously propose that impairments of tau clearance at the periphery by humoral immunity might aggravate the tau pathology in the central nervous system, with implication for the neurodegenerative process of AD.
Collapse
|
88
|
Paranjpe MD, Taubes A, Sirota M. Insights into Computational Drug Repurposing for Neurodegenerative Disease. Trends Pharmacol Sci 2019; 40:565-576. [PMID: 31326236 PMCID: PMC6771436 DOI: 10.1016/j.tips.2019.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022]
Abstract
Computational drug repurposing has the ability to remarkably reduce drug development time and cost in an era where these factors are prohibitively high. Several examples of successful repurposed drugs exist in fields such as oncology, diabetes, leprosy, inflammatory bowel disease, among others, however computational drug repurposing in neurodegenerative disease has presented several unique challenges stemming from the lack of validation methods and difficulty in studying heterogenous diseases of aging. Here, we examine existing approaches to computational drug repurposing, including molecular, clinical, and biophysical methods, and propose data sources and methods to advance computational drug repurposing in neurodegenerative disease using Alzheimer's disease as an example.
Collapse
Affiliation(s)
- Manish D Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA.
| | - Alice Taubes
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA; Gladstone Institutes, San Francisco, CA 94158, USA.
| |
Collapse
|
89
|
Salloway SP, Sperling R, Fox NC, Sabbagh MN, Honig LS, Porsteinsson AP, Rofael H, Ketter N, Wang D, Liu E, Carr S, Black RS, Brashear HR. Long-Term Follow Up of Patients with Mild-to-Moderate Alzheimer's Disease Treated with Bapineuzumab in a Phase III, Open-Label, Extension Study. J Alzheimers Dis 2019; 64:689-707. [PMID: 29914022 DOI: 10.3233/jad-171157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND A 3-year extension of two Phase III parent studies of intravenous (IV) bapineuzumab in patients with mild-to-moderate Alzheimer's disease dementia (apolipoprotein (APOE) ɛ4 carriers and noncarriers) is summarized. OBJECTIVES The primary and secondary objectives were to evaluate the long-term safety, tolerability, and maintenance of efficacy of bapineuzumab. METHODS A multicenter study in patients who had participated in double-blind placebo-controlled parent studies. Patients enrolled in the extension study were assigned to receive IV infusions of bapineuzumab (0.5 or 1.0 mg/kg) every 13 weeks until termination but were blinded to whether they had received bapineuzumab or placebo in the parent studies. RESULTS A total of 1,462 (688 were APOEɛ4 carriers and 774 were noncarriers) patients were enrolled. Extension-onset adverse events occurred in >81% of the patients in each dose group. Fall, urinary tract infection, agitation, and ARIA-E occurred in ≥10% of participants. The incidence proportion of ARIA-E was higher among carriers and noncarriers who received bapineuzumab for the first time in the extension study (11.8% and 5.4%, respectively) versus those who were previously exposed in the parent studies (5.1% and 1.3%, respectively). After 6 to 12 months exposure to bapineuzumab IV in the extension study, similar deterioration of cognition and function occurred with no significant differences between the dose groups. CONCLUSIONS Infusion of bapineuzumab 0.5 or 1.0 mg/kg every 13 weeks for up to 3 years was generally well tolerated, with a safety and tolerability profile similar to that in previous studies.
Collapse
Affiliation(s)
| | - Reisa Sperling
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, MA, USA
| | - Nick C Fox
- Dementia Research Centre, University College London, Institute of Neurology, London, UK
| | | | | | | | - Hany Rofael
- Janssen Alzheimer Immunotherapy Research & Development, LLC, South San Francisco, CA, USA
| | - Nzeera Ketter
- Janssen Alzheimer Immunotherapy Research & Development, LLC, South San Francisco, CA, USA
| | - Daniel Wang
- Janssen Alzheimer Immunotherapy Research & Development, LLC, South San Francisco, CA, USA
| | - Enchi Liu
- Janssen Alzheimer Immunotherapy Research & Development, LLC, South San Francisco, CA, USA
| | - Stephen Carr
- Janssen Alzheimer Immunotherapy Research & Development, LLC, South San Francisco, CA, USA
| | | | | |
Collapse
|
90
|
Chantran Y, Capron J, Alamowitch S, Aucouturier P. Anti-Aβ Antibodies and Cerebral Amyloid Angiopathy Complications. Front Immunol 2019; 10:1534. [PMID: 31333665 PMCID: PMC6620823 DOI: 10.3389/fimmu.2019.01534] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/19/2019] [Indexed: 11/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) corresponds to the deposition of amyloid material in the cerebral vasculature, leading to structural modifications of blood vessel walls. The most frequent form of sporadic CAA involves fibrillar β-amyloid peptide (Aβ) deposits, mainly the 40 amino acid form (Aβ1-40), which are commonly found in the elderly with or without Alzheimer's disease. Sporadic CAA usually remains clinically silent. However, in some cases, acute complications either hemorrhagic or inflammatory can occur. Similar complications occurred after active or passive immunization against Aβ in experimental animal models exhibiting CAA, and in subjects with Alzheimer's disease during clinical trials. The triggering of these adverse events by active immunization and monoclonal antibody administration in CAA-bearing individuals suggests that analogous mechanisms could be involved during spontaneous CAA complications, drawing particular attention to the role of anti-Aβ antibodies. However, antibodies that react with several monomeric and aggregated forms of Aβ spontaneously occur in virtually all human individuals, hence being part of the "natural antibody" repertoire. Natural antibodies are usually described as having low-affinity and high cross-reactivity toward microbial components and autoantigens. Although frequently of the IgM class, they also belong to IgG and IgA isotypes. They likely display homeostatic functions and protective roles in aging. Until recently, the peculiar properties of these natural antibodies have hindered proper analysis of the Aβ-reactive antibody repertoire and the study of their implication in CAA complications. Herein, we review and comment the evidences of an auto-immune nature of spontaneous CAA complications, and discuss implications for forthcoming research and clinical practice.
Collapse
Affiliation(s)
- Yannick Chantran
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département d'Immunologie Biologique, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jean Capron
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département de Neurologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Sonia Alamowitch
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département de Neurologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Pierre Aucouturier
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département d'Immunologie Biologique, Hôpital Saint-Antoine, AP-HP, Paris, France
| |
Collapse
|
91
|
The amyloid cascade and Alzheimer's disease therapeutics: theory versus observation. J Transl Med 2019; 99:958-970. [PMID: 30760863 DOI: 10.1038/s41374-019-0231-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/31/2022] Open
Abstract
The identification of amyloid-β precursor protein (APP) pathogenic mutations in familial early onset Alzheimer's disease (AD), along with knowledge that amyloid-β (Aβ) was the principle protein component of senile plaques, led to the establishment of the amyloid cascade hypothesis. Down syndrome substantiated the hypothesis, given an extra copy of the APP gene and invariable AD pathology hallmarks that occur by middle age. An abundance of support for the amyloid cascade hypothesis followed. Prion-like protein misfolding and non-Mendelian transmission of neurotoxicity are among recent areas of investigation. Aβ-targeted clinical trials have been disappointing, with negative results attributed to inadequacies in patient selection, challenges in pharmacology, and incomplete knowledge of the most appropriate target. There is evidence, however, that proof of concept has been achieved, i.e., clearance of Aβ during life, but with no significant changes in cognitive trajectory in AD. Whether the time, effort, and expense of Aβ-targeted therapy will prove valuable will be determined over time, as Aβ-centered clinical trials continue to dominate therapeutic strategies. It seems reasonable to hypothesize that the amyloid cascade is intimately involved in AD, in parallel with disease pathogenesis, but that removal of toxic Aβ is insufficient for an effective disease modification.
Collapse
|
92
|
Lin CH, Fann JCY, Chen SLS, Chen HH, Yang KC. Heterogeneity in Cost-Effectiveness Analysis of Vaccination for Mild and Moderate Alzheimer's Disease. Curr Alzheimer Res 2019; 16:495-504. [PMID: 31195946 PMCID: PMC6791033 DOI: 10.2174/1567205016666190612162121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 04/04/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Immunotherapy for Alzheimer's disease(AD) has gained momentum in recent years. One of the concerns over its application pertains to Cost-Effectiveness Analysis (CEA) from population average and specific subgroup differences, as such a therapy is imperative for health decisionmakers to allocate limited resources. However, this sort of CEA model considering heterogeneous population with risk factors adjustment has been rarely addressed. METHODS We aimed to show the heterogeneity of CEA in immunotherapy for AD in comparison with the comparator without intervention. Economic evaluation was performed via incremental Cost- Effectiveness Ratio (ICER) and Cost-Effectiveness Acceptability Curve (CEAC) in terms of the Quality- Adjusted Life Years (QALY). First, population-average CEA was performed with and without adjustment for age and gender. Secondly, sub-group CEA was performed with the stratification of gender and age based on Markov process. RESULTS Given the threshold of $20,000 of willingness to pay, the results of ICER without and with adjustment for age and gender revealed similar results ($14,691/QALY and $17,604/QALY). The subgroup ICER results by different age groups and gender showed substantial differences. The CEAC showed that the probability of being cost-effective was only 48.8%-53.3% in terms of QALY at population level but varied from 83.5% in women aged 50-64 years, following women aged 65-74 years and decreased to 0.2% in men≥ 75 years. CONCLUSION There were considerable heterogeneities observed in the CEA of vaccination for AD. As with the development of personalized medicine, the CEA results assessed by health decision-maker should not only be considered by population-average level but also specific sub-group levels.
Collapse
Affiliation(s)
- Chung-Hsien Lin
- Division of New Drug, Center for Drug Evaluation, Taipei, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Jean Ching-Yuan Fann
- Department of Health Industry Management, School of Healthcare Management, Kainan University, Tao-Yuan, Taiwan
| | - Sam Li-Sheng Chen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Hsi Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Kuen-Cheh Yang
- Department of Family Medicine, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan.,Community and Research Center, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| |
Collapse
|
93
|
Liu J, Wang LN. Intravenous immunoglobulins for Alzheimer’s disease and mild cognitive impairment due to Alzheimer’s disease: A systematic review with meta-analysis. Expert Rev Neurother 2019; 19:475-480. [PMID: 31092051 DOI: 10.1080/14737175.2019.1620106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jia Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lu-Ning Wang
- Department of Geriatric Neurology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
94
|
Manolopoulos A, Andreadis P, Malandris K, Avgerinos I, Karagiannis T, Kapogiannis D, Tsolaki M, Tsapas A, Bekiari E. Intravenous Immunoglobulin for Patients With Alzheimer's Disease: A Systematic Review and Meta-Analysis. Am J Alzheimers Dis Other Demen 2019; 34:281-289. [PMID: 30987435 DOI: 10.1177/1533317519843720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM To assess the efficacy and safety of intravenous immunoglobulin (IVIg) for patients with Alzheimer's disease (AD). MATERIALS AND METHODS We searched electronic databases and other sources for randomized controlled trials comparing IVIg with placebo or other treatment for adults with AD. Primary outcome was change from baseline in Alzheimer's Disease Assessment Scale-Cognitive subscale (ADAS-Cog). RESULTS Five placebo-controlled trials were included in the meta-analysis. Compared to placebo, IVIg 0.2 and 0.4 g/kg once every two weeks did not change ADAS-Cog score (weighted mean difference: 0.37, 95% confidence interval: -1.46 to 2.20 and 0.77, -1.34 to 2.88, respectively). Furthermore, except for an increase in the incidence of rash, IVIg did not affect the incidence of other adverse events. CONCLUSION IVIg, albeit safe, is inefficacious for treatment of patients with AD. Future trials targeting earlier stages of disease or applying different dosing regimens may be warranted to clarify its therapeutic potential.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Panagiotis Andreadis
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Konstantinos Malandris
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Ioannis Avgerinos
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Thomas Karagiannis
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Dimitrios Kapogiannis
- 2 Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Magda Tsolaki
- 3 First Department of Neurology, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Apostolos Tsapas
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece.,4 Harris Manchester College, University of Oxford, Oxford, United Kingdom
| | - Eleni Bekiari
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| |
Collapse
|
95
|
Boada M, López O, Núñez L, Szczepiorkowski ZM, Torres M, Grifols C, Páez A. Plasma exchange for Alzheimer's disease Management by Albumin Replacement (AMBAR) trial: Study design and progress. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:61-69. [PMID: 30859122 PMCID: PMC6395854 DOI: 10.1016/j.trci.2019.01.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Preliminary studies have shown that treatment with plasma exchange (PE) plus therapeutic albumin replacement in patients with Alzheimer's disease (AD) induced mobilization of plasma and cerebrospinal fluid amyloid β protein, associated with an improvement in memory and language functions, as well as the stabilization of brain perfusion, which persisted after treatment discontinuation. METHODS Alzheimer's Management By Albumin Replacement (AMBAR) is a multicenter, randomized, blinded and placebo-controlled, parallel-group, phase IIb/III trial enrolling patients with mild to moderate AD. The study evaluates PE with different replacement volumes of therapeutic albumin (5% and 20% Albutein®, Grifols), with or without intravenous immunoglobulin (Flebogamma® 5% DIF, Grifols). Patients are randomized to one of three active treatment groups or one control (sham PE) group (1:1:1:1). The intervention regime includes a first 6-week stage of intensive treatment, followed by a second 12-month stage of maintenance treatment. The change from the baseline to the end of treatment periods in the ADAS-Cog and ADCS-ADL scores are the coprimary efficacy variables. Secondary efficacy variables include change from the baseline in scores on cognitive, functional, behavioral, and overall progression tests; changes in plasma and cerebrospinal fluid levels of amyloid β and tau protein; and assessment of structural and functional changes in brain areas of interest. Safety and tolerability are assessed. RESULTS The study has enrolled 496 patients from 41 centers (19 in Spain and 22 in the USA); 347 of these patients were randomized and underwent close to 5000 PEs, of which approximately 25% were sham PEs. DISCUSSION We present an innovative approach for treating AD. The study has been designed to demonstrate clinical efficacy, defined as slow decline of the patient's cognition and brain function. The sample size has adequate power to detect differences between any of the active treatment groups and the control group, as well as between the three active treatment groups combined and the control group.
Collapse
Affiliation(s)
- Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Oscar López
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura Núñez
- Bioscience Research Group. Grifols S.A., Barcelona, Spain
| | | | - Mireia Torres
- Bioscience Research Group. Grifols S.A., Barcelona, Spain
| | | | - Antonio Páez
- Bioscience Research Group. Grifols S.A., Barcelona, Spain
| |
Collapse
|
96
|
Fu WY, Wang X, Ip NY. Targeting Neuroinflammation as a Therapeutic Strategy for Alzheimer's Disease: Mechanisms, Drug Candidates, and New Opportunities. ACS Chem Neurosci 2019; 10:872-879. [PMID: 30221933 DOI: 10.1021/acschemneuro.8b00402] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease, and its incidence is expected to increase owing to the aging population worldwide. Current therapies merely provide symptomatic relief. Therefore, interventions for AD that delay the disease onset or progression are urgently required. Recent genomics and functional studies suggest that immune/inflammatory pathways are involved in the pathogenesis of AD. Although many anti-inflammatory drug candidates have undergone clinical trials, most have failed. This might be because of our limited understanding of the pathological mechanisms of neuroinflammation in AD. However, recent advances in the understanding of immune/inflammatory pathways in AD and their regulatory mechanisms could open up new avenues for drug development targeting neuroinflammation. In this Review, we discuss the mechanisms and status of different anti-inflammatory drug candidates for AD that have undergone or are undergoing clinical trials and explore new opportunities for targeting neuroinflammation in AD drug development.
Collapse
Affiliation(s)
| | | | - Nancy Y. Ip
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
97
|
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by extracellular β-amyloid plaques and intracellular neurofibrillary tangles (NFTs), which are considered as major targets for AD therapies. However, no effective therapy is available to cure or prevent the progression of AD up until now. Accumulation of NFTs, which consist of abnormally hyperphosphorylated tau, is directly correlated with the degree of dementia in AD patients. Emerging evidence indicates that the prion-like seeding and spreading of tau pathology may be the key driver of AD. In the past decades, greater understanding of tau pathway reveals new targets for the development of specific therapies. Here, we review the recent research progress in the mechanism underlying tau pathology in AD and briefly introduce tau-based therapeutics.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, PR China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York 10314, United States
| |
Collapse
|
98
|
Panza F, Lozupone M, Seripa D, Imbimbo BP. Amyloid-β immunotherapy for alzheimer disease: Is it now a long shot? Ann Neurol 2019; 85:303-315. [PMID: 30635926 DOI: 10.1002/ana.25410] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 01/01/2023]
Abstract
The amyloid-β (Aβ) cascade hypothesis of Alzheimer disease (AD) holds that brain accumulation of Aβ initiates the disease process. Accordingly, drug research has targeted Aβ production, clearance, and deposition as therapeutic strategies. Unfortunately, candidate drugs have failed to show clinical benefit in established, early, or prodromal disease, or in those with high AD risk. Currently, monoclonal antibodies specifically directed against the most neurotoxic Aβ forms are undergoing large-scale trials to confirm initially encouraging results. However, recent findings on the normal physiology of Aβ suggest that accumulation may be compensatory rather than the pathological initiator. If this is true, alternative strategies will be needed to defeat this devastating disease. ANN NEUROL 2019;85:303-315.
Collapse
Affiliation(s)
- Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Neurodegenerative Disease Unit, Department of Clinical Research in Neurology, University of Bari Aldo Moro, Cardinal G. Panico Pious Foundation, Tricase, Italy.,Geriatric Unit, Home Relief of Suffering, Institute of Hospitalization and Scientific Care Foundation, San Giovanni Rotondo, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Davide Seripa
- Geriatric Unit, Home Relief of Suffering, Institute of Hospitalization and Scientific Care Foundation, San Giovanni Rotondo, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Pharmaceuticals, Parma, Italy
| |
Collapse
|
99
|
Zhang L, Xu J, Gao J, Chen P, Yin M, Zhao W. Decreased immunoglobulin G in brain regions of elder female APOE4-TR mice accompany with Aβ accumulation. IMMUNITY & AGEING 2019; 16:2. [PMID: 30700991 PMCID: PMC6347753 DOI: 10.1186/s12979-018-0142-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/20/2018] [Indexed: 11/25/2022]
Abstract
Background Apolipoprotein E4 (APOE4) and ageing are the most important known risk factors for late-onset Alzheimer’s disease (AD). In the present study, we determined the alterations of IgG, CD19, and Aβ in various brain regions of uninfected male and female APOE3- and APOE4-TR mice at the age of 3 and 10 months to elucidate impacts of AD risk factors on alterations of brain IgG. Results Positive staining for IgG was distributed across the brain, including neocortex, entorhinal cortex, hippocampus, thalamus and cerebellum. IgG positive staining was mainly located on microglia, but not astrocytes. Some IgG positive neurons were also observed, but only in mediodorsal thalamic nucleus. Compared with APOE3-TR mice, 10-month-old female APOE4-TR mice had lower IgG level in AD susceptible brain regions such as neocortex, entorhinal cortex and hippocampus, but no significant changes in thalamus and cerebellum, two regions nearly intact in AD. In addition, the expression of CD19, a specific marker for mature B cells, was significantly reduced in the hippocampus of 10-month-old female APOE4-TR mice. Although there were no obvious differences in plasma IgG levels between APOE4- and age matched female APOE3-TR mice, significant decreased B cell amount in blood of 10-month-old female APOE4-TR mice have also been found. Moreover, more obvious positive staining for Aβ was observed in the cortex of 10-month-old female APOE4-TR mice than other groups. Conclusions Our study demonstrated that AD risk factors were associated with IgG alterations in various brain regions, which might result from the defects of humoral immunity and lead to the impairment of IgG-mediated clearance of Aβ by microglia, therefore facilitated AD progression.
Collapse
Affiliation(s)
- Lihang Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Juan Xu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Jinchao Gao
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Peiqing Chen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Ming Yin
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Wenjuan Zhao
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| |
Collapse
|
100
|
Huisa BN, Thomas RG, Jin S, Oltersdorf T, Taylor C, Feldman HH. Memantine and Acetylcholinesterase Inhibitor Use in Alzheimer’s Disease Clinical Trials: Potential for Confounding by Indication. J Alzheimers Dis 2019; 67:707-713. [DOI: 10.3233/jad-180684] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Branko N. Huisa
- Department of Neurosciences, University of California, San Diego, CA, USA
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
| | - Ronald G. Thomas
- Department of Neurosciences, University of California, San Diego, CA, USA
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
- Department of Family Medicine and Public Health, University of California, San Diego, CA, USA
| | - Shelia Jin
- Department of Neurosciences, University of California, San Diego, CA, USA
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
| | - Tilman Oltersdorf
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
| | - Curtis Taylor
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
| | - Howard H. Feldman
- Department of Neurosciences, University of California, San Diego, CA, USA
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, CA, USA
| |
Collapse
|