1
|
Zhai Z, Kong F, Zhu Z, Dai J, Cai J, Xie D, Shen Y, Xu Y, Sun T. Effect and Potential Mechanism of Immunotherapy on Cognitive Deficits in Animal Models of Alzheimer's Disease: A Systematic Review and Meta-Analysis. Am J Geriatr Psychiatry 2024; 32:555-583. [PMID: 38158285 DOI: 10.1016/j.jagp.2023.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE Immunotherapy has been reported to ameliorate Alzheimer's disease (AD) in the animal model; however, the immunologic approaches and mechanisms have not been specifically described. Thus, the systematic review and meta-analysis were conducted to explore the effect and potential mechanism of immunotherapy on AD animal experiments based on behavioral indicators. METHODS According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and the Cochrane Collaboration guidelines and the inclusion/exclusion criteria of immunotherapy in animal studies, 15 studies were systematically reviewed after extraction from a collected database of 3,742 publications. Finally, the effect and mechanism of immunotherapy on AD models were described by performing multiple subgroup analyses. RESULTS After immunotherapy, the escape latency was reduced by 18.15 seconds and the number of crossings over the platform location was increased by 1.60 times in the Morris Water Maze. Furthermore, compared to the control group, active and passive immunization could markedly ameliorate learning and memory impairment in 3 × Tg AD animal models, and active immunization could ameliorate the learning and memory ability of the APPswe/PS1ΔE9 AD animal model. Meanwhile, it could be speculated that cognitive dysfunction was improved by immunotherapy, perhaps mainly via reducing Aβ40, Aβ42, and Tau levels, as well as increasing IL-4 levels. CONCLUSION Immunotherapy significantly ameliorated the cognitive dysfunction of AD animal models by assessing behavioral indicators.
Collapse
Affiliation(s)
- Zhenwei Zhai
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Fanjing Kong
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Zhishan Zhu
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jingyi Dai
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jie Cai
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy (DX, YS, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuzhao Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy (DX, YS, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province (YX), Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Tao Sun
- School of Intelligent Medicine (ZZ, FK, ZZ, JD, JC, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy (DX, YS, TS), Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
2
|
Loeffler DA. Antibody-Mediated Clearance of Brain Amyloid-β: Mechanisms of Action, Effects of Natural and Monoclonal Anti-Aβ Antibodies, and Downstream Effects. J Alzheimers Dis Rep 2023; 7:873-899. [PMID: 37662616 PMCID: PMC10473157 DOI: 10.3233/adr-230025] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Immunotherapeutic efforts to slow the clinical progression of Alzheimer's disease (AD) by lowering brain amyloid-β (Aβ) have included Aβ vaccination, intravenous immunoglobulin (IVIG) products, and anti-Aβ monoclonal antibodies. Neither Aβ vaccination nor IVIG slowed disease progression. Despite conflicting phase III results, the monoclonal antibody Aducanumab received Food and Drug Administration (FDA) approval for treatment of AD in June 2021. The only treatments unequivocally demonstrated to slow AD progression to date are the monoclonal antibodies Lecanemab and Donanemab. Lecanemab received FDA approval in January 2023 based on phase II results showing lowering of PET-detectable Aβ; phase III results released at that time indicated slowing of disease progression. Topline results released in May 2023 for Donanemab's phase III trial revealed that primary and secondary end points had been met. Antibody binding to Aβ facilitates its clearance from the brain via multiple mechanisms including promoting its microglial phagocytosis, activating complement, dissolving fibrillar Aβ, and binding of antibody-Aβ complexes to blood-brain barrier receptors. Antibody binding to Aβ in peripheral blood may also promote cerebral efflux of Aβ by a peripheral sink mechanism. According to the amyloid hypothesis, for Aβ targeting to slow AD progression, it must decrease downstream neuropathological processes including tau aggregation and phosphorylation and (possibly) inflammation and oxidative stress. This review discusses antibody-mediated mechanisms of Aβ clearance, findings in AD trials involving Aβ vaccination, IVIG, and anti-Aβ monoclonal antibodies, downstream effects reported in those trials, and approaches which might improve the Aβ-clearing ability of monoclonal antibodies.
Collapse
Affiliation(s)
- David A. Loeffler
- Beaumont Research Institute, Department of Neurology, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
3
|
An Aβ3-10-KLH vaccine reduced Alzheimer’s disease-like pathology and had a sustained effect in Tg-APPswe/PSEN1dE9 mice. Brain Res 2017; 1673:72-77. [DOI: 10.1016/j.brainres.2017.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 11/24/2022]
|
4
|
Meng Y, Ding L, Zhang H, Yin W, Yan Y, Cao Y. Immunization of Tg-APPswe/PSEN1dE9 mice with Aβ3-10-KLH vaccine prevents synaptic deficits of Alzheimer’s disease. Behav Brain Res 2017; 332:64-70. [DOI: 10.1016/j.bbr.2017.05.056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/04/2017] [Indexed: 01/12/2023]
|
5
|
Cheng L, Zhang J, Li XY, Yuan L, Pan YF, Chen XR, Gao TM, Qiao JT, Qi JS. A novel antibody targeting sequence 31-35 in amyloid β protein attenuates Alzheimer's disease-related neuronal damage. Hippocampus 2016; 27:122-133. [PMID: 27784133 DOI: 10.1002/hipo.22676] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 11/10/2022]
Abstract
Amyloid β protein (Aβ) plays a critical role in pathogenesis of Alzheimer's disease (AD). Our previous studies indicated that the sequence 31-35 in Aβ molecule is an effective active center responsible for Aβ neurotoxicity in vivo and in vitro. In the present study, we prepared a novel antibody specifically targeting the sequence 31-35 of amyloid β protein, and investigated the neuroprotection of the anti-Aβ31-35 antibody against Aβ1-42 -induced impairments in neuronal viability, spatial memory, and hippocampal synaptic plasticity in rats. The results showed that the anti-Aβ31-35 antibody almost equally bound to both Aβ31-35 and Aβ1-42 , and pretreatment with the antibody dose-dependently prevented Aβ1-42 -induced cytotoxicity on cultured primary cortical neurons. In behavioral study, intracerebroventricular (i.c.v.) injection of anti-Aβ31-35 antibody efficiently attenuated Aβ1-42 -induced impairments in spatial learning and memory of rats. In vivo electrophysiological experiments further indicated that Aβ1-42 -induced suppression of hippocampal synaptic plasticity was effectively reversed by the antibody. These results demonstrated that the sequence 31-35 of Aβ may be a new therapeutic target, and the anti-Aβ31-35 antibody could be a novel immunotheraputic approach for the treatment of AD. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Li Cheng
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China.,The General Hospital of TISCO Affiliated to Shanxi Medical University, Taiyuan, 030003, China
| | - Jun Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xin-Yi Li
- Department of Neurology, Shanxi Dayi Hospital, Taiyuan, 030032, China
| | - Li Yuan
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yan-Fang Pan
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao-Rong Chen
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Tian-Ming Gao
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China.,Department of Neurobiology, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Tian Qiao
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin-Shun Qi
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
6
|
Li N, Liu Y, Li W, Zhou L, Li Q, Wang X, He P. A UPLC/MS-based metabolomics investigation of the protective effect of ginsenosides Rg1 and Rg2 in mice with Alzheimer's disease. J Ginseng Res 2015; 40:9-17. [PMID: 26843817 PMCID: PMC4703800 DOI: 10.1016/j.jgr.2015.04.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/20/2015] [Accepted: 04/20/2015] [Indexed: 01/10/2023] Open
Abstract
Background Alzheimer's disease (AD) is a progressive brain disease, for which there is no effective drug therapy at present. Ginsenoside Rg1 (G-Rg1) and G-Rg2 have been reported to alleviate memory deterioration. However, the mechanism of their anti-AD effect has not yet been clearly elucidated. Methods Ultra performance liquid chromatography tandem MS (UPLC/MS)-based metabolomics was used to identify metabolites that are differentially expressed in the brains of AD mice with or without ginsenoside treatment. The cognitive function of mice and pathological changes in the brain were also assessed using the Morris water maze (MWM) and immunohistochemistry, respectively. Results The impaired cognitive function and increased hippocampal Aβ deposition in AD mice were ameliorated by G-Rg1 and G-Rg2. In addition, a total of 11 potential biomarkers that are associated with the metabolism of lysophosphatidylcholines (LPCs), hypoxanthine, and sphingolipids were identified in the brains of AD mice and their levels were partly restored after treatment with G-Rg1 and G-Rg2. G-Rg1 and G-Rg2 treatment influenced the levels of hypoxanthine, dihydrosphingosine, hexadecasphinganine, LPC C 16:0, and LPC C 18:0 in AD mice. Additionally, G-Rg1 treatment also influenced the levels of phytosphingosine, LPC C 13:0, LPC C 15:0, LPC C 18:1, and LPC C 18:3 in AD mice. Conclusion These results indicate that the improvements in cognitive function and morphological changes produced by G-Rg1 and G-Rg2 treatment are caused by regulation of related brain metabolic pathways. This will extend our understanding of the mechanisms involved in the effects of G-Rg1 and G-Rg2 on AD.
Collapse
Affiliation(s)
- Naijing Li
- Department of Gerontology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, China
| | - Ying Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Wei Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Ling Zhou
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qing Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueqing Wang
- Department of Gastroenterology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, China
| | - Ping He
- Department of Gerontology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
7
|
|
8
|
Alves R, Yang M, Batista M, Ferreira L. Alzheimer's disease: is a vaccine possible? Braz J Med Biol Res 2014; 47:438-44. [PMID: 24878604 PMCID: PMC4086169 DOI: 10.1590/1414-431x20143434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 03/31/2014] [Indexed: 12/21/2022] Open
Abstract
The cause of Alzheimer's disease is still unknown, but the disease is distinctively characterized by the accumulation of β-amyloid plaques and neurofibrillary tangles in the brain. These features have become the primary focus of much of the research looking for new treatments for the disease, including immunotherapy and vaccines targeting β-amyloid in the brain. Adverse effects observed in a clinical trial based on the β-amyloid protein were attributed to the presence of the target antigen and emphasized the relevance of finding safer antigen candidates for active immunization. For this kind of approach, different vaccine formulations using DNA, peptide, and heterologous prime-boost immunization regimens have been proposed. Promising results are expected from different vaccine candidates encompassing B-cell epitopes of the β-amyloid protein. In addition, recent results indicate that targeting another protein involved in the etiology of the disease has opened new perspectives for the effective prevention of the illness. Collectively, the evidence indicates that the idea of finding an effective vaccine for the control of Alzheimer's disease, although not without challenges, is a possibility.
Collapse
Affiliation(s)
- R.P.S. Alves
- Universidade de São Paulo, Instituto de Ciências Biomédicas II,
Departamento de Microbiologia, Laboratório de Desenvolvimento de Vacinas, São Paulo,
SP, Brasil, Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia,
Instituto de Ciências Biomédicas II, Universidade de São Paulo, São Paulo, SP,
Brasil
| | - M.J. Yang
- Instituto Butantan, Laboratório de Genética, São Paulo, SP, Brasil,
Laboratório de Genética, Instituto Butantan, São Paulo, SP, Brasil
| | - M.T. Batista
- Universidade de São Paulo, Instituto de Ciências Biomédicas II,
Departamento de Microbiologia, Laboratório de Desenvolvimento de Vacinas, São Paulo,
SP, Brasil, Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia,
Instituto de Ciências Biomédicas II, Universidade de São Paulo, São Paulo, SP,
Brasil
| | - L.C.S. Ferreira
- Universidade de São Paulo, Instituto de Ciências Biomédicas II,
Departamento de Microbiologia, Laboratório de Desenvolvimento de Vacinas, São Paulo,
SP, Brasil, Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia,
Instituto de Ciências Biomédicas II, Universidade de São Paulo, São Paulo, SP,
Brasil
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW We reviewed clinical trials on active and passive anti-β-amyloid (Aβ) immunotherapy for the treatment of Alzheimer's disease with a particular focus on monoclonal antibodies against Aβ. RECENT FINDINGS Studies on anti-Alzheimer's disease immunotherapy published in the period from January 2012 to October 2013 were reviewed. SUMMARY Both active and passive anti-Aβ immunotherapies were shown to clear brain Aβ deposits. However, an active anti-Aβ vaccine (AN1792) has been discontinued because it caused meningoencephalitis in 6% of Alzheimer's disease patients treated. Among passive immunotherapeutics, two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients with bapineuzumab, a humanized monoclonal antibody directed at the N-terminal sequence of Aβ, were disappointing. Another antibody, solanezumab, directed at the mid-region of Aβ, failed in two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients. A third Phase III study with solanezumab is ongoing in mildly affected Alzheimer's disease patients based on encouraging results in this subgroup of patients. Second-generation active Aβ vaccines (ACC-001, CAD106, and Affitope AD02) and new passive anti-Aβ immunotherapies (gantenerumab and crenezumab) are being tested in prodromal Alzheimer's disease patients, in presymptomatic individuals with Alzheimer's disease-related mutations, or in asymptomatic individuals at risk of developing Alzheimer's disease to definitely test the Aβ cascade hypothesis of Alzheimer's disease.
Collapse
|
10
|
Panza F, Solfrizzi V, Imbimbo BP, Tortelli R, Santamato A, Logroscino G. Amyloid-based immunotherapy for Alzheimer's disease in the time of prevention trials: the way forward. Expert Rev Clin Immunol 2014; 10:405-19. [DOI: 10.1586/1744666x.2014.883921] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
11
|
Guo W, Sha S, Xing X, Jiang T, Cao Y. Reduction of cerebral Aβ burden and improvement in cognitive function in Tg-APPswe/PSEN1dE9 mice following vaccination with a multivalent Aβ3-10 DNA vaccine. Neurosci Lett 2013; 549:109-15. [PMID: 23800542 DOI: 10.1016/j.neulet.2013.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/28/2013] [Accepted: 06/13/2013] [Indexed: 12/22/2022]
Abstract
To develop a safe and efficient Aβ vaccine for Alzheimer's disease, we constructed a plasmid DNA vaccine encoding ten repeats of Aβ3-10 and three copies of C3d-p28 as a molecular adjuvant and administered it intramuscularly in 12-month-old female Tg-APPswe/PSEN1dE9 mice. Therapeutic immunization with p(Aβ3-10)10-C3d-p28.3 stimulated a Th2 immune response that elicited therapeutic levels of anti-Aβ antibodies and improved cognitive function. In addition, the vaccine reduced the cerebral Aβ burden and astrocytosis without increasing the incidence of microhemorrhage. Our results indicate that the p(Aβ3-10)10-C3d-p28.3 vaccine is a promising immunotherapeutic option for Aβ vaccination in Alzheimer's disease.
Collapse
Affiliation(s)
- Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, Liao Ning Province, China
| | | | | | | | | |
Collapse
|