1
|
Moawad MHED, Serag I, Alkhawaldeh IM, Abbas A, Sharaf A, Alsalah S, Sadeq MA, Shalaby MMM, Hefnawy MT, Abouzid M, Meshref M. Exploring the Mechanisms and Therapeutic Approaches of Mitochondrial Dysfunction in Alzheimer's Disease: An Educational Literature Review. Mol Neurobiol 2025; 62:6785-6810. [PMID: 39254911 PMCID: PMC12078384 DOI: 10.1007/s12035-024-04468-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Alzheimer's disease (AD) presents a significant challenge to global health. It is characterized by progressive cognitive deterioration and increased rates of morbidity and mortality among older adults. Among the various pathophysiologies of AD, mitochondrial dysfunction, encompassing conditions such as increased reactive oxygen production, dysregulated calcium homeostasis, and impaired mitochondrial dynamics, plays a pivotal role. This review comprehensively investigates the mechanisms of mitochondrial dysfunction in AD, focusing on aspects such as glucose metabolism impairment, mitochondrial bioenergetics, calcium signaling, protein tau and amyloid-beta-associated synapse dysfunction, mitophagy, aging, inflammation, mitochondrial DNA, mitochondria-localized microRNAs, genetics, hormones, and the electron transport chain and Krebs cycle. While lecanemab is the only FDA-approved medication to treat AD, we explore various therapeutic modalities for mitigating mitochondrial dysfunction in AD, including antioxidant drugs, antidiabetic agents, acetylcholinesterase inhibitors (FDA-approved to manage symptoms), nutritional supplements, natural products, phenylpropanoids, vaccines, exercise, and other potential treatments.
Collapse
Affiliation(s)
- Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Abdulrahman Sharaf
- Department of Clinical Pharmacy, Salmaniya Medical Complex, Government Hospital, Manama, Bahrain
| | - Sumaya Alsalah
- Ministry of Health, Primary Care, Governmental Health Centers, Manama, Bahrain
| | | | | | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
2
|
Shanazz K, Xie K, Oliver T, Bogan J, Vale FL, Sword J, Kirov SA, Terry A, O'Herron P, Blake DT. Cortical acetylcholine response to deep brain stimulation of the basal forebrain in mice. J Neurophysiol 2025; 133:825-838. [PMID: 39829107 DOI: 10.1152/jn.00476.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Deep brain stimulation (DBS) using electrical stimulation of neuronal tissue in the basal forebrain to enhance release of the neurotransmitter acetylcholine is under consideration to improve executive function in patients with dementia. Although some small studies indicate a positive response in the clinical setting, the relationship between DBS and acetylcholine pharmacokinetics is incompletely understood. We examined the cortical acetylcholine response to different stimulation parameters of the basal forebrain. Two-photon in vivo imaging was combined with deep brain stimulation in C57BL/6J mice. Stimulating electrodes were implanted in the subpallidal basal forebrain, and the ipsilateral somatosensory cortex was imaged. Acetylcholine activity was determined using the GRABACh-3.0 acetylcholine receptor sensor, and blood vessels were visualized with Texas red. Experiments manipulating stimulation frequency demonstrated that integrated acetylcholine-induced fluorescence was insensitive to frequency with the same number of pulses, and that maximum peak levels were achieved with frequencies from 60 to 130 Hz. Altering pulse train length indicated that longer stimulation resulted in higher peaks and more activation with sublinear summation. The acetylcholinesterase inhibitor, donepezil, increased the peak response to 600 pulses of stimulation at 60 Hz, and the integrated response increased by 57% with the 2 mg/kg dose and 126% with the 4 mg/kg dose. Acetylcholine levels returned to baseline with a time constant of 14-18 s. Donepezil increases total acetylcholine receptor activation associated with DBS but does not change temporal kinetics. The long time constants observed in the cerebral cortex add to the evidence supporting volume and synaptic neurotransmission.NEW & NOTEWORTHY Peak acetylcholine responses to deep brain stimulation of the subpallidal basal forebrain increases with increased frequency and number of pulses. Long recovery periods in the 10s of seconds support "volume" versus "phasic" transmission of acetylcholine. Donepezil administration enhances the effect of stimulation on cortical acetylcholine release.
Collapse
Affiliation(s)
- Khadijah Shanazz
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Kun Xie
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Tucker Oliver
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jamal Bogan
- Department of Science and Mathematics, Augusta University, Augusta, Georgia, United States
| | - Fernando L Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jeremy Sword
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Sergei A Kirov
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Alvin Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Philip O'Herron
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - David T Blake
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
3
|
Mo M, Abzhandadze T, Hoang MT, Sacuiu S, Jurado PG, Pereira JB, Naia L, Kele J, Maioli S, Xu H, Eriksdotter M, Garcia-Ptacek S. Antidepressant use and cognitive decline in patients with dementia: a national cohort study. BMC Med 2025; 23:82. [PMID: 39994788 PMCID: PMC11854023 DOI: 10.1186/s12916-025-03851-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/08/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Dementia is associated with psychiatric symptoms but the effects of antidepressants on cognitive function in dementia are understudied. We aimed to investigate the association between antidepressants and cognitive decline in patients with dementia, and the risk of severe dementia, fractures and death, depending on antidepressant class, drug, and dose. METHODS This is a national cohort study. Patients with dementia registered in the Swedish Registry for Cognitive/Dementia Disorders-SveDem from May 1, 2007, until October 16, 2018, with at least one follow-up after dementia diagnosis, and who were new users of antidepressants, were included. Antidepressant use as a time varying exposure defined during the 6 months leading up to dementia diagnosis or each subsequent follow-up. We used linear mixed models to examine the association between antidepressant use and cognitive trajectories assessed by Mini-Mental State Examination (MMSE) scores. We used Cox proportional hazards models to calculate the hazard ratios for severe dementia (MMSE score < 10), fracture, and death. We compared antidepressant classes and drugs, and analyzed dose-response. RESULTS We included 18740 patients (10 205 women [54.5%]; mean [SD] age, 78.2[7.4] years), of which 4271 (22.8%) received at least one prescription for an antidepressant. During follow-up, a total of 11912 prescriptions for antidepressants were issued, with selective serotonin reuptake inhibitors (SSRI) being the most common (64.8%). Antidepressant use was associated with faster cognitive decline (β (95% CI) = - 0.30(- 0.39, - 0.21) points/year), in particular sertraline (- 0.25(- 0.43, - 0.06) points/year), citalopram (- 0.41(- 0.55, - 0.27) points/year), escitalopram (- 0.76(- 1.09, - 0.44) points/year), and mirtazapine (- 0.19(- 0.34, - 0.04) points/year) compared with non-use. The association was stronger in patients with severe dementia (initial MMSE scores 0-9). Escitalopram showed a greater decline rate than sertraline. Compared with non-use, dose response of SSRIs on greater cognitive decline and higher risks of severe dementia, all-cause mortality, and fracture were observed. CONCLUSIONS In this cohort study, current antidepressant use was associated with faster cognitive decline; furthermore, higher dispensed doses of SSRIs were associated with higher risk for severe dementia, fractures, and all-cause mortality. These findings highlight the significance of careful and regular monitoring to assess the risks and benefits of different antidepressants use in patients with dementia.
Collapse
Affiliation(s)
- Minjia Mo
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden.
| | - Tamar Abzhandadze
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
- Department of Occupational Therapy and Physiotherapy, Sahlgrenska University Hospital, Rehabiliteringsmedicin, Vita Stråket 12, Vån 4, Gothenburg, 41345, Sweden
| | - Minh Tuan Hoang
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12a, Stockholm, 17165, Sweden
| | - Simona Sacuiu
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
- Department of Neuropsychiatry, Sahlgrenska University Hospital Mölndal, Region Västra Götaland, Sweden, Wallinsgatan 6, Mölndal, 43141, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology (EPINEP), Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 3, Göteborg, 413 90, Sweden
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Karolinska Vägen 37A, Stockholm, 171 64, Sweden
| | - Pol Grau Jurado
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
| | - Joana B Pereira
- Department of Clinical Neurosciences, Karolinska Institutet, Nobels Väg 9, Stockholm, D3, 17165, Sweden
| | - Luana Naia
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
| | - Julianna Kele
- Department of Laboratory Medicine, Team Neurovascular Biology and Health, Clinical Immunology, Karolinska Institutet, H5 Laboratoriemedicin, H5 Klin Immunologi Bergman, Huddinge, 14152, Sweden
| | - Silvia Maioli
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
| | - Hong Xu
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Karolinska Vägen 37A, Stockholm, 171 64, Sweden
| | - Sara Garcia-Ptacek
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, Stockholm, 14152, Sweden.
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Karolinska Vägen 37A, Stockholm, 171 64, Sweden.
| |
Collapse
|
4
|
Shimokawa H, Akishita M, Ihara M, Teramukai S, Ishiki A, Nagai Y, Fukushima M. Pivotal trial of low-intensity pulsed ultrasound therapy for early Alzheimer's disease: Rationale and design. J Alzheimers Dis Rep 2025; 9:25424823241312108. [PMID: 40034530 PMCID: PMC11864257 DOI: 10.1177/25424823241312108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/02/2024] [Indexed: 03/05/2025] Open
Abstract
Background There are lines of evidence suggesting that cerebral microcirculatory dysfunction is involved in the pathogenesis of Alzheimer's disease (AD). We have developed a low-intensity pulsed ultrasound (LIPUS) therapy that upregulates endothelial NO synthase with therapeutic angiogenesis. We demonstrated that the LIPUS therapy ameliorates cognitive declines in mouse models of AD and tended to do so in patients with early AD (mild AD and mild cognitive impairment due to AD) in the pilot trial. Thus, the Japanese government has designated our LIPUS device as the first breakthrough medical device in Japan. Objective We are performing a pivotal clinical trial (LIPUS-AD) to finally address the efficacy and safety of our LIPUS therapy in patients with early AD in Japan. Methods LIPUS-AD is a randomized, double-blind, placebo-controlled trial, in which a total of 220 patients with early AD, who are positive for amyloid-β (Aβ) PET, will be randomized in a 1:1 fashion. The LIPUS therapy is performed for the whole brain for one hour 3 times a week as one session under the special conditions (32 cycles, 0.5 MHz, 0.25 W/cm2). It is performed for 6 sessions with 3-month intervals in the LIPUS group for 72 weeks, while the placebo group receives placebo therapy. Before and at 72 weeks of the trial, all subjects undergo brain Aβ PET and MRI and 9 cognitive functions tests. The primary efficacy endpoint is the changes in ADAS-J-cog-14 scores from baseline to 72 weeks. Conclusions LIPUS-AD addresses efficacy and safety of the LIPUS therapy in patients with early AD.Clinical Trial Gov. No.: NCT05983575.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- Sound Wave Innovation, Inc., Tokyo, Japan
- Graduate School, International University of Health and Welfare, Narita, Japan
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Akishita
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Satoshi Teramukai
- Division of Data Science, The Clinical and Translational Research Center, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aiko Ishiki
- Division of Geriatric and Community Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoji Nagai
- Department of Clinical Research Facilitation, Institute for Advancement of Clinical and Translational Science, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
5
|
Geldmacher DS. Treatment of Alzheimer Disease. Continuum (Minneap Minn) 2024; 30:1823-1844. [PMID: 39620846 DOI: 10.1212/con.0000000000001503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Symptom-oriented treatment has been the mainstay of Alzheimer disease (AD) pharmacotherapy for decades. This article reviews the evidence basis for symptomatic treatments for AD and the emerging data on amyloid-lowering therapies with possible disease-slowing effects. LATEST DEVELOPMENT Amyloid-lowering monoclonal antibody therapies entered clinical use in 2021. In July 2023, lecanemab became the first of these to gain full US Food and Drug Administration (FDA) approval and limited Medicare payment coverage. Donanemab gained similar approval status in July 2024. The approved agents remove amyloid plaque from the brain and appear to slow clinical disease progression but can produce significant adverse events known as amyloid-related imaging abnormalities with cerebral edema or effusion and with cerebral hemorrhages. Extensive safety monitoring is therefore required, including scheduled MRI scans. Also in 2023, brexpiprazole became the first agent specifically approved by the FDA for agitation associated with AD. Suvorexant, an orexin receptor antagonist, previously was approved for the treatment of insomnia in people with mild and moderate AD. ESSENTIAL POINTS There is robust evidence for the use of acetylcholinesterase inhibitors for patients with mild, moderate, and severe dementia due to AD, including outcomes beyond changes in cognitive screening test scores. More limited studies support the use of memantine in moderate and severe stages. These agents have a primary effect of delaying decline in cognition and function and postponing the emergence of adverse behaviors. Pharmacotherapy for behavioral and psychological symptoms is less predictable, and most clinical trials have had negative results. Anti-amyloid therapies provide the first FDA-approved option to alter AD pathology, but an understanding of overall utility and value to patients remains in its infancy.
Collapse
|
6
|
Lee YC, Shi SM, Sison SM, Park CM, Oh G, Jeong S, McCarthy EP, Kim DH. Discontinuation of Cholinesterase Inhibitors Following Initiation of Memantine and Admission to Long-Term Care Among Older Adults. JAMA Netw Open 2024; 7:e2445878. [PMID: 39560943 DOI: 10.1001/jamanetworkopen.2024.45878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2024] Open
Abstract
Importance Discontinuing cholinesterase inhibitors when initiating memantine in patients with dementia may be reasonable to reduce treatment burden, costs, and the risk of adverse drug events. Objective To assess the association of cholinesterase inhibitor discontinuation on long-term care institutionalization among older adults with dementia who initiate memantine. Design, Setting, and Participants This retrospective propensity score-matched cohort study used Medicare claims data from January 2014 to December 2019. Participants included fee-for-service Medicare beneficiaries with dementia. Data were analyzed from September 2021 to August 2024. Exposures Discontinuation vs continuation of cholinesterase inhibitor. Main Outcomes and Measures The primary outcome was 1-year long-term care institutionalization-free days. Secondary outcomes include all-cause death and adverse drug events over 1 year. We performed subgroup analyses based on age, sex, dementia type (Alzheimer disease vs other), frailty, and dementia severity (mild vs moderate or severe) based on claims-based algorithms. The primary outcome was analyzed using nonparametric restricted mean survival time analysis. Results Among 16 292 beneficiaries who initiated memantine, 1820 (11.2%) discontinued cholinesterase inhibitors. In the propensity score-matched cohort of 3612 beneficiaries, the mean (SD) age was 80.7 (6.7) years, 2261 (62.6%) were female, and 1989 (55.0%) had a diagnosis of Alzheimer disease. Over 1 year, long-term care institutionalization occurred in 51 of 1806 beneficiaries (2.8%) who discontinued cholinesterase inhibitors (3.4 per 100 person-years) and 62 of 1806 beneficiaries (3.4%) who continued (4.1 per 100 person-years). There was no statistically significant difference in the 1-year mean institutionalization-free days between discontinuation and continuation groups (360.6 [95% CI, 359.3 to 362.0] days vs 359.1 [95% CI, 357.5 to 360.6] days; mean difference, 1.5 [95% CI,-0.5 to 3.6] days). The mean difference in the long-term care institutionalization-free days did not differ by age category, sex, dementia type, frailty, or dementia stage. Individuals who discontinued had a lower rate of fall-related injury (0.9 vs 2.0 per 100 person-years; hazard ratio [HR], 0.47 [95% CI, 0.25 to 0.88]). There was no difference between the discontinuation and continuation groups in all-cause death (10.4 vs 11.6 per 100 person-years; HR, 0.89 [95% CI, 0.72 to 1.10]). Conclusions and Relevance In this study, discontinuing cholinesterase inhibitors upon memantine initiation was not associated with an increased risk of long-term care institutionalization but with a lower risk of fall-related injury among older adults with dementia. These findings offer valuable insights for clinicians aiming to reduce treatment burden in this population.
Collapse
Affiliation(s)
- Yu-Chien Lee
- Department of Family Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
| | - Sandra M Shi
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
| | - Stephanie M Sison
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester
| | - Chan Mi Park
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
| | - Gahee Oh
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
| | - Sohyun Jeong
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
- Department of Pharmacy Practice, Massachusetts College of Pharmacy and Health Science, Boston
| | - Ellen P McCarthy
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
- Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Dae Hyun Kim
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
- Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
7
|
Costa ACS. On the Therapeutic Use of Monoclonal Antibodies Against Amyloid Plaques in Older Adults with Down Syndrome: A Narrative Review and Perspective. Brain Sci 2024; 14:1084. [PMID: 39595846 PMCID: PMC11591668 DOI: 10.3390/brainsci14111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by an extra copy of chromosome 21 (trisomy 21 or T21) and is associated with an increased risk of early-onset Alzheimer's disease (AD), also known as DS-associated AD (DSAD). Individuals with DS typically develop amyloid neuropathology in their late-thirties to early-forties and the mean age of onset of clinical dementia is approximately 55 years. Recent advances in AD clinical research have focused on monoclonal antibodies (mAbs) targeting amyloid-β (Aβ) plaques as a potential therapeutic approach. Therefore, there has been guarded enthusiasm about using anti-amyloid mAbs in the prevention/treatment of DSAD. This narrative review and perspective explores the current understanding of amyloid pathology in AD and DSAD, the rationale for using anti-amyloid mAbs in the treatment of DSAD, and the challenges and opportunities for research toward the application of this therapeutic strategy to older adults with DS.
Collapse
Affiliation(s)
- Alberto C S Costa
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| |
Collapse
|
8
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
9
|
Weng YX, Yang CC, Hsu WC, Kuo RN. Effects of traditional Chinese medicine on outcomes and costs of dementia care: results from a retrospective real-world study. Aging Clin Exp Res 2024; 36:204. [PMID: 39395084 PMCID: PMC11470846 DOI: 10.1007/s40520-024-02858-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024]
Abstract
OBJECTIVES This study aims to assess the impact of Traditional Chinese Medicine (TCM) on dementia patients, utilizing real-world data. Specifically, it seeks to evaluate how TCM influences clinical outcomes by examining changes in the Clinical Dementia Rating (CDR) and Mini-Mental State Examination (MMSE) scores, as well as its effect on medical expenses over a two-year period. Data from a multi-center research database spanning from 2004 to 2021 will be used to achieve these objectives, addressing the current gap in empirical data concerning intuitive outcomes and cognitive function assessments. METHODS Propensity score matching was adopted to improve comparability among the intervention and control groups. Due to repeated dependent variable measurements, the generalized estimating equation was used to control for socio-demographic characteristics, regional characteristics, and Western medicine treatments for dementia. RESULTS After propensity score matching, a total of 441 research subjects were included: 90 in the TCM intervention group and 351 in the non-TCM intervention group. The results of multivariate regression analysis showed that compared with the non-TCM intervention group, the MMSE scores in the TCM intervention group increased by 0.608 points each year. The annual change in CDR scores in the TCM intervention group was 0.702 times that of the non-TCM utilization group. After TCM intervention, annual outpatient expenses increased by US$492.2, hospitalization expenses increased by US$324.3, and total medical expenses increased by US$815.9, compared with the non-intervention group. CONCLUSIONS TCM interventions significantly decelerate cognitive decline in dementia patients, evidenced by slower reductions in MMSE scores and mitigated increases in CDR scores. However, these benefits are accompanied by increased medical expenses, particularly for outpatient care. Future healthcare strategies should balance the cognitive benefits of TCM with its economic impact, advocating for its inclusion in dementia care protocols.
Collapse
Affiliation(s)
- Yi-Xiang Weng
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Health Policy and Management, College of Public Health, National Taiwan University, Room 632, No.17, Syujhou Rd., Taipei City, 100, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Chuin Hsu
- Dementia Center, Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Raymond N Kuo
- Institute of Health Policy and Management, College of Public Health, National Taiwan University, Room 632, No.17, Syujhou Rd., Taipei City, 100, Taiwan.
- Population Health Research Center, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Tobin AB. A golden age of muscarinic acetylcholine receptor modulation in neurological diseases. Nat Rev Drug Discov 2024; 23:743-758. [PMID: 39143241 DOI: 10.1038/s41573-024-01007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/16/2024]
Abstract
Over the past 40 years, the muscarinic acetylcholine receptor family, particularly the M1-receptor and M4-receptor subtypes, have emerged as validated targets for the symptomatic treatment of neurological diseases such as schizophrenia and Alzheimer disease. However, despite considerable effort and investment, no drugs have yet gained clinical approval. This is largely attributable to cholinergic adverse effects that have halted the majority of programmes and resulted in a waning of interest in these G-protein-coupled receptor targets. Recently, this trend has been reversed. Driven by advances in structure-based drug design and an appreciation of the optimal pharmacological properties necessary to deliver clinical efficacy while minimizing adverse effects, a new generation of M1-receptor and M4-receptor orthosteric agonists and positive allosteric modulators are now entering the clinic. These agents offer the prospect of novel therapeutic solutions for 'hard to treat' neurological diseases, heralding a new era of muscarinic drug discovery.
Collapse
Affiliation(s)
- Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, The Advanced Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
11
|
Carrier-Auclair J, Lavoie M, Tastevin M, Laforce R. Efficacy of Acetylcholinesterase Inhibitors in the Logopenic Variant of Primary Progressive Aphasia. Dement Geriatr Cogn Disord 2024; 54:40-51. [PMID: 39255780 PMCID: PMC11797939 DOI: 10.1159/000540932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION For over 25 years, cholinesterase inhibitors (ChEIs) have been the main symptomatic treatment for Alzheimer's disease (AD). Several meta-analyses have supported their effectiveness in various neurocognitive, functional, and behavioral aspects of amnestic AD. Over 86% of cases of the logopenic variant of primary progressive aphasia (lvPPA), also named language variant AD, are caused by a similar pathologic process than AD, yet no study has examined the efficacy of ChEIs in this AD variant. We aimed to explore the efficacy of ChEIs in the treatment of lvPPA by comparing their evolution on the MMSE, and other functional and behavioral parameters, to that of treated amnestic AD patients. METHODS A retrospective chart review was performed in 45 patients with lvPPA and 52 patients with amnestic AD. Both groups were similar in terms of age, level of education, and onset of symptoms. Drug history and MMSE scores, as well as functional (activities of daily living [ADLs] and instrumental activities of daily living [IADLs]), neurocognitive and neuropsychiatric symptoms were collected on several time points before and after the introduction of ChEIs. Data were analyzed using ANOVA and a generalized linear mixed model. RESULTS Patients with lvPPA showed a similar trajectory of decline than amnestic AD patients on serial MMSEs up to 12-24 months after the introduction of ChEIs. There was a significant impact on ADLs but not IADLs and neuropsychiatric symptoms remained stable over time. CONCLUSION This study provides preliminary evidence for efficacy of ChEIs in patients with lvPPA and suggests similar benefits to those seen in amnestic AD patients, hence reassuring patients and their physicians. INTRODUCTION For over 25 years, cholinesterase inhibitors (ChEIs) have been the main symptomatic treatment for Alzheimer's disease (AD). Several meta-analyses have supported their effectiveness in various neurocognitive, functional, and behavioral aspects of amnestic AD. Over 86% of cases of the logopenic variant of primary progressive aphasia (lvPPA), also named language variant AD, are caused by a similar pathologic process than AD, yet no study has examined the efficacy of ChEIs in this AD variant. We aimed to explore the efficacy of ChEIs in the treatment of lvPPA by comparing their evolution on the MMSE, and other functional and behavioral parameters, to that of treated amnestic AD patients. METHODS A retrospective chart review was performed in 45 patients with lvPPA and 52 patients with amnestic AD. Both groups were similar in terms of age, level of education, and onset of symptoms. Drug history and MMSE scores, as well as functional (activities of daily living [ADLs] and instrumental activities of daily living [IADLs]), neurocognitive and neuropsychiatric symptoms were collected on several time points before and after the introduction of ChEIs. Data were analyzed using ANOVA and a generalized linear mixed model. RESULTS Patients with lvPPA showed a similar trajectory of decline than amnestic AD patients on serial MMSEs up to 12-24 months after the introduction of ChEIs. There was a significant impact on ADLs but not IADLs and neuropsychiatric symptoms remained stable over time. CONCLUSION This study provides preliminary evidence for efficacy of ChEIs in patients with lvPPA and suggests similar benefits to those seen in amnestic AD patients, hence reassuring patients and their physicians.
Collapse
Affiliation(s)
- Julie Carrier-Auclair
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques du CHU de Québec, Quebec City, Québec, Canada,
- Chaire de Recherche sur les Aphasies Primaires Progressives, Fondation de la Famille Lemaire, Quebec City, Québec, Canada,
| | - Monica Lavoie
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques du CHU de Québec, Quebec City, Québec, Canada
- Chaire de Recherche sur les Aphasies Primaires Progressives, Fondation de la Famille Lemaire, Quebec City, Québec, Canada
| | - Maud Tastevin
- Chaire de Recherche sur les Aphasies Primaires Progressives, Fondation de la Famille Lemaire, Quebec City, Québec, Canada
- CH Montperrin, Aix-en-Provence, France
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques du CHU de Québec, Quebec City, Québec, Canada
- Chaire de Recherche sur les Aphasies Primaires Progressives, Fondation de la Famille Lemaire, Quebec City, Québec, Canada
- Faculté de Médecine, Université Laval, Quebec City, Québec, Canada
| |
Collapse
|
12
|
Shanazz K, Xie K, Oliver T, Bogan J, Vale F, Sword J, Kirov SA, Terry A, O'Herron P, Blake DT. Cortical Acetylcholine Response to Deep Brain Stimulation of the Basal Forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605828. [PMID: 39131297 PMCID: PMC11312592 DOI: 10.1101/2024.07.30.605828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Deep brain stimulation (DBS), the direct electrical stimulation of neuronal tissue in the basal forebrain to enhance release of the neurotransmitter acetylcholine, is under consideration as a method to improve executive function in patients with dementia. While some small studies indicate a positive response in the clinical setting, the relationship between DBS and acetylcholine pharmacokinetics is incompletely understood. Objective We examined the cortical acetylcholine response to different stimulation parameters of the basal forebrain. Methods 2-photon imaging was combined with deep brain stimulation. Stimulating electrodes were implanted in the subpallidal basal forebrain, and the ipsilateral somatosensory cortex was imaged. Acetylcholine activity was determined using the GRABACh-3.0 muscarinic acetylcholine receptor sensor, and blood vessels were imaged with Texas red. Results Experiments manipulating pulse train frequency demonstrated that integrated acetylcholine induced fluorescence was insensitive to frequency, and that peak levels were achieved with frequencies from 60 to 130 Hz. Altering pulse train length indicated that longer stimulation resulted in higher peaks and more activation with sublinear summation. The acetylcholinesterase inhibitor donepezil increased the peak response to 10s of stimulation at 60Hz, and the integrated response increased 57% with the 2 mg/kg dose, and 126% with the 4 mg/kg dose. Acetylcholine levels returned to baseline with a time constant of 14 to 18 seconds in all experiments. Conclusions These data demonstrate that acetylcholine receptor activation is insensitive to frequency between 60 and 130 Hz. High peak responses are achieved with up to 900 pulses. Donepezil increases total acetylcholine receptor activation associated with DBS but did not change temporal kinetics. The long time constants observed in the cerebral cortex add to the evidence supporting volume in addition to synaptic transmission.
Collapse
Affiliation(s)
- Khadijah Shanazz
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Kun Xie
- Dept of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Tucker Oliver
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jamal Bogan
- Dept of Science and Mathematics, Augusta University, Augusta, GA
| | - Fernando Vale
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jeremy Sword
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sergei A Kirov
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
- Dept of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Alvin Terry
- Dept of Pharmacology and Toxicology , Medical College of Georgia, Augusta University, Augusta, GA
| | - Philip O'Herron
- Dept of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - David T Blake
- Dept of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
13
|
Hu K, Wu S, Xu J, Zhang Y, Zhang Y, Wu X, Miao J, Yao Y, Zhu S, Chen G, Ren J. Pongamol Alleviates Neuroinflammation and Promotes Autophagy in Alzheimer's Disease by Regulating the Akt/mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38841893 DOI: 10.1021/acs.jafc.4c00836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD), one of the neurodegenerative disorders, is highly correlated with the abnormal hyperphosphorylation of Tau and aggregation of β-amyloid (Aβ). Oxidative stress, neuroinflammation, and abnormal autophagy are key drivers of AD and how they contribute to neuropathology remains largely unknown. The flavonoid compound pongamol is reported to possess a variety of pharmacological activities, such as antioxidant, antibacterial, and anti-inflammatory. This study investigated the neuroprotective effect and its mechanisms of pongamol in lipopolysaccharide (LPS)-induced BV2 cells, d-galactose/sodium nitrite/aluminum chloride (d-gal/NaNO2/AlCl3)-induced AD mice, and Caenorhabditis elegans models. Our research revealed that pongamol reduced the release of inflammatory factors IL-1β, TNF-α, COX-2, and iNOS in LPS-induced BV2 cells. Pongamol also protected neurons and significantly restored memory function, inhibited Tau phosphorylation, downregulated Aβ aggregation, and increased oxidoreductase activity in the hippocampus of AD mice. In addition, pongamol reversed the nuclear transfer of NF-κB and increased the levels of Beclin 1 and LC3 II/LC3 I. Most importantly, the anti-inflammatory and promoter autophagy effects of pongamol may be related to the regulation of the Akt/mTOR signaling pathway. In summary, these results showed that pongamol has a potential neuroprotective effect, which greatly enriched the research on the pharmacological activity of pongamol for improving AD.
Collapse
Affiliation(s)
- Kun Hu
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Shaojun Wu
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Jiaxin Xu
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Yongzhen Zhang
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Yanan Zhang
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Xinyuan Wu
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Jie Miao
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Yongxu Yao
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Susu Zhu
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| | - Guangtong Chen
- School of Pharmacy, Nantong University, No. 19. Qixiu Road, Nantong, Jiangsu 226001, China
| | - Jie Ren
- School of Pharmacy, Changzhou University, No. 1. Gehu Middle Road, Changzhou, Jiangsu 213164, China
| |
Collapse
|
14
|
Ashique S, Sirohi E, Kumar S, Rihan M, Mishra N, Bhatt S, Gautam RK, Singh SK, Gupta G, Chellappan DK, Dua K. Aducanumab in Alzheimer's Disease: A Critical Update. Curr Med Chem 2024; 31:5004-5026. [PMID: 37497712 DOI: 10.2174/0929867331666230727103553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/23/2023] [Accepted: 05/19/2023] [Indexed: 07/28/2023]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder that results in cognitive decline. The incidence rates of AD have been increasing, particularly among individuals 60 years of age or older. In June 2021, the US FDA approved aducanumab, the first humanized monoclonal antibody, as a potential therapeutic option for AD. Clinical trials have shown this drug to effectively target the accumulation of Aβ (beta-amyloid) plaques in the brain, and its effectiveness is dependent on the dosage and duration of treatment. Additionally, aducanumab has been associated with improvements in cognitive function. Biogen, the pharmaceutical company responsible for developing and marketing aducanumab, has positioned it as a potential breakthrough for treating cerebral damage in AD. However, the drug has raised concerns due to its high cost, limitations, and potential side effects. AD is a progressive neurological condition that affects memory, cognitive function, and behaviour. It significantly impacts the quality of life of patients and caregivers and strains healthcare systems. Ongoing research focuses on developing disease-modifying therapies that can halt or slow down AD progression. The pathogenesis of AD involves various molecular cascades and signaling pathways. However, the formation of extracellular amyloid plaques is considered a critical mechanism driving the development and progression of the disease. Aducanumab, as a monoclonal antibody, has shown promising results in inhibiting amyloid plaque formation, which is the primary pathological feature of AD. This review explores the signaling pathways and molecular mechanisms through which aducanumab effectively prevents disease pathogenesis in AD.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Science, School of Pharmacy, Bharat Institute of Technology (BIT), Meerut 250103, UP, India
| | - Ekta Sirohi
- Department of Pharmaceutical Science, School of Pharmacy, Bharat Institute of Technology (BIT), Meerut 250103, UP, India
| | - Shubneesh Kumar
- Department of Pharmaceutical Science, School of Pharmacy, Bharat Institute of Technology (BIT), Meerut 250103, UP, India
| | - Mohd Rihan
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior 474005, Madhya Pradesh, India
| | - Shvetank Bhatt
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Gwalior 474005, Madhya Pradesh, India
| | - Rupesh K Gautam
- MM School of Pharmacy, Maharishi Markandeshwar University, Sadopur, Ambala, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| |
Collapse
|
15
|
Wang D, Zheng J, Sun X, Xie L, Yang Y. Study on the Pharmacological Mechanism of Icariin for the Treatment of Alzheimer's Disease Based on Network Pharmacology and Molecular Docking Techniques. Metabolites 2023; 14:1. [PMID: 38276291 PMCID: PMC10820555 DOI: 10.3390/metabo14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study is to explore the pharmacological mechanism of icariin (ICA) in the treatment of Alzheimer's disease (AD) based on network pharmacology and network molecular docking technology. In order to investigate the regulatory effect of ICA on the expression level of AD pathological phosphorylation regulatory proteins, this study further explored the possible molecular mechanism of ICA regulating AD autophagy through network pharmacology. Macromolecular docking network was verified by Autodock Vina 1.1.2 software. The main active ingredients of ICA, the physicochemical properties, and pharmacokinetic information of ICA were predicted using online databases and relevant information. The results showed that the targets of MAPK3, AKT1, HSP90AA1, ESR1, and HSP90AA1 were more critical in the treatment of AD. Autophagy, apoptosis, senescence factors, phosphatidylinositide 3-kinase/protein kinase B (P13K/AKT) signaling pathway, MAKP, mTOR, and other pathways were significantly associated with AD. Docking of ICA with HIF-1, BNIP3, PINK1, and Parkin pathway molecules showed that the key targets of the signaling pathway were more stably bound to ICA, which may provide a better pathway for ICA to regulate autophagy by providing a better pathway. ICA can improve AD, and its mechanism may be related to the P13K/AKT, MAKP, and mTOR signaling pathways, thereby regulating autophagy-related proteins.
Collapse
Affiliation(s)
- Dongwei Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Jilong Zheng
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Xingsheng Sun
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Liuwei Xie
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
16
|
Vesikansa A, Halminen O, Mehtälä J, Hörhammer I, Mikkola T, Ylisaukko-Oja T, Linna M. Early start of anti-dementia medication is associated with lower health and social care costs in Alzheimer´s patients: a Finnish nationwide register study. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2023; 24:1421-1428. [PMID: 36449133 DOI: 10.1007/s10198-022-01553-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 11/15/2022] [Indexed: 06/17/2023]
Abstract
OBJECTIVES To evaluate the association between health and social care costs and early start of anti-dementia medication in a nationwide cohort of Finnish Alzheimer's disease (AD) patients. METHODS The cohort included 7454 Finnish AD patients who had their first AD diagnosis in 2012 and lived at home at the time of diagnosis. Data were collected retrospectively from the Finnish national health and social care registers. The primary outcome was 2-year cumulative direct costs after the incident AD diagnosis. The exploratory variable was early anti-dementia medication start (anti-dementia medication started within 3 months of the incident AD diagnosis). Sociodemographic variables, admission to 24-h care and care intensity level, as well as comorbidities were considered as adjusting variables. RESULTS Of all patients, 88.9% started AD medication within 3 months of diagnosis. The 2-year cumulative costs were €30,787 and €40,484 per patient for early and late starters, respectively. When adjusted for possible confounders, early start of anti-dementia medication was associated with 26.5% lower 2-year cumulative costs compared to late starters (relative cost 0.735; p < 0.001). CONCLUSIONS Early diagnosis and start of anti-dementia medication is important for managing the costs of increasing number of AD patients.
Collapse
Affiliation(s)
| | - Olli Halminen
- Aalto University/Health Care, Engineering, Management and architecture (HEMA), Espoo , Finland
| | - Juha Mehtälä
- MedEngine Oy, Eteläranta 14, 00130, Helsinki, Finland
| | - Iiris Hörhammer
- Aalto University/Health Care, Engineering, Management and architecture (HEMA), Espoo , Finland
| | | | - Tero Ylisaukko-Oja
- MedEngine Oy, Eteläranta 14, 00130, Helsinki, Finland
- Faculty of Medicine, Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Miika Linna
- Aalto University/Health Care, Engineering, Management and architecture (HEMA), Espoo , Finland
- Department of Health and Social Management, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
17
|
Eggli Y, Halfon P, Zeukeng MJ, Kherad O, Schaller P, Raetzo MA, Klay MF, Favre BM, Schaller D, Marti J. Potentially Inappropriate Medication Dispensing in Outpatients: Comparison of Different Measurement Approaches. Risk Manag Healthc Policy 2023; 16:2565-2578. [PMID: 38024485 PMCID: PMC10680376 DOI: 10.2147/rmhp.s427516] [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: 07/20/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose of the Research This paper aims at comparing different approaches to measure potentially inappropriate medication (PIM) with routinely collected data on prescriptions, patient age institutionalization status (ie in nursing home or in the community). A secondary objective is to measure the rate and prevalence of PIM dispensing and to identify problematic practices in Switzerland. Material and Methods The studied population includes about 90,000 insured over 17 years old from a Swiss health maintenance organization in 2019 and 2020. We computed and compared the number of PIM per patient for Beers criteria, Priscus list, Laroche, NORGEP and Prescrire approaches. We also created a composite indicator that accounts for the specificities of the Swiss context (adaptation to the Swiss drugs' market, recommendations in force related to sleeping pills, anxiolytics and NSAIDs). We also stratified the analysis per physician, including initiation and cessation of PIM prescription. Results Our comparison revealed similarities between the approaches, but also that each of them had specific gaps that provides further motivation for the development of a composite approach. PIM rate was particularly high for sleeping pills, anxiolytics, NSAIDs, even when analyses were limited to chronic use. Drugs with anticholinergic effect were also frequently prescribed. Based on our composite indicator, 27% of insured over 64 years old received at least one PIM in 2020, and 8% received more than one. Our analyses also reveal that for sleeping pills and anxiolytics, half of the volume (or prevalence?) occurs in the <65 population. We observed strong variations between physicians and a significant proportion of new users among patients with PIM. Conclusion Our results show that PIMs prescribing is very frequent in Switzerland and is driven mostly by a few drug categories. There is important physician variation in PIM prescribing that warrants the development of intervention targeted at high PIM-prescribers.
Collapse
Affiliation(s)
- Yves Eggli
- Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Patricia Halfon
- Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | | | - Omar Kherad
- Internal Medicine Department, La Tour Hospital and University of Geneva, Geneva, Switzerland
| | | | | | | | | | | | - Joachim Marti
- Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Bittner N, Funk CSM, Schmidt A, Bermpohl F, Brandl EJ, Algharably EEA, Kreutz R, Riemer TG. Psychiatric Adverse Events of Acetylcholinesterase Inhibitors in Alzheimer's Disease and Parkinson's Dementia: Systematic Review and Meta-Analysis. Drugs Aging 2023; 40:953-964. [PMID: 37682445 PMCID: PMC10600312 DOI: 10.1007/s40266-023-01065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The acetylcholinesterase inhibitors (AChEIs) donepezil, galantamine, and rivastigmine are commonly used in the management of various forms of dementia. OBJECTIVES While these drugs are known to induce classic cholinergic adverse events such as diarrhea, their potential to cause psychiatric adverse events has yet to be thoroughly examined. METHODS We sought to determine the risk of psychiatric adverse events associated with the use of AChEIs through a systematic review and meta-analysis of double-blind randomized controlled trials involving patients with Alzheimer's dementia and Parkinson's dementia. RESULTS A total of 48 trials encompassing 22,845 patients were included in our analysis. Anorexia was the most commonly reported psychiatric adverse event, followed by agitation, insomnia, and depression. Individuals exposed to AChEIs had a greater risk of experiencing appetite disorders, insomnia, or depression compared with those who received placebo (anorexia: odds ratio [OR] 2.93, 95% confidence interval [CI] 2.29-3.75; p < 0.00001; decreased appetite: OR 1.93, 95% CI 1.33-2.82; p = 0.0006; insomnia: OR 1.55, 95% CI 1.25-1.93; p < 0.0001; and depression: OR 1.59, 95% CI 1.23-2.06, p = 0.0004). Appetite disorders were also more frequent with high-dose versus low-dose therapy. A subgroup analysis revealed that the risk of insomnia was higher for donepezil than for galantamine. CONCLUSIONS Our findings suggest that AChEI therapy may negatively impact psychological health, and careful monitoring of new psychiatric symptoms is warranted. Lowering the dose may resolve some psychiatric adverse events, as may switching to galantamine in the case of insomnia. CLINICAL TRIAL REGISTRATION The study was pre-registered on PROSPERO (CRD42021258376).
Collapse
Affiliation(s)
- Nadine Bittner
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Cleo S. M. Funk
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Alexander Schmidt
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Felix Bermpohl
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Eva J. Brandl
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Engi E. A. Algharably
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Thomas G. Riemer
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| |
Collapse
|
19
|
Liu KY, Walsh S, Brayne C, Merrick R, Richard E, Howard R. Evaluation of clinical benefits of treatments for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2023; 4:e645-e651. [PMID: 37924845 DOI: 10.1016/s2666-7568(23)00193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 11/06/2023] Open
Abstract
The need for regulatory approval of new therapies for the treatment of Alzheimer's disease-a progressive neurodegenerative condition-has made the assessment of treatment efficacy an urgent priority for discussion and investigation in the field. In the first part of this Personal View, we summarise current views on what constitutes a clinically meaningful benefit from treatment for Alzheimer's disease, including the concept of a minimum treatment effect against which to compare trial outcomes and its limitations. Considering existing and divergent definitions of clinically meaningful change, we define this concept in the second part of the Personal View by proposing a new approach that consecutively considers whether a treatment benefit for Alzheimer's disease is noticeable, valuable, and worthwhile in the context of costs and risks. This approach could be a useful foundation from which the field can move forwards on this issue and address existing gaps in understanding.
Collapse
Affiliation(s)
- Kathy Y Liu
- Division of Psychiatry, University College London, London, UK.
| | - Sebastian Walsh
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Richard Merrick
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands; Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
20
|
Salemme S, Ancidoni A, Vanacore N. Responder analyses for anti-amyloid immunotherapies for Alzheimer's disease: a paradigm shift by regulatory authorities is urgently needed. Brain Commun 2023; 5:fcad276. [PMID: 37901042 PMCID: PMC10612473 DOI: 10.1093/braincomms/fcad276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 08/31/2023] [Accepted: 10/20/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Simone Salemme
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Antonio Ancidoni
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome 00161, Italy
- Department of Public Health and Infectious Diseases, Sapienza University, Rome 00185, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome 00161, Italy
| |
Collapse
|
21
|
Dávila G, Torres-Prioris MJ, López-Barroso D, Berthier ML. Turning the Spotlight to Cholinergic Pharmacotherapy of the Human Language System. CNS Drugs 2023; 37:599-637. [PMID: 37341896 PMCID: PMC10374790 DOI: 10.1007/s40263-023-01017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Even though language is essential in human communication, research on pharmacological therapies for language deficits in highly prevalent neurodegenerative and vascular brain diseases has received little attention. Emerging scientific evidence suggests that disruption of the cholinergic system may play an essential role in language deficits associated with Alzheimer's disease and vascular cognitive impairment, including post-stroke aphasia. Therefore, current models of cognitive processing are beginning to appraise the implications of the brain modulator acetylcholine in human language functions. Future work should be directed further to analyze the interplay between the cholinergic system and language, focusing on identifying brain regions receiving cholinergic innervation susceptible to modulation with pharmacotherapy to improve affected language domains. The evaluation of language deficits in pharmacological cholinergic trials for Alzheimer's disease and vascular cognitive impairment has thus far been limited to coarse-grained methods. More precise, fine-grained language testing is needed to refine patient selection for pharmacotherapy to detect subtle deficits in the initial phases of cognitive decline. Additionally, noninvasive biomarkers can help identify cholinergic depletion. However, despite the investigation of cholinergic treatment for language deficits in Alzheimer's disease and vascular cognitive impairment, data on its effectiveness are insufficient and controversial. In the case of post-stroke aphasia, cholinergic agents are showing promise, particularly when combined with speech-language therapy to promote trained-dependent neural plasticity. Future research should explore the potential benefits of cholinergic pharmacotherapy in language deficits and investigate optimal strategies for combining these agents with other therapeutic approaches.
Collapse
Affiliation(s)
- Guadalupe Dávila
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - María José Torres-Prioris
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Diana López-Barroso
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Marcelo L Berthier
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain.
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain.
| |
Collapse
|
22
|
Bava JM, Wang Z, Bick SK, Englot DJ, Constantinidis C. Improving Visual Working Memory with Cholinergic Deep Brain Stimulation. Brain Sci 2023; 13:917. [PMID: 37371395 PMCID: PMC10296349 DOI: 10.3390/brainsci13060917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Acetylcholine is a critical modulatory neurotransmitter for cognitive function. Cholinergic drugs improve cognitive performance and enhance neuronal activity in the sensory and association cortices. An alternative means of improving cognitive function is through the use of deep brain stimulation. Prior animal studies have demonstrated that stimulation of the nucleus basalis of Meynert through DBS improves cognitive performance on a visual working memory task to the same degree as cholinesterase inhibitors. Additionally, unlike current pharmacological treatments for neurocognitive disorders, DBS does not lose efficacy over time and adverse effects are rare. These findings suggest that DBS may be a promising alternative for treating cognitive impairments in neurodegenerative disorders such as Alzheimer's disease. Thus, further research and human trials should be considered to assess the potential of DBS as a therapeutic treatment for these disorders.
Collapse
Affiliation(s)
- Janki M. Bava
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; (J.M.B.); (D.J.E.)
| | - Zhengyang Wang
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235, USA;
| | - Sarah K. Bick
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Dario J. Englot
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; (J.M.B.); (D.J.E.)
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Christos Constantinidis
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; (J.M.B.); (D.J.E.)
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235, USA;
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
23
|
Ajenikoko MK, Ajagbe AO, Onigbinde OA, Okesina AA, Tijani AA. Review of Alzheimer's disease drugs and their relationship with neuron-glia interaction. IBRO Neurosci Rep 2023; 14:64-76. [PMID: 36593897 PMCID: PMC9803919 DOI: 10.1016/j.ibneur.2022.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Because Alzheimer's disease has no known treatment, sufferers and their caregivers must concentrate on symptom management. Astrocytes and microglia are now known to play distinct physiological roles in synaptic function, the blood-brain barrier, and neurovascular coupling. Consequently, the search for drugs that can slow the degenerative process in dementia sufferers continues because existing drugs are designed to alleviate the symptoms of Alzheimer's disease. Drugs that address pathological changes without interfering with the normal function of glia, such as eliminating amyloid-beta deposits, are prospective treatments for neuroinflammatory illnesses. Because neuron-astrocytes-microglia interactions are so complex, developing effective, preventive, and therapeutic medications for AD will necessitate novel methodologies and strategic targets. This review focused on existing medications used in treating AD amongst which include Donepezil, Choline Alphoscerate, Galantamine, Dextromethorphan, palmitoylethanolamide, citalopram, resveratrol, and solanezumab. This review summarizes the effects of these drugs on neurons, astrocytes, and microglia interactions based on their pharmacokinetic properties, mechanism of action, dosing, and clinical presentations.
Collapse
Affiliation(s)
- Michael Kunle Ajenikoko
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, Ishaka, Uganda
| | - Abayomi Oyeyemi Ajagbe
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, P.M.B. 900001 Abuja, Nigeria
| | - Oluwanisola Akanji Onigbinde
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, P.M.B. 900001 Abuja, Nigeria
| | - Akeem Ayodeji Okesina
- Department of Clinical Medicine and Community Health, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Ahmad Adekilekun Tijani
- Department of Anatomy, Faculty of Basic Medical Sciences, Modibbo Adama University, Yola, Nigeria
| |
Collapse
|
24
|
Lee JE, Kang HW, Jung SA, Lee SY, Kim JY, Lee DE, Jeong JH, Jung IC, Cho E. The effects of herbal medicine (Jujadokseo-hwan) on quality of life in patients with mild cognitive impairment: Cost-effectiveness analysis alongside randomized controlled trial. Integr Med Res 2023; 12:100914. [PMID: 36632128 PMCID: PMC9826841 DOI: 10.1016/j.imr.2022.100914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/17/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Background Mild cognitive impairment (MCI), the early stage of dementia, requires effective intervention for symptom management and improving patients' quality of life (QoL). Jujadokseo-hwan (JDH) is a Korean herbal medicine prescription used to improve MCI symptoms, such as memory deficit. This study evaluates the improvement in QoL through JDH. Alongside a clinical trial, it estimates the cost-effectiveness of JDH, compared to placebo, for MCI over 24 weeks. Methods Changes in QoL were measured using the EuroQol-5 Dimensions (EQ-5D) and Korean version QoL-Alzheimer's Disease (KQOL-AD). Direct medical and non-medical costs were surveyed and incremental cost-effectiveness ratios (ICER) per QALY for JDH were produced. Results In total, 64 patients were included in the economic evaluation (n = 35 in JDH, n = 29 in placebo). In the JDH group, EQ-5D and KQOL-AD improved by 0.020 (p = .318) and 3.40 (p = .011) over 24 weeks, respectively. In the placebo group, they increased by 0.001 (p=.920) and 1.07 (p=.130), respectively. The ICER was KRW 76,400,000 per QALY and KRW 108,000 per KQOL-AD for JDH, compared to the placebo group. Conclusion JDH is not considered a cost-effective treatment option compared with placebo; however, it positively affects QoL improvement in patients with MCI.
Collapse
Affiliation(s)
- Ji-Eun Lee
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hyung Won Kang
- Department of Neuropsychiatry, College of Oriental Medicine, Wonkwang University, Iksan, Republic of Korea
| | - Sun-A Jung
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - So-Young Lee
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ju Yeon Kim
- Department of Oriental Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea.,Department of Neuropsychiatry, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Da Eun Lee
- Department of Neuropsychiatry, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Jin-Hyung Jeong
- Department of Oriental Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea.,Department of Neuropsychiatry, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - In Chul Jung
- Department of Oriental Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea.,Department of Neuropsychiatry, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea.,Corresponding authors at: Department of Oriental Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, 34520, Republic of Korea (I. C. Jung); College of Pharmacy, Sookmyung Women's University, Cheongpa-ro 47-gil, 100, Yongsan-gu, Seoul, 04310, Republic of Korea (E. Cho)
| | - Eun Cho
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea,Corresponding authors at: Department of Oriental Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, 34520, Republic of Korea (I. C. Jung); College of Pharmacy, Sookmyung Women's University, Cheongpa-ro 47-gil, 100, Yongsan-gu, Seoul, 04310, Republic of Korea (E. Cho)
| |
Collapse
|
25
|
Perlett L, Smith EE. Treatment of Vascular and Neurodegenerative Forms of Cognitive Impairment and Dementias. Clin Geriatr Med 2023; 39:135-149. [PMID: 36404026 DOI: 10.1016/j.cger.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ideally, dementia care should be provided by a collaborative team. Eligible patients should be treated with the cognitive-enhancing medications, the cholinesterase inhibitors and memantine. For most of the common causes of dementia, there are no disease-modifying medications, with the exception that vascular dementia can be prevented by treating vascular risk factors to prevent stroke. There is hope that Alzheimer disease can be treated by using monoclonal antibodies that target amyloid beta, although more trials are needed. Holistic, patient-centered care can enhance quality and extend the time that the patient can live safely in the community.
Collapse
Affiliation(s)
- Landon Perlett
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Eric E Smith
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
26
|
Brody M, Agronin M, Herskowitz BJ, Bookheimer SY, Small GW, Hitchinson B, Ramdas K, Wishard T, McInerney KF, Vellas B, Sierra F, Jiang Z, Mcclain-Moss L, Perez C, Fuquay A, Rodriguez S, Hare JM, Oliva AA, Baumel B. Results and insights from a phase I clinical trial of Lomecel-B for Alzheimer's disease. Alzheimers Dement 2023; 19:261-273. [PMID: 35357079 PMCID: PMC10084163 DOI: 10.1002/alz.12651] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 01/18/2023]
Abstract
HYPOTHESIS We hypothesized that Lomecel-B, an allogeneic medicinal signaling cell (MSC) therapeutic candidate for Alzheimer's disease (AD), is safe and potentially disease-modifying via pleiotropic mechanisms of action. KEY PREDICTIONS We prospectively tested the predictions that Lomecel-B administration to mild AD patients is safe (primary endpoint) and would provide multiple exploratory indications of potential efficacy in clinical and biomarker domains (prespecified secondary/exploratory endpoints). STRATEGY AND KEY RESULTS Mild AD patient received a single infusion of low- or high-dose Lomecel-B, or placebo, in a double-blind, randomized, phase I trial. The primary safety endpoint was met. Fluid-based and imaging biomarkers indicated significant improvement in the Lomecel-B arms versus placebo. The low-dose Lomecel-B arm showed significant improvements versus placebo on neurocognitive and other assessments. INTERPRETATION Our results support the safety of Lomecel-B for AD, suggest clinical potential, and provide mechanistic insights. This early-stage study provides important exploratory information for larger efficacy-powered clinical trials.
Collapse
Affiliation(s)
- Mark Brody
- Brain Matters Research, Delray Beach, Florida, USA
| | | | | | - Susan Y Bookheimer
- Dept. of Psychiatry and Biobehavioral Sciences, and Semel Institute For Neuroscience and Human Behavior, UCLA David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Gary W Small
- Psychiatry, Hackensack Meridian Health, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | | | | | - Tyler Wishard
- Interdepartmental Program in Neuroscience, UCLA, and Semel Institute For Neuroscience and Human Behavior, UCLA David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | - Bruno Vellas
- Gérontopôle, Department of Geriatric Internal Medicine, University of Toulouse, Toulouse, France
| | - Felipe Sierra
- National Institute of Aging, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Carmen Perez
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ana Fuquay
- Brain Matters Research, Delray Beach, Florida, USA
| | | | - Joshua M Hare
- Longeveron Inc., Miami, Florida, USA.,Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Bernard Baumel
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
27
|
Repetitive TMS applied to the precuneus stabilizes cognitive status in Alzheimer’s disease. Brain 2022; 145:3730-3732. [DOI: 10.1093/brain/awac322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/23/2022] Open
Abstract
This scientific commentary refers to ‘Precuneus magnetic stimulation for Alzheimer's disease: a randomized, sham-controlled trial’ by Koch et al. (https://doi.org/10.1093/brain/awac285).
Collapse
|
28
|
Dwomoh L, Rossi M, Scarpa M, Khajehali E, Molloy C, Herzyk P, Mistry SN, Bottrill AR, Sexton PM, Christopoulos A, Conn J, Lindsley CW, Bradley SJ, Tobin AB. M 1 muscarinic receptor activation reduces the molecular pathology and slows the progression of prion-mediated neurodegenerative disease. Sci Signal 2022; 15:eabm3720. [PMID: 36378750 PMCID: PMC7616172 DOI: 10.1126/scisignal.abm3720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Many dementias are propagated through the spread of "prion-like" misfolded proteins. This includes prion diseases themselves (such as Creutzfeldt-Jakob disease) and Alzheimer's disease (AD), for which no treatments are available to slow or stop progression. The M1 acetylcholine muscarinic receptor (M1 receptor) is abundant in the brain, and its activity promotes cognitive function in preclinical models and in patients with AD. Here, we investigated whether activation of the M1 receptor might slow the progression of neurodegeneration associated with prion-like misfolded protein in a mouse model of prion disease. Proteomic and transcriptomic analysis of the hippocampus revealed that this model had a molecular profile that was similar to that of human neurodegenerative diseases, including AD. Chronic enhancement of the activity of the M1 receptor with the positive allosteric modulator (PAM) VU0486846 reduced the abundance of prion-induced molecular markers of neuroinflammation and mitochondrial dysregulation in the hippocampus and normalized the abundance of those associated with neurotransmission, including synaptic and postsynaptic signaling components. PAM treatment of prion-infected mice prolonged survival and maintained cognitive function. Thus, allosteric activation of M1 receptors may reduce the severity of neurodegenerative diseases caused by the prion-like propagation of misfolded protein.
Collapse
Affiliation(s)
- Louis Dwomoh
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mario Rossi
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Miriam Scarpa
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Elham Khajehali
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Colin Molloy
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Pawel Herzyk
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Shailesh N Mistry
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Andrew R Bottrill
- Research Technology Platforms, University of Warwick, School of Life Sciences, Coventry CV4 7AL, UK
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Australian Research Council Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Australian Research Council Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jeffrey Conn
- Warren Centre for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Warren Centre for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Sophie J Bradley
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
29
|
Britton A, Zimmermann M. Informal dementia care: The carer's lived experience at the divides between policy and practice. DEMENTIA 2022; 21:2117-2127. [PMID: 35838118 PMCID: PMC9483676 DOI: 10.1177/14713012221112234] [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] [Indexed: 11/15/2022]
Abstract
Support for informal dementia care at a local community level is not working for most carers today. Carers looking after a person with dementia have long lamented the absence of an empowered named support and an effectively actioned care plan. Drawing on literary writing and social research, we argue in this article that these challenges have existed since dementia emerged as a major condition in the West during the 1980s. Based on this historical context, we ask: Why has this issue persisted over the last four decades? How have healthcare politics and policy initiatives responded to these requests? And what can we learn from this for the current, COVID-19 exacerbated crisis of care? This article focuses on the English context, to discuss these ongoing challenges in the light of a series of policy papers, and to ask what is hampering the implementation of such policy initiatives. In England, local authorities are responsible for dementia support. This article focuses on the situation in a county in the Midlands where one of us (AB) has been lobbying local government for over a decade. The discussion contextualises the lived experience of dementia care within the situation exacerbated by the COVID-19 pandemic, ensuing politics of crises and persistent emphasis on cure over care. We find that the absence on two points centrally challenges care: a joined-up approach between health and social care and adequate information on available care support services, accessible through an empowered named contact. To enhance the lived experience of dementia care, consistent provision of individual named support and professional care support, as and when required, should become essential to local implementation of the care policy.
Collapse
Affiliation(s)
- Anthony Britton
- Founder Trustee, The Pam Britton Trust for Dementia, Leamington Spa, UK
| | - Martina Zimmermann
- UKRI Future Leaders Fellow and Lecturer, Health Humanities and Health Sciences, Department of English, 4616King's College London, London, UK
| |
Collapse
|
30
|
Lee JH. The listed, delisted, and sustainability of therapeutic medicines for dementia patients: the study is specific to South Korea. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:535-546. [PMID: 35122115 PMCID: PMC8989833 DOI: 10.1007/s00210-022-02209-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/19/2022] [Indexed: 01/02/2023]
Abstract
The Dementia Management Act (DMA) came into effect on August 4, 2011, in South Korea. Diagnosis and medication were rapidly performed for dementia in a short time. We investigated the cardiac effects of increased drug prescription following DMA. We observed a correlation between Alzheimer’s disease (AD) and anti-AD drug (AAD) groups from 2010 to 2019 on the National Health Insurance System (NHIS) of South Korea. This study investigated the increase and decrease in deaths of AD patients with AAD. We analysed the mortality per 100,000 population with the R2 Calculator. Moreover, we made the up or down datum line for the simple decision on the listed, delisted, and sustainable drug examined by a linear equation and R2. We observed that life expectancy was diminished by AAD in Sorokdo National Hospital. In the NHIS, donepezil and rivastigmine increased the number of deaths decided on R2 > 0.75. Memantine was sustainable. We could not decide on galantamine because it is one of the other groups. We made a straightforward decision-maker of delisted, listed, or sustainable criteria based on mortality and datum line.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science & Research Center, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
31
|
Huo Z, Lin J, Bat BKK, Chan TK, Yip BHK, Tsoi KKF. Cost-effectiveness of pharmacological therapies for people with Alzheimer's disease and other dementias: a systematic review and meta-analysis. Cost Eff Resour Alloc 2022; 20:19. [PMID: 35443684 PMCID: PMC9022294 DOI: 10.1186/s12962-022-00354-3] [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: 11/04/2021] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
Objectives This study aims to synthesize the empirical economic evidence of pharmaceutical therapies for people with dementia. Study design Systematic review and meta-analysis. Literature evaluating the costs and effects of drug therapies for dementia was indexed until December 2021. Quality of study was assessed using the Cochrane Risk of Bias Tool and Consensus on Health Economic Criteria list. Cost data were standardized to 2020 US dollars and analyzed from healthcare service and societal perspectives. Random-effects models were used to synthesize economic and clinical data, based on mean differences (MDs) and standardized MDs. Results Ten unique studies were identified from 11,771 records. Acetylcholinesterase inhibitors (AChEIs) and memantine improved dementia-related symptoms, alongside nonsignificant savings in societal cost (AChEIs: MD-2002 [− 4944 ~ 939]; memantine: MD-6322 [− 14355 ~ 1711]). Despite decreases in cost, antidepressants of mirtazapine and sertraline and second-generation antipsychotics were limited by their significant side effects on patients’ cognitive and activity functions. Subgroup analysis indicated that the impacts of AChEIs on cost were affected by different analytical perspectives, follow-up periods, and participant age. Conclusions AChEIs and memantine are cost-effective with improvements in dementia-related symptoms and trends of cost-savings. More empirical evidence with non-industrial sponsorships and rigorous design in different settings is warranted.
Supplementary Information The online version contains supplementary material available at 10.1186/s12962-022-00354-3.
Collapse
Affiliation(s)
- Zhaohua Huo
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiaer Lin
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Baker K K Bat
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Tak Kit Chan
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Benjamin H K Yip
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kelvin K F Tsoi
- JC School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China. .,Stanley Ho Big Data Decision Analytics Research Centre, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
32
|
Design, Synthesis, and Evaluation of Novel 2 H-Benzo[b][1,4]thiazin-3(4 H)-one Derivatives as New Acetylcholinesterase Inhibitors. Molecules 2022; 27:molecules27072121. [PMID: 35408519 PMCID: PMC9000418 DOI: 10.3390/molecules27072121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a slowly progressive neurodegenerative disease that causes dementia in people aged 65 and over. In the present study, a series of thiadiazole hybrid compounds with benzothiazine derivatives as acetylcholinesterase inhibitors were developed and evaluated for their biological activity. The AChE and BChE inhibition potentials of all compounds were evaluated by using the in vitro Ellman method. The biological evaluation showed that compounds 3i and 3j displayed significant inhibitory activity against AChE. Compounds 3i and 3j showed IC50 values of 0.027 µM and 0.025 µM against AChE, respectively. The reference drug donepezil (IC50 = 0.021 µM) also showed significant inhibition against AChE. Further docking simulation also revealed that these compounds (3i and 3j) interacted with the active site of the enzyme similarly to donepezil. The antioxidant study revealed that compounds 3i and 3j exhibited greater antioxidant effects. An in vitro blood–brain barrier permeability study showed that compounds 3i and 3j are promising compounds against AD. The cytotoxicity study of compounds 3i and 3j showed non-cytotoxic with an IC50 value of 98.29 ± 3.98 µM and 159.68 ± 5.53 µM against NIH/3T3 cells, respectively.
Collapse
|
33
|
Abstract
Primary palliative care is a fundamental aspect of high-quality care for patients with a serious illness such as dementia. The clinician caring for a patient and family suffering with dementia can provide primary palliative care in numerous ways. Perhaps the most important aspects are high quality communication while sharing a diagnosis, counseling the patient through progression of illness and prognostication, and referral to hospice when appropriate. COVID-19 presents additional risks of intensive care requirement and mortality which we must help patients and families navigate. Throughout all of these discussions, the astute clinician must monitor the patient's decision making capacity and balance respect for autonomy with protection against uninformed consent. Excellent primary palliative care also involves discussion of deprescribing medications of uncertain benefit such as long term use of cholinesterase inhibitors and memantine and being vigilant in the monitoring of pain with its relationship to behavioral disturbance in patients with dementia. Clinicians should follow a standardized approach to pain management in this vulnerable population. Caregiver burden is high for patients with dementia and comprehensive care should also address this burden and implement reduction strategies. When these aspects of care are particularly complex or initial managements strategies fall short, palliative care specialists can be an important additional resource not only for the patient and family, but for the care team struggling to guide the way through a disease with innumerable challenges.
Collapse
Affiliation(s)
- Neal Weisbrod
- Department of Neurology, University of Florida, 1505 SW Archer, Gainesville, FL, 32608, USA.
- Department of Medicine Division of Palliative Care, University of Florida, 1505 SW Archer, Gainesville, FL, 32608, USA.
| |
Collapse
|
34
|
Biased M1 muscarinic receptor mutant mice show accelerated progression of prion neurodegenerative disease. Proc Natl Acad Sci U S A 2021; 118:2107389118. [PMID: 34893539 PMCID: PMC8685681 DOI: 10.1073/pnas.2107389118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/14/2023] Open
Abstract
The M1 muscarinic acetylcholine receptor (M1-receptor) plays a crucial role in learning and memory and is a validated drug target for the treatment of Alzheimer’s disease (AD). Furthermore, M1-receptor ligands have been demonstrated to display disease-modifying effects in preclinical models of neurodegenerative disease. By employing a genetic mouse model expressing a G protein–biased M1-receptor in combination with a mouse model of terminal neurodegenerative disease, we demonstrate here that the M1-receptor exerts an inherent neuroprotective activity that is dependent on its phosphorylation status. Thus, in AD drug development programs, M1-receptor ligands that maintain the receptor phosphorylation status will be more likely to lead to beneficial neuroprotective outcomes. There are currently no treatments that can slow the progression of neurodegenerative diseases, such as Alzheimer’s disease (AD). There is, however, a growing body of evidence that activation of the M1 muscarinic acetylcholine receptor (M1-receptor) can not only restore memory loss in AD patients but in preclinical animal models can also slow neurodegenerative disease progression. The generation of an effective medicine targeting the M1-receptor has however been severely hampered by associated cholinergic adverse responses. By using genetically engineered mouse models that express a G protein–biased M1-receptor, we recently established that M1-receptor mediated adverse responses can be minimized by ensuring activating ligands maintain receptor phosphorylation/arrestin-dependent signaling. Here, we use these same genetic models in concert with murine prion disease, a terminal neurodegenerative disease showing key hallmarks of AD, to establish that phosphorylation/arrestin-dependent signaling delivers neuroprotection that both extends normal animal behavior and prolongs the life span of prion-diseased mice. Our data point to an important neuroprotective property inherent to the M1-receptor and indicate that next generation M1-receptor ligands designed to drive receptor phosphorylation/arrestin-dependent signaling would potentially show low adverse responses while delivering neuroprotection that will slow disease progression.
Collapse
|
35
|
Cherkasov NS, Kolykhalov IV. [Non-cognitive psychopathological symptoms in mild cognitive impairment]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:41-51. [PMID: 34870913 DOI: 10.17116/jnevro202112110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Noncognitive psychopathological symptoms (NPS) in elderly patients are increasingly attracting the attention of researchers in the field of neurodegenerative diseases. The question is raised whether these symptoms are risk factors or initial manifestations of the neurodegeneration process. This article provides information on the prevalence of late-onset NPS together with mild cognitive impairment (MCI), combination of which reflects the risk of developing dementia. The characteristic of mild behavioral impairment syndrome, which is currently used along with the concept of MCI, is given. The authors summarized data of the studies published over the past 10 years on the effect of NPS on the progression of cognitive impairment. Topics related to the differential diagnosis of these disorders, as well as existing approaches to treatment, are considered.
Collapse
|
36
|
Bago Rožanković P, Rožanković M, Badžak J, Stojić M, Šušak Sporiš I. Impact of Donepezil and Memantine on Behavioral and Psychological Symptoms of Alzheimer Disease: Six-month Open-label Study. Cogn Behav Neurol 2021; 34:288-294. [PMID: 34851866 DOI: 10.1097/wnn.0000000000000285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/01/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Behavioral and psychological symptoms of dementia (BPSD) are common in individuals with Alzheimer disease (AD). Donepezil and memantine are both widely used for the treatment of moderate AD. OBJECTIVE To evaluate the effects of donepezil and memantine in relieving BPSD in individuals with moderate AD. METHOD We conducted a prospective, randomized, 6-month clinical trial involving 85 individuals with moderate AD divided into two groups: group 1 (n = 42) was treated with donepezil; group 2 (n = 43) was treated with memantine. We used the Neuropsychiatric Inventory (NPI) to assess the prevalence and severity of BPSD at baseline and after 6 months of treatment with donepezil or memantine. RESULTS The two groups' baseline characteristics, including age, sex, mean length of education, and disease duration, were comparable, as were their baseline Mini-Mental State Examination scores. The NPI Total score improved from baseline to month 6 in both groups (P < 0.0001). Analyses of the NPI subdomains revealed that both donepezil treatment and memantine treatment produced statistically significant improvement in all of the NPI domains except euphoria and apathy, for which no improvement was observed after memantine treatment. Both treatments were well tolerated, with mostly mild and transient adverse effects. CONCLUSION Specific drugs for AD, including donepezil and memantine, may be effective in treating BPSD in individuals with moderate AD, with a favorable safety profile.
Collapse
|
37
|
Brown AJH, Bradley SJ, Marshall FH, Brown GA, Bennett KA, Brown J, Cansfield JE, Cross DM, de Graaf C, Hudson BD, Dwomoh L, Dias JM, Errey JC, Hurrell E, Liptrot J, Mattedi G, Molloy C, Nathan PJ, Okrasa K, Osborne G, Patel JC, Pickworth M, Robertson N, Shahabi S, Bundgaard C, Phillips K, Broad LM, Goonawardena AV, Morairty SR, Browning M, Perini F, Dawson GR, Deakin JFW, Smith RT, Sexton PM, Warneck J, Vinson M, Tasker T, Tehan BG, Teobald B, Christopoulos A, Langmead CJ, Jazayeri A, Cooke RM, Rucktooa P, Congreve MS, Weir M, Tobin AB. From structure to clinic: Design of a muscarinic M1 receptor agonist with potential to treatment of Alzheimer's disease. Cell 2021; 184:5886-5901.e22. [PMID: 34822784 PMCID: PMC7616177 DOI: 10.1016/j.cell.2021.11.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 04/29/2021] [Accepted: 11/01/2021] [Indexed: 12/31/2022]
Abstract
Current therapies for Alzheimer's disease seek to correct for defective cholinergic transmission by preventing the breakdown of acetylcholine through inhibition of acetylcholinesterase, these however have limited clinical efficacy. An alternative approach is to directly activate cholinergic receptors responsible for learning and memory. The M1-muscarinic acetylcholine (M1) receptor is the target of choice but has been hampered by adverse effects. Here we aimed to design the drug properties needed for a well-tolerated M1-agonist with the potential to alleviate cognitive loss by taking a stepwise translational approach from atomic structure, cell/tissue-based assays, evaluation in preclinical species, clinical safety testing, and finally establishing activity in memory centers in humans. Through this approach, we rationally designed the optimal properties, including selectivity and partial agonism, into HTL9936-a potential candidate for the treatment of memory loss in Alzheimer's disease. More broadly, this demonstrates a strategy for targeting difficult GPCR targets from structure to clinic.
Collapse
Affiliation(s)
- Alastair J H Brown
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Sophie J Bradley
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK; The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Fiona H Marshall
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Giles A Brown
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Kirstie A Bennett
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Jason Brown
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Julie E Cansfield
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - David M Cross
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK; Cross Pharma Consulting Ltd, 20-22 Wenlock Road, London, N17GU, UK
| | - Chris de Graaf
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Brian D Hudson
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Louis Dwomoh
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - João M Dias
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - James C Errey
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Edward Hurrell
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Jan Liptrot
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Giulio Mattedi
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Colin Molloy
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Pradeep J Nathan
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK; Brain Mapping Unit, University of Cambridge, Department of Psychiatry, Herchel Smith Building, Cambridge, CB20SZ, UK
| | - Krzysztof Okrasa
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Greg Osborne
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Jayesh C Patel
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Mark Pickworth
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Nathan Robertson
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Shahram Shahabi
- Eli Lilly & Co, Neuroscience Discovery, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Christoffer Bundgaard
- Eli Lilly & Co, Neuroscience Discovery, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK; H. Lundbeck A/S, Neuroscience Research, Ottiliavej 9, 2500 Valby, Copenhagen, Denmark
| | - Keith Phillips
- Eli Lilly & Co, Neuroscience Discovery, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Lisa M Broad
- Eli Lilly & Co, Neuroscience Discovery, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Anushka V Goonawardena
- Center for Neuroscience, Biosciences Division, SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Stephen R Morairty
- Center for Neuroscience, Biosciences Division, SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Michael Browning
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX12JD, UK; P1vital, Manor house, Howbery Buisness Park, Wallingford, OX108BA, UK
| | - Francesca Perini
- Centre for Cognitive Neuroscience - Duke-NUS Medical School, 8 College Road, 169857, Singapore
| | - Gerard R Dawson
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX12JD, UK
| | - John F W Deakin
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, M139PT UK
| | - Robert T Smith
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville 3052, Victoria, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Julie Warneck
- Protogenia Consulting Ltd, PO-Box 289, Ely, CB79DR, UK
| | - Mary Vinson
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Tim Tasker
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Benjamin G Tehan
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Barry Teobald
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville 3052, Victoria, Australia
| | - Christopher J Langmead
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville 3052, Victoria, Australia
| | - Ali Jazayeri
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Robert M Cooke
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Prakash Rucktooa
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Miles S Congreve
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK
| | - Malcolm Weir
- Sosei-Heptares, Steinmetz Building, Granta Park, Cambridge, CB21 6DG, UK.
| | - Andrew B Tobin
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
38
|
Gunawardena IPC, Retinasamy T, Shaikh MF. Is Aducanumab for LMICs? Promises and Challenges. Brain Sci 2021; 11:1547. [PMID: 34827546 PMCID: PMC8615623 DOI: 10.3390/brainsci11111547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/02/2022] Open
Abstract
Aducanumab, a human monoclonal antibody, was approved in June of 2021 as the first disease-modifying treatment for Alzheimer's disease by the United States Food and Drug Administration (U.S. FDA). A substantial proportion of patients with Alzheimer's disease live in low- and middle-income countries (LMICs), and the debilitating effects of this disease exerts burdens on patients and caregivers in addition to the significant economic strains many nations bear. While the advantages of a disease-modifying therapy are clear in delaying the progression of disease to improve patient outcomes, aducanumab's approval by the U.S. FDA was met with controversy for a plethora of reasons. This paper will provide precursory insights into aducanumab's role, appropriateness, and cost-effectiveness in low- and middle-income countries. We extend some of the controversies associated with aducanumab, including the contradicting evidence from the two trials (EMERGE and ENGAGE) and the resources required to deliver the treatment safely and effectively to patients, among other key considerations.
Collapse
Affiliation(s)
- Illangage P. C. Gunawardena
- Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Johor, Malaysia;
| | - Thaarvena Retinasamy
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia;
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia;
| |
Collapse
|
39
|
Pople CB, Meng Y, Li DZ, Bigioni L, Davidson B, Vecchio LM, Hamani C, Rabin JS, Lipsman N. Neuromodulation in the Treatment of Alzheimer's Disease: Current and Emerging Approaches. J Alzheimers Dis 2021; 78:1299-1313. [PMID: 33164935 DOI: 10.3233/jad-200913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neuromodulation as a treatment strategy for psychiatric and neurological diseases has grown in popularity in recent years, with the approval of repetitive transcranial magnetic stimulation (rTMS) for the treatment of depression being one such example. These approaches offer new hope in the treatment of diseases that have proven largely intractable to traditional pharmacological approaches. For this reason, neuromodulation is increasingly being explored for the treatment of Alzheimer's disease. However, such approaches have variable, and, in many cases, very limited evidence for safety and efficacy, with most human evidence obtained in small clinical trials. Here we review work in animal models and humans with Alzheimer's disease exploring emerging neuromodulation modalities. Approaches reviewed include deep brain stimulation, transcranial magnetic stimulation, transcranial electrical stimulation, ultrasound stimulation, photobiomodulation, and visual or auditory stimulation. In doing so, we clarify the current evidence for these approaches in treating Alzheimer's disease and identify specific areas where additional work is needed to facilitate their clinical translation.
Collapse
Affiliation(s)
- Christopher B Pople
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ying Meng
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Daniel Z Li
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Luca Bigioni
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Benjamin Davidson
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Laura M Vecchio
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Jennifer S Rabin
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medicine (Neurology), University of Toronto, Toronto, Ontario, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto ON, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
40
|
Martinez JL, Zammit MD, West NR, Christian BT, Bhattacharyya A. Basal Forebrain Cholinergic Neurons: Linking Down Syndrome and Alzheimer's Disease. Front Aging Neurosci 2021; 13:703876. [PMID: 34322015 PMCID: PMC8311593 DOI: 10.3389/fnagi.2021.703876] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022] Open
Abstract
Down syndrome (DS, trisomy 21) is characterized by intellectual impairment at birth and Alzheimer's disease (AD) pathology in middle age. As individuals with DS age, their cognitive functions decline as they develop AD pathology. The susceptibility to degeneration of a subset of neurons, known as basal forebrain cholinergic neurons (BFCNs), in DS and AD is a critical link between cognitive impairment and neurodegeneration in both disorders. BFCNs are the primary source of cholinergic innervation to the cerebral cortex and hippocampus, as well as the amygdala. They play a critical role in the processing of information related to cognitive function and are directly engaged in regulating circuits of attention and memory throughout the lifespan. Given the importance of BFCNs in attention and memory, it is not surprising that these neurons contribute to dysfunctional neuronal circuitry in DS and are vulnerable in adults with DS and AD, where their degeneration leads to memory loss and disturbance in language. BFCNs are thus a relevant cell target for therapeutics for both DS and AD but, despite some success, efforts in this area have waned. There are gaps in our knowledge of BFCN vulnerability that preclude our ability to effectively design interventions. Here, we review the role of BFCN function and degeneration in AD and DS and identify under-studied aspects of BFCN biology. The current gaps in BFCN relevant imaging studies, therapeutics, and human models limit our insight into the mechanistic vulnerability of BFCNs in individuals with DS and AD.
Collapse
Affiliation(s)
- Jose L. Martinez
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, United States
- Waisman Center, University of Wisconsin, Madison, WI, United States
| | - Matthew D. Zammit
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Nicole R. West
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, United States
- Waisman Center, University of Wisconsin, Madison, WI, United States
| | - Bradley T. Christian
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
- Department of Psychiatry, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
41
|
Adib M, Islam R, Ahsan M, Rahman A, Hossain M, Rahman MM, Alshehri SM, Kazi M, Mazid MA. Cholinesterase inhibitory activity of tinosporide and 8-hydroxytinosporide isolated from Tinospora cordifolia: In vitro and in silico studies targeting management of Alzheimer's disease. Saudi J Biol Sci 2021; 28:3893-3900. [PMID: 34220245 PMCID: PMC8241625 DOI: 10.1016/j.sjbs.2021.03.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Tinosporide and 8-hydroxytinosporide isolated from Tinospora cordifolia were evaluated for acetylcholinesterase (AChE) and butylcholinesterase (BuChE) inhibitory activities. The structure of the compound was confirmed by spectroscopic analysis, whereas cholinesterase inhibition was investigated by Ellman method using donepezil as standard drug and the data were presented as IC50 (μg/ml ± SEM). Furthermore, donepezil, tinosporide and 8-hydroxytinosporide were executed for docking analysis. The results from the isolated compounds TC-16R confirmed as tinosporide promisingly inhibited AChE with IC50 value of 13.45 ± 0.144, whereas TC-19R confirmed as 8-hydroxytinosporide moderately inhibited AChE with IC50 value of 46.71 ± 0.511. In case of BuChE inhibition, the IC50 values were found to be 408.50 ± 17.197 and 317.26 ± 6.918 for tinosporide and 8-hydroxytinosporide, respectively. The in silico studies revealed that the ligand tinosporide fit with the binding sites and inhibited AChE. Overall, the study findings suggested that tinosporide would be a complementary noble molecule of donepezil which is correlated with its pharmacological activity through in vitro studies, while 8-hydroxytinosporide modestly inhibited BuChE and the results are very close to the standard donepezil.
Collapse
Affiliation(s)
- Mohiminul Adib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Rashedul Islam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Monira Ahsan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Arifur Rahman
- Department of Pharmacy, Jagannath University, Dhaka 1100, Bangladesh
| | - Mahmud Hossain
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | | | - Sultan M. Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Pharmaceutical Sciences, College of Pharmacy, Almaarefa University, Riyadh 11597, Saudi Arabia
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Md Abdul Mazid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
42
|
Proulx É, Power SK, Oliver DK, Sargin D, McLaurin J, Lambe EK. Apamin Improves Prefrontal Nicotinic Impairment in Mouse Model of Alzheimer's Disease. Cereb Cortex 2021; 30:563-574. [PMID: 31188425 DOI: 10.1093/cercor/bhz107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/11/2022] Open
Abstract
Disruption of attention is an early and disabling symptom of Alzheimer's disease (AD). The underlying cellular mechanisms are poorly understood and treatment options for patients are limited. These early attention deficits are evident in the TgCRND8 mouse, a well-established murine model of AD that recapitulates several features of the disease. Here, we report severe impairment of the nicotinic receptor-mediated excitation of prefrontal attentional circuitry in TgCRND8 mice relative to wild-type littermate controls. We demonstrate that this impairment can be remedied by apamin, a bee venom neurotoxin peptide that acts as a selective antagonist to the SK family of calcium-sensitive potassium channels. We probe this seeming upregulation of calcium-sensitive inhibition and find that the attenuated nicotinic firing rates in TgCRND8 attention circuits are mediated neither by greater cellular calcium signals nor by elevated SK channel expression. Instead, we find that TgCRND8 mice show enhanced functional coupling of nicotinic calcium signals to inhibition. This SK-mediated inhibition exerts a powerful negative feedback on nicotinic excitation, dampening attention-relevant signaling in the TgCRND8 brain. These mechanistic findings identify a new cellular target involved in the modulation of attention and a novel therapeutic target for early attention deficits in AD.
Collapse
Affiliation(s)
- É Proulx
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - S K Power
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - D K Oliver
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - D Sargin
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - J McLaurin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Biological Sciences and Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada M4N 3M5
| | - E K Lambe
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada M5G 1E2.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada M5T 1R8
| |
Collapse
|
43
|
Halminen O, Vesikansa A, Mehtälä J, Hörhammer I, Mikkola T, Virta LJ, Ylisaukko-Oja T, Linna M. Early Start of Anti-Dementia Medication Delays Transition to 24-Hour Care in Alzheimer's Disease Patients: A Finnish Nationwide Cohort Study. J Alzheimers Dis 2021; 81:1103-1115. [PMID: 33843673 DOI: 10.3233/jad-201502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dementia is one of the strongest predictors of admission to a 24-hour care facility among older people, and 24-hour care is the major cost of Alzheimer's disease (AD). OBJECTIVE The aim of this study was to evaluate the association of early start of anti-dementia medication and other predisposing factors with 2-year risk of transition to 24-hour care in the nationwide cohort of Finnish AD patients. METHODS This was a retrospective, non-interventional study based on individual-level data from Finnish national health and social care registers. The incident cohort included 7,454 AD patients (ICD-10, G30) comprised of two subgroups: those living unassisted at home (n = 5,002), and those receiving professional home care (n = 2,452). The primary outcome was admission to a 24-hour care facility. Exploratory variables were early versus late anti-dementia medication start, sociodemographic variables, care intensity level, and comorbidities. RESULTS Early anti-dementia medication reduced the risk of admission to 24-hour care both in patients living unassisted at home, with a hazard ratio (HR) of 0.58 (p < 0.001), and those receiving professional home care (HR, 0.84; p = 0.039). Being unmarried (HR, 1.69; p < 0.001), having an informal caregiver (HR, 1.69; p = 0.003), or having a diagnosis of additional neurological disorder (HR, 1.68; p = 0.006) or hip fracture (HR, 1.61; p = 0.004) were associated with higher risk of admission to 24-hour care in patients living unassisted at home. CONCLUSION To support living at home, early start of anti-dementia medication should be a high priority in newly diagnosed AD patients.
Collapse
Affiliation(s)
- Olli Halminen
- Department of Industrial Engineering and Management, School of Science, Aalto University, Espoo, Finland
| | | | | | - Iiris Hörhammer
- Department of Industrial Engineering and Management, School of Science, Aalto University, Espoo, Finland
| | | | | | - Tero Ylisaukko-Oja
- MedEngine Oy, Helsinki, Finland.,Faculty of Medicine, Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Miika Linna
- Department of Industrial Engineering and Management, School of Science, Aalto University, Espoo, Finland
| |
Collapse
|
44
|
Riedel G, Klein J, Niewiadomska G, Kondak C, Schwab K, Lauer D, Magbagbeolu M, Steczkowska M, Zadrozny M, Wydrych M, Cranston A, Melis V, Santos RX, Theuring F, Harrington CR, Wischik CM. Mechanisms of Anticholinesterase Interference with Tau Aggregation Inhibitor Activity in a Tau-Transgenic Mouse Model. Curr Alzheimer Res 2021; 17:285-296. [PMID: 32091331 PMCID: PMC7403648 DOI: 10.2174/1567205017666200224120926] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/08/2020] [Accepted: 02/12/2020] [Indexed: 01/18/2023]
Abstract
Background Symptomatic treatments of Alzheimer’s Disease (AD) with cholinesterase inhibitors and/or memantine are relatively ineffective and there is a need for new treatments targeting the underlying pathology of AD. In most of the failed disease-modifying trials, patients have been allowed to continue taking symptomatic treatments at stable doses, under the assumption that they do not impair efficacy. In recently completed Phase 3 trials testing the tau aggregation inhibitor leuco-methylthioninium bis (hydromethane-sulfonate) (LMTM), we found significant differences in treatment response according to whether patients were taking LMTM either as monotherapy or as an add-on to symptomatic treatments. Methods We have examined the effect of either LMTM alone or chronic rivastigmine prior to LMTM treatment of tau transgenic mice expressing the short tau fragment that constitutes the tangle filaments of AD. We have measured acetylcholine levels, synaptosomal glutamate release, synaptic proteins, mitochondrial complex IV activity, tau pathology and Choline Acetyltransferase (ChAT) immunoreactivity. Results LMTM given alone increased hippocampal Acetylcholine (ACh) levels, glutamate release from synaptosomal preparations, synaptophysin levels in multiple brain regions and mitochondrial complex IV activity, reduced tau pathology, partially restored ChAT immunoreactivity in the basal forebrain and reversed deficits in spatial learning. Chronic pretreatment with rivastigmine was found to reduce or eliminate almost all these effects, apart from a reduction in tau aggregation pathology. LMTM effects on hippocampal ACh and synaptophysin levels were also reduced in wild-type mice. Conclusion The interference with the pharmacological activity of LMTM by a cholinesterase inhibitor can be reproduced in a tau transgenic mouse model and, to a lesser extent, in wild-type mice. Long-term pretreatment with a symptomatic drug alters a broad range of brain responses to LMTM across different transmitter systems and cellular compartments at multiple levels of brain function. There is, therefore, no single locus for the negative interaction. Rather, the chronic neuronal activation induced by reducing cholinesterase function produces compensatory homeostatic downregulation in multiple neuronal systems. This reduces a broad range of treatment responses to LMTM associated with a reduction in tau aggregation pathology. Since the interference is dictated by homeostatic responses to prior symptomatic treatment, it is likely that there would be similar interference with other drugs tested as add-on to the existing symptomatic treatment, regardless of the intended therapeutic target or mode of action. The present findings outline key results that now provide a working model to explain interference by symptomatic treatment.
Collapse
Affiliation(s)
- Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Jochen Klein
- Department of Pharmacology and Clinical Pharmacy, Goethe University, Max-von-Laue Str. 9, Frankfurt, 60438 Frankfurt am Main, Germany
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Constantin Kondak
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.,Department of Pharmacology and Clinical Pharmacy, Goethe University, Max-von-Laue Str. 9, Frankfurt, 60438 Frankfurt am Main, Germany
| | - Karima Schwab
- Charite-Institute of Pharmacology, Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Dilyara Lauer
- Charite-Institute of Pharmacology, Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Mandy Magbagbeolu
- Charite-Institute of Pharmacology, Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Marta Steczkowska
- Mossakowski Medical Research Centre, 5 Pawinski Street, 02-106 Warsaw, Poland
| | - Maciej Zadrozny
- Mossakowski Medical Research Centre, 5 Pawinski Street, 02-106 Warsaw, Poland
| | - Malgorzata Wydrych
- Mossakowski Medical Research Centre, 5 Pawinski Street, 02-106 Warsaw, Poland
| | - Anna Cranston
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Valeria Melis
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Renato X Santos
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Franz Theuring
- Charite-Institute of Pharmacology, Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.,TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, United Kingdom
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.,TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, United Kingdom
| |
Collapse
|
45
|
Isla AG, Balleza-Tapia H, Fisahn A. Efficacy of preclinical pharmacological interventions against alterations of neuronal network oscillations in Alzheimer's disease: A systematic review. Exp Neurol 2021; 343:113743. [PMID: 34000250 DOI: 10.1016/j.expneurol.2021.113743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 12/29/2022]
Abstract
Despite the development of multiple pharmacological approaches over the years aimed at treating Alzheimer's Disease (AD) only very few have been approved for clinical use in patients. To date there still exists no disease-modifying treatment that could prevent or rescue the cognitive impairment, particularly of memory aquisition, that is characteristic of AD. One of the possibilities for this state of affairs might be that the majority of drug discovery efforts focuses on outcome measures of decreased neuropathological biomarkers characteristic of AD, without taking into acount neuronal processes essential to the generation and maintenance of memory processes. Particularly, the capacity of the brain to generate theta (θ) and gamma (γ) oscillatory activity has been strongly correlated to memory performance. Using a systematic review approach, we synthesize the existing evidence in the literature on pharmacological interventions that enhance neuronal theta (θ) and/or gamma (γ) oscillations in non-pathological animal models and in AD animal models. Additionally, we synthesize the main outcomes and neurochemical systems targeted. We propose that functional biomarkers such as cognition-relevant neuronal network oscillations should be used as outcome measures during the process of research and development of novel drugs against cognitive impairment in AD.
Collapse
Affiliation(s)
- Arturo G Isla
- Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Akademiska Stråket 1, J10:30, 17164 Solna, Stockholm, Sweden
| | - Hugo Balleza-Tapia
- Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Akademiska Stråket 1, J10:30, 17164 Solna, Stockholm, Sweden
| | - André Fisahn
- Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Akademiska Stråket 1, J10:30, 17164 Solna, Stockholm, Sweden.
| |
Collapse
|
46
|
Howard R, Gathercole R, Bradley R, Harper E, Davis L, Pank L, Lam N, Talbot E, Hooper E, Winson R, Scutt B, Ordonez Montano V, Nunn S, Lavelle G, Bateman A, Bentham P, Burns A, Dunk B, Forsyth K, Fox C, Poland F, Leroi I, Newman S, O’Brien J, Henderson C, Knapp M, Woolham J, Gray R. The effectiveness and cost-effectiveness of assistive technology and telecare for independent living in dementia: a randomised controlled trial. Age Ageing 2021; 50:882-890. [PMID: 33492349 PMCID: PMC8099012 DOI: 10.1093/ageing/afaa284] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 11/14/2022] Open
Abstract
Objectives The use of assistive technology and telecare (ATT) has been promoted to manage risks associated with independent living in people with dementia but with little evidence for effectiveness. Methods Participants were randomly assigned to receive an ATT assessment followed by installation of all appropriate ATT devices or limited control of appropriate ATT. The primary outcomes were time to institutionalisation and cost-effectiveness. Key secondary outcomes were number of incidents involving risks to safety, burden and stress in family caregivers and quality of life. Results Participants were assigned to receive full ATT (248 participants) or the limited control (247 participants). After adjusting for baseline imbalance of activities of daily living score, HR for median pre-institutionalisation survival was 0.84; 95% CI, 0.63 to 1.12; P = 0.20. There were no significant differences between arms in health and social care (mean -£909; 95% CI, -£5,336 to £3,345, P = 0.678) and societal costs (mean -£3,545; 95% CI, -£13,914 to £6,581, P = 0.499). ATT group members had reduced participant-rated quality-adjusted life years (QALYs) at 104 weeks (mean − 0.105; 95% CI, −0.204 to −0.007, P = 0.037) but did not differ in QALYs derived from proxy-reported EQ-5D. Discussion Fidelity of the intervention was low in terms of matching ATT assessment, recommendations and installation. This, however, reflects current practice within adult social care in England. Conclusions Time living independently outside a care home was not significantly longer in participants who received full ATT and ATT was not cost-effective. Participants with full ATT attained fewer QALYs based on participant-reported EQ-5D than controls at 104 weeks.
Collapse
Affiliation(s)
| | | | - Rosie Bradley
- Medical Research Council Population Health Research Unit, UK
| | - Emma Harper
- Medical Research Council Population Health Research Unit, UK
| | - Lucy Davis
- Medical Research Council Population Health Research Unit, UK
| | - Lynn Pank
- Medical Research Council Population Health Research Unit, UK
| | - Natalie Lam
- Medical Research Council Population Health Research Unit, UK
| | - Emma Talbot
- Norfolk and Suffolk NHS Foundation Trust, UK
| | - Emma Hooper
- Lancashire Care NHS Foundation Trust and University of Manchester, UK
| | - Rachel Winson
- Cambridgeshire and Peterborough NHS Foundation Trust, UK
| | | | | | | | | | | | - Peter Bentham
- Birmingham and Solihull Mental Health NHS Foundation Trust, UK
| | | | - Barbara Dunk
- South London and Maudsley NHS Foundation Trust, UK
| | | | | | | | | | | | | | | | - Martin Knapp
- London School of Economics and Political Science, UK
| | | | - Richard Gray
- Medical Research Council Population Health Research Unit, UK
| |
Collapse
|
47
|
Xu H, Garcia-Ptacek S, Jönsson L, Wimo A, Nordström P, Eriksdotter M. Long-term Effects of Cholinesterase Inhibitors on Cognitive Decline and Mortality. Neurology 2021; 96:e2220-e2230. [PMID: 33741639 PMCID: PMC8166426 DOI: 10.1212/wnl.0000000000011832] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Objective To investigate whether cholinesterase inhibitors (ChEIs) are associated with slower cognitive decline in Alzheimer dementia and decreased risk of severe dementia or death. Methods Patients with Alzheimer dementia from the Swedish Dementia Registry starting on ChEIs within 3 months of the dementia diagnosis were included and compared to nontreated patients with Alzheimer dementia. In a propensity score–matched cohort, the association between ChEI use and cognitive trajectories assessed by Mini-Mental State Examination (MMSE) scores was examined with a mixed model, and severe dementia (MMSE score <10) or death as an outcome was assessed with Cox proportional hazards models. Results The matched cohort included 11,652 ChEI users and 5,826 nonusers. During an average of 5 years of follow-up, 255 cases developed severe dementia, and 6,055 (35%) died. ChEI use was associated with higher MMSE score at each visit (0.13 MMSE points per year; 95% confidence interval [CI] 0.06–0.20). ChEI users had a 27% lower risk of death (0.73, 95% CI 0.69–0.77) compared with nonusers. Galantamine was associated with lower risk of death (0.71, 95% CI 0.65–0.76) and lower risk of severe dementia (0.69, 95% CI 0.47–1.00) and had the strongest effect on cognitive decline of all the ChEIs (0.18 MMSE points per year, 95% CI 0.07–0.28). Conclusions ChEIs are associated with cognitive benefits that are modest but persist over time and with reduced mortality risk, which could be explained partly by their cognitive effects. Galantamine was the only ChEI demonstrating a significant reduction in the risk of developing severe dementia. Classification of Evidence This study provides Class III evidence that for patients with Alzheimer dementia ChEIs decrease long-term cognitive decline and risk of death and that galantamine decreases the risk of severe dementia.
Collapse
Affiliation(s)
- Hong Xu
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden.
| | - Sara Garcia-Ptacek
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden
| | - Linus Jönsson
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden
| | - Anders Wimo
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden
| | - Peter Nordström
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Division of Neurogeriatrics (L.J., A.W.), Department of Neurobiology, Care Sciences and Society, and Department of Medical Epidemiology and Biostatistics (H.X.), Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset, Stockholm, Sweden; H. Lundbeck A/S (L.J.), Copenhagen, Denmark; Department of Community Medicine and Rehabilitation (P.N.), Geriatric Medicine, Umeå University; and Theme Aging (S.G.-P., M.A.), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
48
|
Gathercole R, Bradley R, Harper E, Davies L, Pank L, Lam N, Davies A, Talbot E, Hooper E, Winson R, Scutt B, Montano VO, Nunn S, Lavelle G, Lariviere M, Hirani S, Brini S, Bateman A, Bentham P, Burns A, Dunk B, Forsyth K, Fox C, Henderson C, Knapp M, Leroi I, Newman S, O'Brien J, Poland F, Woolham J, Gray R, Howard R. Assistive technology and telecare to maintain independent living at home for people with dementia: the ATTILA RCT. Health Technol Assess 2021; 25:1-156. [PMID: 33755548 DOI: 10.3310/hta25190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Assistive technology and telecare have been promoted to manage the risks associated with independent living for people with dementia, but there is limited evidence of their effectiveness. OBJECTIVES This trial aimed to establish whether or not assistive technology and telecare assessments and interventions extend the time that people with dementia can continue to live independently at home and whether or not they are cost-effective. Caregiver burden, the quality of life of caregivers and of people with dementia and whether or not assistive technology and telecare reduce safety risks were also investigated. DESIGN This was a pragmatic, randomised controlled trial. Blinding was not undertaken as it was not feasible to do so. All consenting participants were included in an intention-to-treat analysis. SETTING This trial was set in 12 councils in England with adult social services responsibilities. PARTICIPANTS Participants were people with dementia living in the community who had an identified need that might benefit from assistive technology and telecare. INTERVENTIONS Participants were randomly assigned to receive either assistive technology and telecare recommended by a health or social care professional to meet their assessed needs (a full assistive technology and telecare package) or a pendant alarm, non-monitored smoke and carbon monoxide detectors and a key safe (a basic assistive technology and telecare package). MAIN OUTCOME MEASURES The primary outcomes were time to admission to care and cost-effectiveness. Secondary outcomes assessed caregivers using the 10-item Center for Epidemiological Studies Depression Scale, the State-Trait Anxiety Inventory 6-item scale and the Zarit Burden Interview. RESULTS Of 495 participants, 248 were randomised to receive full assistive technology and telecare and 247 received the limited control. Comparing the assistive technology and telecare group with the control group, the hazard ratio for institutionalisation was 0.76 (95% confidence interval 0.58 to 1.01; p = 0.054). After adjusting for an imbalance in the baseline activities of daily living score between trial arms, the hazard ratio was 0.84 (95% confidence interval 0.63 to 1.12; p = 0.20). At 104 weeks, there were no significant differences between groups in health and social care resource use costs (intervention group - control group difference: mean -£909, 95% confidence interval -£5336 to £3345) or in societal costs (intervention group - control group difference: mean -£3545; 95% confidence interval -£13,914 to £6581). At 104 weeks, based on quality-adjusted life-years derived from the participant-rated EuroQol-5 Dimensions questionnaire, the intervention group had 0.105 (95% confidence interval -0.204 to -0.007) fewer quality-adjusted life-years than the control group. The number of quality-adjusted life-years derived from the proxy-rated EuroQol-5 Dimensions questionnaire did not differ between groups. Caregiver outcomes did not differ between groups over 24 weeks. LIMITATIONS Compliance with the assigned trial arm was variable, as was the quality of assistive technology and telecare needs assessments. Attrition from assessments led to data loss additional to that attributable to care home admission and censoring events. CONCLUSIONS A full package of assistive technology and telecare did not increase the length of time that participants with dementia remained in the community, and nor did it decrease caregiver burden, depression or anxiety, relative to a basic package of assistive technology and telecare. Use of the full assistive technology and telecare package did not increase participants' health and social care or societal costs. Quality-adjusted life-years based on participants' EuroQol-5 Dimensions questionnaire responses were reduced in the intervention group compared with the control group; groups did not differ in the number of quality-adjusted life-years based on the proxy-rated EuroQol-5 Dimensions questionnaire. FUTURE WORK Future work could examine whether or not improved assessment that is more personalised to an individual is beneficial. TRIAL REGISTRATION Current Controlled Trials ISRCTN86537017. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 25, No. 19. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
| | - Rosie Bradley
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Emma Harper
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Lucy Davies
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Lynn Pank
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Natalie Lam
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Anna Davies
- School of Health Sciences, City, University of London, London, UK.,Population Health Sciences, University of Bristol, Bristol, UK
| | - Emma Talbot
- Norfolk and Suffolk NHS Foundation Trust, Stowmarket, UK
| | - Emma Hooper
- Lancashire Care NHS Foundation Trust, Preston, UK.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rachel Winson
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Bethany Scutt
- Department of Old Age Psychiatry, King's College London, London, UK
| | | | - Samantha Nunn
- Cambridgeshire Community Services NHS Trust, Cambridge, UK
| | - Grace Lavelle
- Department of Old Age Psychiatry, King's College London, London, UK
| | - Matthew Lariviere
- Centre for International Research on Care, Labour and Equalities, University of Sheffield, Sheffield, UK
| | | | - Stefano Brini
- School of Health Sciences, City, University of London, London, UK
| | - Andrew Bateman
- School of Health and Social Care, University of Essex, Colchester, UK
| | - Peter Bentham
- Birmingham and Solihull Mental Health NHS Foundation Trust, Birmingham, UK
| | - Alistair Burns
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Barbara Dunk
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Kirsty Forsyth
- School of Health Sciences, Queen Margaret University, Edinburgh, UK
| | - Chris Fox
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Catherine Henderson
- Care Policy and Evaluation Centre, London School of Economics and Political Science, London, UK
| | - Martin Knapp
- Care Policy and Evaluation Centre, London School of Economics and Political Science, London, UK
| | - Iracema Leroi
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Stanton Newman
- School of Health Sciences, City, University of London, London, UK
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Fiona Poland
- School of Health Sciences, University of East Anglia, Norwich, UK
| | - John Woolham
- National Institute for Health Research (NIHR) Health & Social Care Workforce Research Unit, King's College London, London, UK
| | - Richard Gray
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
49
|
Sood A, Pavlik V, Darby E, Chan W, Doody R. Different Cognitive Profiles Are Associated with Progression Rate and Age at Death in Probable Alzheimer's Disease. J Alzheimers Dis 2021; 80:735-747. [PMID: 33579838 DOI: 10.3233/jad-201124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cognitive profiles characterized by primarily language or visuospatial deficits have been documented in individuals meeting diagnostic criteria for probable Alzheimer's disease (AD), but their association with progression rate or overall survival is not well described. OBJECTIVE To compare time from diagnosis to severe disease stage and death in probable AD patients classified into three groups based on neuropsychological test performance: marked verbal impairment (Verb-PI) with relatively preserved visuospatial function, marked visuospatial impairment with preserved verbal function (Vis-PI), and balanced verbal and visuospatial impairments (Bal-PI). METHODS This prospective cohort study included 540 probable AD patients attending an academic memory clinic who were enrolled from 1995-2013 and followed annually. Eligible individuals had a Mini-Mental State Exam (MMSE) score ≥10 at baseline, and at least one annual follow up visit. We used Cox proportional hazards modeling to analyze the association of cognitive profiles with time to decline in MMSE and CDR Global Score. RESULTS Sixty-one (11.3%) individuals had a Verb-PI profile, 86 (16%) had a Vis-PI profile, and 393 (72.8%) a Bal-PI profile. MMSE decline to <10 was faster in Verb-PI than Vis-PI (HR 2.004, 95%CI, 1.062-3.780; p = 0.032). Progression to CDR-GS = 3 was faster in Verb-PI individuals compared to Bal-PI (HR 1.604, 95%CI, 1.022-2.515; p = 0.040) or Vis-PI (HR 2.388, 95%CI, 1.330-4.288; p = 0.004) individuals. Baseline cognitive profile did not affect mortality. CONCLUSION A recognition of different AD profiles may help to personalize care by providing a better understanding of pathogenesis and expected progression.
Collapse
Affiliation(s)
- Ajay Sood
- Rush Alzheimer's Disease Center (RADC), Rush University Medical Center, Chicago, IL, USA
| | - Valory Pavlik
- Department of Neurology and Alzheimer's Disease and Memory Disorders Center, Baylor College of Medicine, Houston, TX, USA
| | - Eveleen Darby
- Department of Neurology and Alzheimer's Disease and Memory Disorders Center, Baylor College of Medicine, Houston, TX, USA
| | - Wenyaw Chan
- Department of Biostatistics and Data Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rachelle Doody
- Genentech, San Francisco, CA, USA.,Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
50
|
Parsons C, Lim WY, Loy C, McGuinness B, Passmore P, Ward SA, Hughes C. Withdrawal or continuation of cholinesterase inhibitors or memantine or both, in people with dementia. Cochrane Database Syst Rev 2021; 2:CD009081. [PMID: 35608903 PMCID: PMC8094886 DOI: 10.1002/14651858.cd009081.pub2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Dementia is a progressive syndrome characterised by deterioration in memory, thinking and behaviour, and by impaired ability to perform daily activities. Two classes of drug - cholinesterase inhibitors (donepezil, galantamine and rivastigmine) and memantine - are widely licensed for dementia due to Alzheimer's disease, and rivastigmine is also licensed for Parkinson's disease dementia. These drugs are prescribed to alleviate symptoms and delay disease progression in these and sometimes in other forms of dementia. There are uncertainties about the benefits and adverse effects of these drugs in the long term and in severe dementia, about effects of withdrawal, and about the most appropriate time to discontinue treatment. OBJECTIVES To evaluate the effects of withdrawal or continuation of cholinesterase inhibitors or memantine, or both, in people with dementia on: cognitive, neuropsychiatric and functional outcomes, rates of institutionalisation, adverse events, dropout from trials, mortality, quality of life and carer-related outcomes. SEARCH METHODS We searched the Cochrane Dementia and Cognitive Improvement Group's Specialised Register up to 17 October 2020 using terms appropriate for the retrieval of studies of cholinesterase inhibitors or memantine. The Specialised Register contains records of clinical trials identified from monthly searches of a number of major healthcare databases, numerous trial registries and grey literature sources. SELECTION CRITERIA We included all randomised, controlled clinical trials (RCTs) which compared withdrawal of cholinesterase inhibitors or memantine, or both, with continuation of the same drug or drugs. DATA COLLECTION AND ANALYSIS Two review authors independently assessed citations and full-text articles for inclusion, extracted data from included trials and assessed risk of bias using the Cochrane risk of bias tool. Where trials were sufficiently similar, we pooled data for outcomes in the short term (up to 2 months after randomisation), medium term (3-11 months) and long term (12 months or more). We assessed the overall certainty of the evidence for each outcome using GRADE methods. MAIN RESULTS We included six trials investigating cholinesterase inhibitor withdrawal, and one trial investigating withdrawal of either donepezil or memantine. No trials assessed withdrawal of memantine only. Drugs were withdrawn abruptly in five trials and stepwise in two trials. All participants had dementia due to Alzheimer's disease, with severities ranging from mild to very severe, and were taking cholinesterase inhibitors without known adverse effects at baseline. The included trials randomised 759 participants to treatment groups relevant to this review. Study duration ranged from 6 weeks to 12 months. There were too few included studies to allow planned subgroup analyses. We considered some studies to be at unclear or high risk of selection, performance, detection, attrition or reporting bias. Compared to continuing cholinesterase inhibitors, discontinuing treatment may be associated with worse cognitive function in the short term (standardised mean difference (SMD) -0.42, 95% confidence interval (CI) -0.64 to -0.21; 4 studies; low certainty), but the effect in the medium term is very uncertain (SMD -0.40, 95% CI -0.87 to 0.07; 3 studies; very low certainty). In a sensitivity analysis omitting data from a study which only included participants who had shown a relatively poor prior response to donepezil, inconsistency was reduced and we found that cognitive function may be worse in the discontinuation group in the medium term (SMD -0.62; 95% CI -0.94 to -0.31). Data from one longer-term study suggest that discontinuing a cholinesterase inhibitor is probably associated with worse cognitive function at 12 months (mean difference (MD) -2.09 Standardised Mini-Mental State Examination (SMMSE) points, 95% CI -3.43 to -0.75; moderate certainty). Discontinuation may make little or no difference to functional status in the short term (SMD -0.25, 95% CI -0.54 to 0.04; 2 studies; low certainty), and its effect in the medium term is uncertain (SMD -0.38, 95% CI -0.74 to -0.01; 2 studies; very low certainty). After 12 months, discontinuing a cholinesterase inhibitor probably results in greater functional impairment than continuing treatment (MD -3.38 Bristol Activities of Daily Living Scale (BADLS) points, 95% CI -6.67 to -0.10; one study; moderate certainty). Discontinuation may be associated with a worsening of neuropsychiatric symptoms over the short term and medium term, although we cannot exclude a minimal effect (SMD - 0.48, 95% CI -0.82 to -0.13; 2 studies; low certainty; and SMD -0.27, 95% CI -0.47 to -0.08; 3 studies; low certainty, respectively). Data from one study suggest that discontinuing a cholinesterase inhibitor may result in little to no change in neuropsychiatric status at 12 months (MD -0.87 Neuropsychiatric Inventory (NPI) points; 95% CI -8.42 to 6.68; moderate certainty). We found no clear evidence of an effect of discontinuation on dropout due to lack of medication efficacy or deterioration in overall medical condition (odds ratio (OR) 1.53, 95% CI 0.84 to 2.76; 4 studies; low certainty), on number of adverse events (OR 0.85, 95% CI 0.57 to 1.27; 4 studies; low certainty) or serious adverse events (OR 0.80, 95% CI 0.46 to 1.39; 4 studies; low certainty), and on mortality (OR 0.75, 95% CI 0.36 to 1.55; 5 studies; low certainty). Institutionalisation was reported in one trial, but it was not possible to extract data for the groups relevant to this review. AUTHORS' CONCLUSIONS This review suggests that discontinuing cholinesterase inhibitors may result in worse cognitive, neuropsychiatric and functional status than continuing treatment, although this is supported by limited evidence, almost all of low or very low certainty. As all participants had dementia due to Alzheimer's disease, our findings are not transferable to other dementia types. We were unable to determine whether the effects of discontinuing cholinesterase inhibitors differed with baseline dementia severity. There is currently no evidence to guide decisions about discontinuing memantine. There is a need for further well-designed RCTs, across a range of dementia severities and settings. We are aware of two ongoing registered trials. In making decisions about discontinuing these drugs, clinicians should exercise caution, considering the evidence from existing trials along with other factors important to patients and their carers.
Collapse
Affiliation(s)
- Carole Parsons
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Wei Yin Lim
- Centre for Clinical Epidemiology, Institute for Clinical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Malaysia
| | - Clement Loy
- Brain and Mind Centre and Sydney School of Public Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | | | - Peter Passmore
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Stephanie A Ward
- Monash Aging Research Center, The Kingston Centre, Cheltenham, Australia
| | - Carmel Hughes
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|