1
|
Serag I, Abouzid M, Moawad MHED, Jaradat JH, Hendawy M, Hendi NI, Alkhawaldeh IM, Abdullah JA, Elsakka MM, Muneer MA, Elnagar MA, Fakher MA, Elkenani AJ, Abbas A. Vaccines for Alzheimer's disease: a brief scoping review. Neurol Sci 2025:10.1007/s10072-025-08073-2. [PMID: 40111670 DOI: 10.1007/s10072-025-08073-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the most common cause of dementia among older adults. Existing treatments-such as cholinesterase inhibitors, N-methyl-D-aspartate receptor antagonists, and monoclonal antibodies targeting amyloid beta-can improve functional and neuropsychiatric outcomes but fail to prevent disease onset, halt progression, or adequately reduce amyloid-beta burden. Consequently, research efforts have shifted to primary prevention through immunization, although the efficacy of these strategies remains uncertain. This review explores the efficacy, safety, and adverse events of current immunotherapies for AD and discusses future research and clinical implications. METHODS A scoping review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Scoping Reviews (PRISMA-SR) checklist. A systematic search was carried out using PubMed, Scopus, and Web of Science. RESULTS A total of 145 studies were included. Preclinical research often employed transgenic mouse models to investigate AD pathology and vaccine benefits, while Phase I and II clinical trials centered on safety and preliminary efficacy in humans. Most studies were conducted in the USA, China, and Japan, highlighting these countries' strong clinical trial infrastructure. Vaccination frequently reduced amyloid-beta or tau pathology in preclinical settings, although cognitive outcomes were inconsistent. Clinical trials primarily focused on safety and immune response, with newer vaccines such as ABvac40 demonstrating encouraging results and minimal adverse events. CONCLUSION Although AD vaccines show promise in preclinical settings, longer and more comprehensive clinical trials are necessary to determine their long-term efficacy and safety. Standardized protocols and efforts to reduce regional disparities in research would facilitate better comparability and generalizability of findings, thereby guiding the future development of effective immunotherapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - 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 Hossam El Din Moawad
- Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Mohamed Hendawy
- Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | | | | | | | | | | | | | - Aya J Elkenani
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
2
|
Jin Y, Du Q, Song M, Kang R, Zhou J, Zhang H, Ding Y. Amyloid-β-targeting immunotherapies for Alzheimer's disease. J Control Release 2024; 375:346-365. [PMID: 39271059 DOI: 10.1016/j.jconrel.2024.09.012] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Recent advances in clinical passive immunotherapy have provided compelling evidence that eliminating amyloid-β (Aβ) slows cognitive decline in Alzheimer's disease (AD). However, the modest benefits and side effects observed in clinical trials indicate that current immunotherapy therapy is not a panacea, highlighting the need for a deeper understanding of AD mechanisms and the significance of early intervention through optimized immunotherapy or immunoprevention. This review focuses on the centrality of Aβ pathology in AD and summarizes recent clinical progress in passive and active immunotherapies targeting Aβ, discussing their lessons and failures to inform future anti-Aβ biotherapeutics design. Various delivery strategies to optimize Aβ-targeting immunotherapies are outlined, highlighting their benefits and drawbacks in overcoming challenges such as poor stability and limited tissue accessibility of anti-Aβ biotherapeutics. Additionally, the perspectives and challenges of immunotherapy and immunoprevention targeting Aβ are concluded in the end, aiming to guide the development of next-generation anti-Aβ immunotherapeutic agents towards improved efficacy and safety.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qiaofei Du
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Mingjie Song
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixin Kang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jianping Zhou
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yang Ding
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
3
|
Lambracht-Washington D, Fu M, Wight-Carter M, Riegel M, Hynan LS, Rosenberg RN. DNA Aβ42 immunization via needle-less Jet injection in mice and rabbits as potential immunotherapy for Alzheimer's disease. J Neurol Sci 2023; 446:120564. [PMID: 36731358 DOI: 10.1016/j.jns.2023.120564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia found in the elderly and disease progression is associated with accumulation of Amyloid beta 1-42 (Aβ42) in brain. An immune-mediated approach as a preventive intervention to reduce amyloid plaques without causing brain inflammation is highly desirable for future clinical use. Genetic immunization, in which the immunizing agent is DNA encoding Aβ42, has great potential because the immune response to DNA delivered into the skin is generally non-inflammatory, and thus differs quantitatively and qualitatively from immune responses elicited by peptides, which are inflammatory with production of IFNγ and IL-17 cytokines by activated T cells. DNA immunization has historically been proven difficult to apply to larger mammals. A potential barrier to use DNA immunization in large mammals is the method for delivery of the DNA antigen. We tested jet injection in mice and rabbits and found good antibody production and safe immune responses (no inflammatory cytokines). We found significant reduction of amyloid plaques and Aβ peptides in brains of the DNA Aβ42 immunized 3xTg-AD mouse model. This study was designed to optimize DNA delivery for possible testing of the DNA Aβ42 vaccine for AD prevention in a clinical trial.
Collapse
Affiliation(s)
| | - Min Fu
- Department of Neurology, UT Southwestern Medical Center Dallas, TX, USA.
| | - Mary Wight-Carter
- Animal Resource Center, UT Southwestern Medical Center Dallas, TX, USA.
| | - Matthew Riegel
- Animal Resource Center, UT Southwestern Medical Center Dallas, TX, USA; University of Kansas, Lawrence, KS, USA.
| | - Linda S Hynan
- Departments of Population and Data Sciences (Biostatistics) & Psychiatry, UT Southwestern Medical Center Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology, UT Southwestern Medical Center Dallas, TX, USA.
| |
Collapse
|
4
|
Arora S, Sharma D, Layek B, Singh J. A Review of Brain-Targeted Nonviral Gene-Based Therapies for the Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:4237-4255. [PMID: 34705472 DOI: 10.1021/acs.molpharmaceut.1c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases of the central nervous system (CNS) are difficult to treat owing to the complexity of the brain and the presence of a natural blood-brain-barrier (BBB). Alzheimer's disease (AD) is one of the major progressive and currently incurable neurodegenerative disorders of the CNS, which accounts for 60-80% of cases of dementia. The pathophysiology of AD involves the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Additionally, synaptic loss and imbalance of neuronal signaling molecules are characterized as important markers of AD. Existing treatments of AD help in the management of its symptoms and aim toward the maintenance of cognitive functions, behavior, and attenuation of gradual memory loss. Over the past decade, nonviral gene therapy has attracted increasing interest due to its various advantages over its viral counterparts. Moreover, advancements in nonviral gene technology have led to their increasing contributions in clinical trials. However, brain-targeted nonviral gene delivery vectors come across various extracellular and intracellular barriers, limiting their ability to transfer the therapeutic gene into the target cells. Chief barriers to nonviral gene therapy have been discussed briefly in this review. We have also highlighted the rapid advancement of several nonviral gene therapies for AD, which are broadly categorized into physical and chemical methods. These methods aim to modulate Aβ, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), apolipoprotein E, or neurotrophic factors' expression in the CNS. Overall, this review discusses challenges and recent advancements of nonviral gene therapy for AD.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
5
|
Low-Dose Phosphodiesterase III Inhibitor Reduces the Vascular Amyloid Burden in Amyloid-β Protein Precursor Transgenic Mice. Int J Mol Sci 2020; 21:ijms21072295. [PMID: 32225099 PMCID: PMC7177305 DOI: 10.3390/ijms21072295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 01/13/2023] Open
Abstract
A previous study reported that relatively high-dose cilostazol (0.3%) promoted the drainage of cerebrovascular amyloid-β (Aβ) protein in Aβ Precursor Protein (APP) transgenic mice overexpressing vasculotropic Aβ. We investigated whether lower-dose cilostazol can decrease micro-hemorrhages and Aβ deposition in the brain using APP transgenic mice. At baseline, 14-month-old female Tg2576 mice were randomly assigned to a control group (vehicle), aspirin group (0.01% aspirin), or cilostazol group (0.01% cilostazol). The severity of cerebral micro-hemorrhages (i.e., number), area of senile plaque, and severity of vascular amyloid burden (quantified with cerebral amyloid angiopathy (CAA) score (=number of Aβ-positive vessels × severity of amyloid burden of Aβ-positive vessels) were evaluated in the brain of mice aged 15 and 21–23 months. At 15 months, no differences were shown in each pathological change among the three groups. At 21–23 months, there were no differences in the severity of cerebral micro-hemorrhages or area of senile plaque among the three groups. However, the CAA score was significantly lower in the cilostazol compared to the control group (p = 0.046, Mann–Whitney U test), although no difference was seen between the control and aspirin group. Our study showed that lower-dose cilostazol could reduce the vascular amyloid burden without increasing cerebral micro-hemorrhages in APP transgenic mice.
Collapse
|
6
|
Abstract
This paper considers ethical issues related to early diagnosis and all forms of prevention of Alzheimer disease and related conditions. It offers a critical view of the current state of scientific, clinical, and social responses to the growing number of older people with cognitive challenges, and suggests how priorities going forward should be different from those receiving most attention today. We begin with a review of global policy efforts, consider the fundamental goals of prevention, examine issues surrounding early diagnosis, explore more deeply values associated with efforts to prevent age associated cognitive decline, and conclude by considering often unexplored ethical issues that contextualize the field and should influence our approaches to the future.
Collapse
Affiliation(s)
- Peter J Whitehouse
- Case Western Reserve University, Cleveland, Ohio, USA; Baycrest Health Center, Cleveland, Ohio, USA; Institute of Life Course and Aging, University of Toronto; Intergenerational Schools International, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Rosenberg RN, Fu M, Lambracht-Washington D. Intradermal active full-length DNA Aβ42 immunization via electroporation leads to high anti-Aβ antibody levels in wild-type mice. J Neuroimmunol 2018; 322:15-25. [PMID: 29958693 PMCID: PMC6192700 DOI: 10.1016/j.jneuroim.2018.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 11/24/2022]
Abstract
Aβ immunotherapies with anti-Aβ antibody responses have high potential as possible prevention treatment for Alzheimer's disease. We have previously shown that active DNA Aβ1-42 immunization via gene gun delivery led to a non-inflammatory immune response resulting in decreased Aβ levels in brains of an immunized AD mouse model. To make DNA vaccination more applicable for clinical use, we used here intradermal electroporation. With fine tuning of the electropulse parameters, high antibody levels and low levels of inflammatory cytokines in the cellular immunoassays were observed. Full-length DNA Aβ1-42 immunization delivered via electroporation has potential to be used in the clinical setting.
Collapse
Affiliation(s)
- Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA
| | - Min Fu
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA
| | - Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA.
| |
Collapse
|
8
|
Martins YA, Tsuchida CJ, Antoniassi P, Demarchi IG. Efficacy and Safety of the Immunization with DNA for Alzheimer's Disease in Animal Models: A Systematic Review from Literature. J Alzheimers Dis Rep 2017; 1:195-217. [PMID: 30480238 PMCID: PMC6159633 DOI: 10.3233/adr-170025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that does not have a proven cure; however, one of the most promising strategies for its treatment has been DNA vaccines. OBJECTIVE The present review is aimed to report the new developments of the efficacy and safety of DNA vaccines for AD in animal models. METHOD The method PRISMA was used for this review. The article search was made in the electronic databases PubMed, LILACS, and Scopus using the descriptors ''Alzheimer disease" and ''Vaccine, DNA". Articles published between January 2001 and September 2017 in English, Portuguese, and Spanish were included. RESULTS Upon the consensus, the researchers identified 28 original articles. The studies showed satisfying results as for the decrease of amyloid plaques in mouse, rabbits, and monkeys brains using mostly the DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11, mainly with a gene gun. In addition to a reduction in tau by the first DNA vaccine (AV-1980D) targeting this protein. The use of adjuvants and boosters also had positive results as they increased the destruction of the amyloid plaques and induced an anti-inflammatory response profile without side effects. CONCLUSION The results of DNA vaccines targeting the amyloid-β and the tau protein with or without adjuvants and boosters were promising in reducing amyloid plaques and tau protein without side effects in animals. Although there are many vaccines being tested in animals, few reach clinical trials. Thus, as a future perspective, we suggest that clinical studies should be conducted with vaccines that have been promising in animal models (e.g., DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11).
Collapse
|
9
|
Abstract
IMPORTANCE To provide a comprehensive review of knowledge of the genomics of Alzheimer disease (AD) and DNA amyloid β 42 (Aβ42) vaccination as a potential therapy. OBSERVATIONS Genotype-phenotype correlations of AD are presented to provide a comprehensive appreciation of the spectrum of disease causation. Alzheimer disease is caused in part by the overproduction and lack of clearance of Aβ protein. Oligomer Aβ, the most toxic species of Aβ, causes direct injury to neurons, accompanied by enhanced neuroinflammation, astrocytosis and gliosis, and eventually neuronal loss. The strongest genetic evidence supporting this hypothesis derives from mutations in the amyloid precursor protein (APP) gene. A detrimental APP mutation at the β-secretase cleavage site linked to early-onset AD found in a Swedish pedigree enhances Aβ production, in contrast to a beneficial mutation 2 residues away in APP that reduces Aβ production and protects against the onset of sporadic AD. A number of common variants associated with late-onset AD have been identified including apolipoprotein E, BIN1, ABC7, PICALM, MS4A4E/MS4A6A, CD2Ap, CD33, EPHA1, CLU, CR1, and SORL1. One or 2 copies of the apolipoprotein E ε4 allele are a major risk factor for late-onset AD. With DNA Aβ42 vaccination, a Th2-type noninflammatory immune response was achieved with a downregulation of Aβ42-specific effector (Th1, Th17, and Th2) cell responses at later immunization times. DNA Aβ42 vaccination upregulated T regulator cells (CD4+, CD25+, and FoxP3+) and its cytokine interleukin 10, resulting in downregulation of T effectors. CONCLUSIONS AND RELEVANCE Mutations in APP and PS-1 and PS-2 genes that are associated with early-onset, autosomal, dominantly inherited AD, in addition to the at-risk gene polymorphisms responsible for late-onset AD, all indicate a direct and early role of Aβ in the pathogenesis of AD. A translational result of genomic research has been Aβ-reducing therapies including DNA Aβ42 vaccination as a promising approach to delay or prevent this disease.
Collapse
Affiliation(s)
- Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas2Editor, JAMA Neurology
| | | | - Gang Yu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas
| | - Weiming Xia
- Geriatric Research, Education and Clinical Center, Bedford Veterans Hospital, Bedford, Massachusetts5Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
10
|
Lambracht-Washington D, Fu M, Frost P, Rosenberg RN. Evaluation of a DNA Aβ42 vaccine in adult rhesus monkeys (Macaca mulatta): antibody kinetics and immune profile after intradermal immunization with full-length DNA Aβ42 trimer. ALZHEIMERS RESEARCH & THERAPY 2017; 9:30. [PMID: 28441965 PMCID: PMC5405538 DOI: 10.1186/s13195-017-0257-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/24/2017] [Indexed: 01/18/2023]
Abstract
Background Aggregated amyloid-β peptide 1–42 (Aβ42), derived from the cellular amyloid precursor protein, is one of the pathological hallmarks of Alzheimer’s disease (AD). Although active immunization against Aβ42 peptide was successful in AD mouse models and led to removal of plaques and improved memory, a similar clinical trial in humans (Aβ42 peptide immunization with QS-21 adjuvant) was stopped in phase II, when 6% of the treated patients developed encephalitis. Currently ongoing passive immunizations with the injection of preformed monoclonal antibodies against different epitopes within the Aβ1–42 peptide, which do not lead to activation of the immune system, have shown some effects in slowing AD pathology. Active DNA Aβ42 immunizations administered with the gene gun into the skin are noninflammatory because they activate a different T-cell population (Th2) with different cytokine responses eliciting a different humoral immune response. We present our findings in rhesus macaques that underwent the DNA Aβ42 immunization via gene gun delivery into the skin. Methods Six rhesus monkeys received two different doses of a DNA Aβ42 trimer vaccine. The humoral immune response was analyzed from blood throughout the study, and cellular immune responses were determined in peripheral blood mononuclear cells (PBMCs) after three and six immunizations. Results DNA Aβ42 trimer immunization led to high titer antibody responses in the nonhuman primate (NHP) model. Antibodies generated in the rhesus monkeys following DNA Aβ42 immunization detected amyloid plaques consisting of human Aβ42 peptide in the brain of the triple-transgenic AD mouse model. T-cell responses showed no interferon (IFN)-γ- and interleukin (IL)-17-producing cells from PBMCs in Enzyme-Linked ImmunoSpot assays after three immunization time points. At six immunization time points, IFN-γ- and IL-17-producing cells were found in immunized animals as well as in control animals and were thus considered nonspecific and not due to the immunization regimen. IFN-γ and IL-17 secretion in response to Aβ42 peptide restimulation became undetectable after a 3-month rest period. Conclusions Intradermal DNA Aβ42 immunization delivered with the gene gun produces a high antibody response in NHPs and is highly likely to be effective and safe in a clinical AD prevention trial in patients.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8813, USA.
| | - Min Fu
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8813, USA
| | - Pat Frost
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8813, USA.,Alzheimer's Disease Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
11
|
Management of Alzheimer’s disease—An insight of the enzymatic and other novel potential targets. Int J Biol Macromol 2017; 97:700-709. [DOI: 10.1016/j.ijbiomac.2017.01.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/15/2017] [Accepted: 01/16/2017] [Indexed: 12/25/2022]
|
12
|
Lambracht-Washington D, Fu M, Wight-Carter M, Riegel M, Rosenberg RN. Evaluation of a DNA Aβ42 Vaccine in Aged NZW Rabbits: Antibody Kinetics and Immune Profile after Intradermal Immunization with Full-Length DNA Aβ42 Trimer. J Alzheimers Dis 2017; 57:97-112. [PMID: 28222511 PMCID: PMC5345648 DOI: 10.3233/jad-160947] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2016] [Indexed: 01/03/2023]
Abstract
A pathological hallmark of Alzheimer's disease (AD) are amyloid plaques in the brain consisting of aggregated amyloid-β 42 peptide (Aβ42) derived from cellular amyloid-β protein precursor (AβPP). Based on successful experiments in mouse AD models, active immunization with Aβ42 peptide and passive immunizations with anti-Aβ42 antibodies were started in clinical trials. Active Aβ42 peptide immunization in humans had led to an inflammatory autoimmune response, and the trial was stopped. Passive immunizations had shown some effects in slowing AD pathology. Active DNA Aβ42 immunizations administered with the gene gun into the skin elicits a different immune response and is non-inflammatory. While in rodents, good responses had been found for this type of immunization, positive results in larger mammals are missing. We present here results from sixteen New Zealand White Rabbits, which underwent intradermal DNA Aβ42 immunization via gene gun. The humoral immune response was analyzed from blood throughout the study, and cellular immune responses were determined from spleens at the end of the study. A good anti-Aβ antibody response was found in the rabbit model. The T cell response after re-stimulation in cell culture showed no IFNγ producing cells when ELISPOT assays were analyzed from PBMC, but low numbers of IFNγ and IL-17 producing cells were found in ELISPOTS from spleens (both 5 immunizations). Brains from immunized rabbits showed no signs of encephalitis. Based on these results, DNA Aβ42 immunization is highly likely to be safe and effective to test in a possible clinical AD prevention trial in patients.
Collapse
MESH Headings
- Aging/immunology
- Aging/pathology
- Alzheimer Disease/immunology
- Amyloid beta-Peptides/immunology
- Animals
- Autoantibodies/immunology
- B-Lymphocytes/immunology
- Biolistics
- Brain/immunology
- Brain/pathology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Epitopes, B-Lymphocyte/immunology
- Female
- Humans
- Injections, Intradermal
- Male
- Mice, Transgenic
- Peptide Fragments/immunology
- Plaque, Amyloid/metabolism
- Plaque, Amyloid/pathology
- Plaque, Amyloid/prevention & control
- Rabbits
- T-Lymphocytes/immunology
- Vaccination
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- alpha-Synuclein/metabolism
Collapse
Affiliation(s)
| | - Min Fu
- Department of Neurology and Neurotherapeutics, UTSouthwestern Medical Center Dallas, Dallas, TX, USA
| | | | | | - Roger N. Rosenberg
- Department of Neurology and Neurotherapeutics, UTSouthwestern Medical Center Dallas, Dallas, TX, USA
- Alzheimer’s Disease Center, Dallas, TX, USA
| |
Collapse
|
13
|
Moosavi B, Mousavi B, Macreadie IG. Yeast Model of Amyloid-β and Tau Aggregation in Alzheimer's Disease. J Alzheimers Dis 2016; 47:9-16. [PMID: 26402750 DOI: 10.3233/jad-150173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The amyloid-β peptide (Aβ) and the phosphorylated protein tau have been widely implicated in Alzheimer's disease and are the focus of most research. Both agents have been extensively studied in mammalian cell culture and in animal studies, but new research is focusing on yeast models. Yeast are eukaryotes, just like us, and are amenable to effects and expression of Aβ and tau and appear able to 'report' with considerable relevance on the effects of these biomolecules. The use of yeast enables powerful new approaches to understanding how to overcome the effects of Aβ and tau, and such advances could lead to new therapies to prevent the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Behrooz Moosavi
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, P.R. China
| | - Bibimaryam Mousavi
- Laboratory of Organometallics, Catalysis and Ordered Materials, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, P.R. China
| | - Ian G Macreadie
- School of Applied Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
14
|
Azizi G, Navabi SS, Al-Shukaili A, Seyedzadeh MH, Yazdani R, Mirshafiey A. The Role of Inflammatory Mediators in the Pathogenesis of Alzheimer's Disease. Sultan Qaboos Univ Med J 2015; 15:e305-16. [PMID: 26357550 DOI: 10.18295/squmj.2015.15.03.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/17/2014] [Accepted: 03/19/2015] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder associated with advanced age, is the most common cause of dementia globally. AD is characterised by cognitive dysfunction, deposition of amyloid plaques, neurofibrillary tangles and neuro-inflammation. Inflammation of the brain is a key pathological hallmark of AD. Thus, clinical and immunopathological evidence of AD could be potentially supported by inflammatory mediators, including cytokines, chemokines, the complement system, acute phase proteins and oxidative mediators. In particular, oxidative mediators may actively contribute to the progression of AD and on-going inflammation in the brain. This review provides an overview of the functions and activities of inflammatory mediators in AD. An improved understanding of inflammatory processes and their role in AD is needed to improve therapeutic research aims in the field of AD and similar diseases.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran; ; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadi S Navabi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmed Al-Shukaili
- Health & Social Services Sector, The Research Council Oman, Muscat, Oman
| | - Mir H Seyedzadeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran ; Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Lambracht-Washington D, Rosenberg RN. A noninflammatory immune response in aged DNA Aβ42-immunized mice supports its safety for possible use as immunotherapy in AD patients. Neurobiol Aging 2014; 36:1274-81. [PMID: 25725942 DOI: 10.1016/j.neurobiolaging.2014.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/02/2014] [Accepted: 12/06/2014] [Indexed: 12/17/2022]
Abstract
Aging in the immune system results in tendency to proinflammatory responses. Intradermal DNA immunization showed Th2 polarized noninflammatory immune responses. We tested here 18-month-old mice which were immunized with Aβ42 peptide, DNA Aβ42 trimer, or 2 different prime boost protocols identical to previous experiments. High Aβ42 antibody levels were found in aged mice which had received peptide immunizations (900 μg/mL plasma), and in mice which had received peptide prime and DNA boost immunizations (500 μg/mL), compared with antibodies in DNA Aβ42 immunized mice with 50 μg/mL. Although we found T-cell proliferation and inflammatory cytokines in mice which had received peptide or prime boost immunization, these were not found in DNA-immunized mice. The results are concordant with proinflammatory responses because of immunosenescence and contraindicate the use of Aβ42 peptide immunizations or prime boost immunization protocols for the use in elderly Alzheimer's disease patients. DNA Aβ42 immunization only on the other hand does lead to effective levels of antibodies without inflammatory cytokine or T-cell responses in the aged animal model tested.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Fujisaki K, Tsuruya K, Yamato M, Toyonaga J, Noguchi H, Nakano T, Taniguchi M, Tokumoto M, Hirakata H, Kitazono T. Cerebral oxidative stress induces spatial working memory dysfunction in uremic mice: neuroprotective effect of tempol. Nephrol Dial Transplant 2014; 29:529-538. [DOI: 10.1093/ndt/gft327] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
17
|
Rosenberg RN, Lambracht-Washington D. DNA Aβ42 vaccination as possible alternative immunotherapy for Alzheimer disease. JAMA Neurol 2013; 70:772-3. [PMID: 23700123 DOI: 10.1001/jamaneurol.2013.1502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9036, USA.
| | | |
Collapse
|
18
|
Lambracht-Washington D, Rosenberg RN. Anti-amyloid beta to tau - based immunization: Developments in immunotherapy for Alzheimer disease. Immunotargets Ther 2013; 2013:105-114. [PMID: 24926455 PMCID: PMC4051350 DOI: 10.2147/itt.s31428] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy might provide an effective treatment for Alzheimer’s disease (AD). A unique feature of AD immunotherapies is that an immune response against a self-antigen needs to be elicited without causing adverse autoimmune reactions. Current research is focused on two possible targets in this regard. One is the inhibition of accumulation and deposition of amyloid beta 1–42 (Aβ42), which is one of the major peptides found in senile plaques, and the second target is hyperphosphorylated tau, which forms neurofibrillary tangles inside the nerve cell and shows association with the progression of dementia. Mouse models have shown that immunotherapy targeting Aβ42 as well as tau with the respective anti-Aβ or anti-tau antibodies can provide significant improvements in these mice. While anti-Aβ immunotherapy (active and passive immunizations) is already in several stages of clinical trials, tau-based immunizations have been analyzed only in mouse models. Recently, as a significant correlation of progression of dementia and levels of phosphorylated tau have been found, high interest has again focused on further development of tau-based therapies. While Aβ immunotherapy might delay the onset of AD, immunotherapy targeting tau might provide benefits in later stages of this disease. Last but not least, targeting Aβ and tau simultaneously with immunotherapy might provide additional therapeutic effects, as these two pathologies are likely synergistic; this is an approach that has not been tested yet. In this review, we will summarize animal models used to test possible therapies for AD, some of the facts about Aβ42 and tau biology, and present an overview on halted, ongoing, and upcoming clinical trials together with ongoing preclinical studies targeting tau or Aβ42.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer's disease: Past, present and future. Neuropharmacology 2013; 76 Pt A:27-50. [PMID: 23891641 DOI: 10.1016/j.neuropharm.2013.07.004] [Citation(s) in RCA: 531] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 06/26/2013] [Accepted: 07/02/2013] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. The etiology is multifactorial, and pathophysiology of the disease is complex. Data indicate an exponential rise in the number of cases of AD, emphasizing the need for developing an effective treatment. AD also imposes tremendous emotional and financial burden to the patient's family and community. The disease has been studied over a century, but acetylcholinesterase inhibitors and memantine are the only drugs currently approved for its management. These drugs provide symptomatic improvement alone but do less to modify the disease process. The extensive insight into the molecular and cellular pathomechanism in AD over the past few decades has provided us significant progress in the understanding of the disease. A number of novel strategies that seek to modify the disease process have been developed. The major developments in this direction are the amyloid and tau based therapeutics, which could hold the key to treatment of AD in the near future. Several putative drugs have been thoroughly investigated in preclinical studies, but many of them have failed to produce results in the clinical scenario; therefore it is only prudent that lessons be learnt from the past mistakes. The current rationales and targets evaluated for therapeutic benefit in AD are reviewed in this article. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- R Anand
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamilnadu, India.
| | | | | |
Collapse
|
20
|
Zhang L, Ma Q, Yang W, Qi X, Yao Z, Liu Y, Liang L, Wang X, Ma C, Huang L, Xu Y, Zhu H, Deng W, Gao Y, Ruan L, Xiao Z, Qin C. Recombinant DNA vaccine against neurite outgrowth inhibitors attenuates behavioral deficits and decreases Abeta in an Alzheimer's disease mouse model. Neuropharmacology 2013; 70:200-210. [PMID: 23201352 DOI: 10.1016/j.neuropharm.2012.10.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 10/15/2012] [Accepted: 10/19/2012] [Indexed: 01/30/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease that causes a progressive loss in learning and memory capabilities and eventually results in dementia. The non-renewable nature of neurons in the central nervous system leads to the basic pathological changes that are related to the various behavioral and psychological symptoms of AD. Oligodendrocyte- and myelin-related neurite outgrowth inhibitors (NOIs) tend to hinder the regeneration of neurons. We designed a recombinant DNA vaccine composed of multiple specific inhibitory domains of NOIs. Vaccination induced effective antibodies against the specific domains in the sera of mice treated with a DNA primed-vaccinia virus boost regimen. The vaccine attenuated neuronal degeneration in the mouse brain and protected the model mice from behavioral deficits. Vaccination also decreased the formation of soluble Aβ oligomer and amyloid plaques in the co-transgenic mice brain. What's more, astrocytosis in brains of APP/PS1 co-transgenic mice was also relieved. The results suggested that immunotherapy with multiple specific domains of myelin- and oligodendrocyte-related NOIs may be a promising approach for Alzheimer's disease and other degenerative central nervous system diseases.
Collapse
Affiliation(s)
- Lingling Zhang
- Comparative Medicine Center, Peking Union Medical College and Institute of Laboratory Animal Science, Chinese Academy of Medical Science, No 5 Pan Jia Yuan Nan Li, Chaoyang District, Beijing 100021, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lambracht-Washington D, Rosenberg RN. Advances in the development of vaccines for Alzheimer's disease. DISCOVERY MEDICINE 2013; 15:319-326. [PMID: 23725605 PMCID: PMC3696351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
One of the challenges of our society is to find a treatment or cure for Alzheimer's disease (AD). AD is the leading form of age-related dementia and with the increase of life expectancy worldwide, the social and economic burden from this disease will increase dramatically. It is a progressive and, in regard to clinical scores, a highly variable disease. AD pathogenesis has been associated with the accumulation, aggregation, and deposition of amyloid beta (Abeta) peptides in the brain. Hallmarks of AD are the amyloid plaques consisting of fibrillar Abeta and neurofibrillary tangles which are intracellular fibrils of hyperphosphorylated tau protein that develop later in this disease. The amyloid cascade hypothesis postulates that Abeta deposition is an initial event in the multifactorial pathogenesis and Abeta deposition may precede AD symptoms in some patients by at least 20 years. Amyloid beta therapy with active and passive immunizations against Abeta has a high possibility to be effective in removing Abeta from brain and might thus prevent the downstream pathology. Since 2000 a number of clinical trials for AD immunotherapy have started, have failed, and are continuing to be pursued. This article will review these clinical trials and ongoing research in this regard.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | |
Collapse
|
22
|
Restrepo L, Stafford P, Johnston SA. Feasibility of an early Alzheimer's disease immunosignature diagnostic test. J Neuroimmunol 2012; 254:154-60. [PMID: 23084373 DOI: 10.1016/j.jneuroim.2012.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/27/2012] [Accepted: 09/28/2012] [Indexed: 01/28/2023]
Abstract
A practical diagnostic test is needed for early Alzheimer's disease (AD) detection. Immunosignaturing, a technology that employs antibody binding to a random-sequence peptide microarray, generates profiles that distinguish transgenic mice engineered with familial AD mutations (APPswe/PSEN1-dE9) from non-transgenic littermates. It can also detect an AD-like signature in humans. Here, we assess the changes in the immunosignature at different time points of the disease in mice and humans. We also evaluate the accuracy of the late-stage signature as a test to discriminate between young mice with familial AD mutations from non-transgenic littermates. Plasma samples from AD patients were assayed 3-12 months apart, while APPswe/PSEN1-dE9 and non-transgenic controls supplied plasma at monthly intervals until they reached 15 months of age. Microarrays with 10,000 random-sequence peptides were used to compare antibody binding patterns. These patterns gradually changed over the life-span of mice. Strong, characteristic signatures were observed in transgenic mice at early, mid and late stages, but these profiles had minimal overlap. The signature of young transgenic mice had an error rate of 18% at classifying plasma samples from late-stage transgenic mice. Conversely, the late-stage transgenic mice signature discriminated between young transgenic mice and littermates with an error rate of 21%. Less distinctive profiles were recognizable throughout the transgenic mice lifespan, being detectable as early as 2 months. The human signature had minimal change on short-term follow-up. Our results call for a reappraisal of the way incipient AD is studied, as biomarkers seen in late-stages of the disease may not be relevant in earlier stages.
Collapse
Affiliation(s)
- Lucas Restrepo
- Center for Innovations in Medicine, The Biodesign Institute, Arizona State University, Tempe, AZ 85287-5901, United States
| | | | | |
Collapse
|
23
|
A peptide prime-DNA boost immunization protocol provides significant benefits as a new generation Aβ42 DNA vaccine for Alzheimer disease. J Neuroimmunol 2012; 254:63-8. [PMID: 23036592 DOI: 10.1016/j.jneuroim.2012.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 09/06/2012] [Accepted: 09/07/2012] [Indexed: 11/20/2022]
Abstract
Immunotherapy has the potential to provide a possible treatment therapy to prevent or delay Alzheimer disease. In a clinical trial (AN1792) in which patients received this immunotherapy and received active Aβ1-42 peptide immunizations, treatment was stopped when 6% of patients showed signs of meningoencephalitis. Follow up on these patients led to the conclusion that the antibody response was beneficial in removing Aβ1-42 from brain but an accompanying inflammatory Th1 T cell response was harmful. As a safe alternative treatment targeting the same self protein, Aβ1-42, in brain, we and others are working on a DNA Aβ1-42 immunization protocol as the immune response to DNA immunizations differs in many aspects from immunizations with peptide antigens. Because the immune response to DNA vaccination has different kinetics and has a significantly lower antibody production, we evaluated two different prime boost regimens, Aβ1-42 DNA prime/Aβ1-42 peptide boost and Aβ1-42 peptide prime/Aβ1-42 DNA boost for their effectiveness in antibody production and possible side effects due to inflammatory T cell responses. While both boost regimes significantly enhanced the specific antibody production with comparable antibody concentrations, the absence of the Aβ1-42 T cell response (no proliferation and no cytokine production) is consistent with our previous findings using this DNA Aβ1-42 trimer immunization and greatly enhances the safety aspect for possible clinical use.
Collapse
|
24
|
Al-Jumaily AM, Mbikou P, Redey PR. Effect of length oscillations on airway smooth muscle reactivity and cross-bridge cycling. Am J Physiol Lung Cell Mol Physiol 2012; 303:L286-94. [DOI: 10.1152/ajplung.00100.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Excessive airway narrowing due to airway smooth muscle (ASM) hyperconstriction is a major symptom in many respiratory diseases. In vitro imposition of length oscillations similar to those produced by tidal breathing on contracted ASM have shown to reduce muscle active forces, which is usually attributed to unconfirmed disruption of actomyosin cross-bridges. This research focuses on an in vitro investigation of the effect of mechanical oscillations on ASM reactivity and actomyosin cross-bridges. A computerized organ bath system was used to test maximally precontracted bovine ASM subjected to length oscillations at frequencies in the range of 10–100 Hz superimposed on tidal breathing oscillation. Using an immunofluorescence technique, two specific antibodies against the phospho-serine19 myosin light chain and the α-smooth muscle actin were used to analyze the colocalization between these two filaments. Data were processed using the plug-in “colocalization threshold” of ImageJ 1.43m software. The results demonstrate that both tidal and superimposed length oscillations reduce the active force in contracted ASM for a relatively long term and that the latter enhances the force reduction of the former. This reduction was also found to be frequency and time dependent. Additionally colocalization analysis indicates that length oscillations cause the detachment of the actomyosin connections and that this condition is sustained even after the cessation of the length oscillations.
Collapse
Affiliation(s)
- Ahmed M. Al-Jumaily
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| | - Prisca Mbikou
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| | - Prachi R. Redey
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
25
|
Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, Sha S, Liu L, Cao YP. Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Aβ3-10 reduces AD-like pathology and cognitive impairment in Tg-APPswe/PSEN1dE9 mice. J Neuroimmunol 2012; 249:16-26. [DOI: 10.1016/j.jneuroim.2012.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 04/09/2012] [Accepted: 04/15/2012] [Indexed: 10/28/2022]
|
26
|
Abstract
As a neurodegenerative disorder, Alzheimer disease (AD) is the most common form of dementia found in the aging population. Immunotherapy with passive or active immunizations targeting amyloid beta (Aβ) build-up in the brain may provide a possible treatment option and may help prevent AD from progressing. A number of passive immunizations with anti-Aβ42 antibodies are in different phases of clinical trials. One active immunization approach, AN-1792, was stopped after the development of autoimmune encephalitis in 6% of patients and a second one, CAD106, in which a small Aβ epitope is used, is currently in safety and tolerability studies. Besides active immunizations with proteins or peptides, active immunizations using DNA which codes for the protein against which the immune response will be directed, so called genetic immunizations, provide additional safety as the immune response in DNA immunizations differs quantitatively and qualitatively from the response elicited by peptide immunizations. In this review, we summarize our data using DNA Aβ42 immunizations in mouse models and discuss the results together with the results presented by other groups working on a DNA vaccine as treatment option for AD.
Collapse
|
27
|
Xing X, Sha S, Li Y, Zong L, Jiang T, Cao Y. Immunization with a new DNA vaccine for Alzheimer's disease elicited Th2 immune response in BALB/c mice by in vivo electroporation. J Neurol Sci 2011; 313:17-21. [PMID: 22029939 DOI: 10.1016/j.jns.2011.09.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 12/18/2022]
Abstract
Immunization with synthetic amyloid β-protein (Aβ) peptide has resulted in preventing and clearing Aβ deposits as well as improving cognitive function in transgenic mouse models of Alzheimer's disease (AD). But similar immunization studies in humans were halted due to the risk of inducing T cell-mediated meningoencephalitis. A safe and effective vaccine for AD requires not only therapeutic levels of anti-Aβ antibodies but also the prevention of an adverse T cell-mediated, proinflammatory autoimmune response. In this study, we developed a DNA vaccine, p(Aβ(3-10))(10)-IL-4, encoding ten tandem repeats of Aβ(3-10) fused with mouse cytokine interleukin-4 (IL-4) as a molecular adjuvant. Wild-type mice were injected intramuscularly with p(Aβ(3-10))(10)-IL-4 followed by in vivo electroporation. The p(Aβ(3-10))(10)-IL-4 vaccine elicited high titer anti-Aβ antibodies which bound to Aβ plaque in brain tissue from a ten-month-old APP/PS1 transgenic mouse. The antibody isotype was mainly IgG(1) and the IgG(1)/IgG(2a) ratio in the p(Aβ(3-10))(10)-IL-4 group was approximately eight times greater than that of the Aβ(42) group. Ex vivo cultured splenocytes isolated from mice immunized with p(Aβ(3-10))(10)-IL-4 exhibited a low IFN-γ response and a high IL-4 response compared with the control group. These results indicate that immunization with the p(Aβ(3-10))(10)-IL-4 vaccine induced effective anti-Aβ antibodies and elicited a Th2-polarized immune response that had a lower potential to cause an inflammatory T cell response. Thus, the DNA vaccine, p(Aβ(3-10))(10)-IL-4, may be a safe and efficient vaccine for AD.
Collapse
Affiliation(s)
- Xiaona Xing
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | | | | | | | | | | |
Collapse
|
28
|
Lambracht-Washington D, Qu BX, Fu M, Anderson LD, Stüve O, Eagar TN, Rosenberg RN. DNA immunization against amyloid beta 42 has high potential as safe therapy for Alzheimer's disease as it diminishes antigen-specific Th1 and Th17 cell proliferation. Cell Mol Neurobiol 2011; 31:867-74. [PMID: 21625960 DOI: 10.1007/s10571-011-9680-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 02/23/2011] [Indexed: 10/18/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) has been strongly associated with the accumulation of amyloid beta (Aβ) peptides in brain, and immunotherapy targeting Aβ provides potential for AD prevention. A clinical trial in which AD patients were immunized with Aβ42 peptide was stopped when 6% of participants showed meningoencephalitis, apparently due to an inflammatory Th1 immune response. Previously, we and other have shown that Aβ42 DNA vaccination via gene gun generates a Th2 cellular immune response, which was shown by analyses of the respective antibody isotype profiles. We also determined that in vitro T cell proliferation in response to Aβ42 peptide re-stimulation was absent in DNA Aβ42 trimer-immunized mice when compared to Aβ42 peptide-immunized mice. To further characterize this observation prospectively and longitudinally, we analyzed the immune response in wild-type mice after vaccination with Aβ42 trimer DNA and Aβ42 peptide with Quil A adjuvant. Wild-type mice were immunized with short-term (1-3× vaccinations) or long-term (6× vacinations) immunization strategies. Antibody titers and isotype profiles of the Aβ42 specific antibodies, as well as cytokine profiles and cell proliferation studies from this longitudinal study were determined. Sufficient antibody titers to effectively reduce Aβ42, but an absent T cell proliferative response and no IFNγ or IL-17 secretion after Aβ42 DNA trimer immunization minimizes the risk of inflammatory activities of the immune system towards the self antigen Aβ42 in brain. Therefore, Aβ42 DNA trimer immunization has a high probability to be effective and safe to treat patients with early AD.
Collapse
|
29
|
Combining NMR and X-ray Crystallography in Fragment-Based Drug Discovery: Discovery of Highly Potent and Selective BACE-1 Inhibitors. Top Curr Chem (Cham) 2011; 317:83-114. [DOI: 10.1007/128_2011_183] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Qu BX, Lambracht-Washington D, Fu M, Eagar TN, Stüve O, Rosenberg RN. Analysis of three plasmid systems for use in DNA A beta 42 immunization as therapy for Alzheimer's disease. Vaccine 2010; 28:5280-7. [PMID: 20562015 PMCID: PMC2926979 DOI: 10.1016/j.vaccine.2010.05.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 04/26/2010] [Accepted: 05/20/2010] [Indexed: 01/01/2023]
Abstract
In an effort to optimize DNA immunization-elicited antibody production responses against A beta 1-42 (A beta 42) as a therapy for Alzheimer's disease (AD), comparisons were made between three distinct plasmid systems using gene gun delivery. Plasmids encoding A beta 42 monomer and a novel A beta 42 trimeric fusion protein were evaluated in conjunction with CMV or Gal4/UAS promoter elements. It was found that vaccination A beta 42 trimer under the Gal4/UAS promoter elicited high levels of anti-A beta 42 antibody production. Serum antibody levels from Gal4/UAS-A beta 42 trimer immunized mice were found to be 16.6+/-5.5 microg/ml compared to 6.5+/-2.5 microg/ml with Gal4/UAS-A beta 42 monomer or even less with CMV-A beta 42 trimer. As compared to monomeric A beta 42 or A beta 42 trimer expressed under the CMV promoter, injection of the Gal4/UAS-A beta 42 trimer induced high levels of A beta 42 antigen expression in tissue suggesting a mechanism for the increase in anti-A beta 42 antibody. Antibodies were found to be primarily IgG1 suggesting a predominant Th2 response (IgG1/IgG2a ratio of 9). Serum from A beta 42 trimer-vaccinated mice was also found to identify amyloid plaques in the brains of APP/PS1 transgenic mice. These results demonstrate the potential therapeutic use of Gal4/UAS DNA A beta 42 trimer immunization in preventing Alzheimer's disease.
Collapse
Affiliation(s)
- Bao-Xi Qu
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9036, USA
| | | | | | | | | | | |
Collapse
|
31
|
Fu HJ, Liu B, Frost JL, Lemere CA. Amyloid-beta immunotherapy for Alzheimer's disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2010; 9:197-206. [PMID: 20205640 PMCID: PMC2895488 DOI: 10.2174/187152710791012017] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 12/12/2009] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a progressive, degenerative disorder of the brain and the most common form of dementia among the elderly. As the population grows and lifespan is extended, the number of AD patients will continue to rise. Current clinical therapies for AD provide partial symptomatic benefits for some patients; however, none of them modify disease progression. Amyloid-beta (Abeta) peptide, the major component of senile plaques in AD patients, is considered to play a crucial role in the pathogenesis of AD thereby leading to Abeta as a target for treatment. Abeta immunotherapy has been shown to induce a marked reduction in amyloid burden and an improvement in cognitive function in animal models. Although preclinical studies were successful, the initial human clinical trial of an active Abeta vaccine was halted due to the development of meningoencephalitis in approximately 6% of the vaccinated AD patients. Some encouraging outcomes, including signs of cognitive stabilization and apparent plaque clearance, were obtained in subset of patients who generated antibody titers. These promising preliminary data support further efforts to refine Abeta immunotherapy to produce highly effective and safer active and passive vaccines for AD. Furthermore, some new human clinical trials for both active and passive Abeta immunotherapy are underway. In this review, we will provide an update of Abeta immunotherapy in animal models and in human beings, as well as discuss the possible mechanisms underlying Abeta immunotherapy for AD.
Collapse
Affiliation(s)
- H J Fu
- Center for Neurologic Diseases, Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | | | | |
Collapse
|
32
|
Zhu Z, Sun ZY, Ye Y, Voigt J, Strickland C, Smith EM, Cumming J, Wang L, Wong J, Wang YS, Wyss DF, Chen X, Kuvelkar R, Kennedy ME, Favreau L, Parker E, McKittrick BA, Stamford A, Czarniecki M, Greenlee W, Hunter JC. Discovery of Cyclic Acylguanidines as Highly Potent and Selective β-Site Amyloid Cleaving Enzyme (BACE) Inhibitors: Part I—Inhibitor Design and Validation. J Med Chem 2009; 53:951-65. [DOI: 10.1021/jm901408p] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
Lambracht-Washington D, Qu BX, Fu M, Eagar TN, Stüve O, Rosenberg RN. DNA beta-amyloid(1-42) trimer immunization for Alzheimer disease in a wild-type mouse model. JAMA 2009; 302:1796-802. [PMID: 19861672 PMCID: PMC2896011 DOI: 10.1001/jama.2009.1547] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CONTEXT DNA beta-amyloid(1-42) (Abeta42) trimer immunization was developed to produce specific T helper 2 cell (T(H)2)-type antibodies to provide an effective and safe therapy for Alzheimer disease (AD) by reducing elevated levels of Abeta42 peptide that occur in the brain of patients with AD. OBJECTIVE To compare the immune response in wild-type mice after immunization with DNA Abeta42 trimer and Abeta42 peptide. DESIGN AND INTERVENTION Wild-type mice received either 4 microg of DNA Abeta42 trimer immunization administered with gene gun (n = 8) or intraperitoneal injection of 100 microg of human Abeta42 peptide with the adjuvant Quil A (n = 8). Titers, epitope mapping, and isotypes of the Abeta42-specific antibodies were analyzed. MAIN OUTCOME MEASURES Antibody titers, mapping of binding sites (epitopes), isotype profiles of the Abeta42-specific antibodies, and T-cell activation. RESULTS DNA Abeta42 trimer immunization resulted in antibody titers with a mean of 15 microg per milliliter of plasma. The isotype profile of the antibodies differed markedly. A predominant IgG1 antibody response was found in the DNA-immunized mice, indicating a T(H)2 type of immune response (IgG1/IgG2a ratio of 10). The peptide-immunized mice showed a mixed T(H)1/T(H)2 immune response (IgG1/IgG2a ratio of 1) (P < .001). No increased T-cell proliferation was observed in the DNA-immunized mice (P = .03). CONCLUSION In this preliminary study in a wild-type mouse model, DNA Abeta42 trimer immunization protocol produced a T(H)2 immune response and appeared to have low potential to cause an inflammatory T-cell response.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9108, USA
| | | | | | | | | | | |
Collapse
|
34
|
Suppression of nuclear factor kappa B ameliorates astrogliosis but not amyloid burden in APPswe/PS1dE9 mice. Neuroscience 2009; 161:53-8. [DOI: 10.1016/j.neuroscience.2009.03.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 03/04/2009] [Accepted: 03/05/2009] [Indexed: 11/23/2022]
|
35
|
DaSilva KA, Brown ME, McLaurin J. Reduced oligomeric and vascular amyloid-beta following immunization of TgCRND8 mice with an Alzheimer's DNA vaccine. Vaccine 2009; 27:1365-76. [PMID: 19150380 DOI: 10.1016/j.vaccine.2008.12.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 12/19/2008] [Accepted: 12/22/2008] [Indexed: 01/16/2023]
Abstract
Immunization with amyloid-beta (Abeta) peptide reduces amyloid load in animal studies and in humans; however clinical trials resulted in the development of a pro-inflammatory cellular response to Abeta. Apoptosis has been employed to stimulate humoral and Th2-biased cellular immune responses. Thus, we sought to investigate whether immunization using a DNA vaccine encoding Abeta in conjunction with an attenuated caspase generates therapeutically effective antibodies. Plasmids encoding Abeta and an attenuated caspase were less effective in reducing amyloid pathology than those encoding Abeta alone. Moreover, use of Abeta with an Arctic mutation (E22G) as an immunogen was less effective than wild-type Abeta in terms of improvements in pathology. Low levels of IgG and IgM were generated in response to immunization with a plasmid encoding wild-type Abeta. These antibodies decreased plaque load by as much as 36+/-8% and insoluble Abeta42 levels by 56+/-3%. Clearance of Abeta was most effective when antibodies were directed against N-terminal epitopes of Abeta. Moreover, immunization reduced CAA by as much as 69+/-12% in TgCRND8 mice. Finally, high-molecular-weight oligomers and Abeta trimers were significantly reduced with immunization. Thus, immunization with a plasmid encoding Abeta alone drives an attenuated immune response that is sufficient to clear amyloid pathology in a mouse model of Alzheimer's disease.
Collapse
Affiliation(s)
- Kevin A DaSilva
- Centre for Research in Neurodegenerative Diseases, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
36
|
Abstract
The amyloid deposits in the brains of patients with Alzheimer's disease have attracted attention and are now considered not only an essential requirement for pathological diagnosis but also an important clue to the pathogenesis of the disease. This article looks critically at the basis of the "amyloid cascade hypothesis" to conclude that there are many issues that need to be resolved since they are inconsistent with the hypothesis.
Collapse
Affiliation(s)
- Amos D Korczyn
- Sieratzki Chair of Neurology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
37
|
Abstract
Alzheimer disease is the most common cause of dementia and represents a major public health problem. The neuropathologic findings of amyloid-beta plaques and tau containing neurofibrillary tangles represent important molecular clues to the underlying pathogenesis. Genetic factors are well recognized, but complicated. Three rare forms of autosomal-dominant early-onset familial Alzheimer disease have been identified and are associated with mutations in amyloid precursor protein, presenilin 1, and presenilin 2 genes. The more common late-onset form of Alzheimer disease is assumed to be polygenic/multifactorial. However, thus far the only clearly identified genetic risk factor for Alzheimer disease is Apo lipoprotein E. The epsilon4 allele of Apo lipoprotein E influences age at onset of Alzheimer disease, but is neither necessary nor sufficient for the disease. The search continues for the discovery of additional genetic influences.
Collapse
Affiliation(s)
- Thomas D Bird
- University of Washington, Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA.
| |
Collapse
|
38
|
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized pathologically by the deposition of beta-amyloid (A beta)-containing extracellular neuritic plaques, intracellular neurofibrillary tangles and neuronal loss. Much evidence supports the hypothesis that A beta peptide aggregation contributes to AD pathogenesis, however, currently approved therapeutic treatments do nothing to stop or reverse A beta deposition. The success of active and passive anti-A beta immunotherapies in both preventing and clearing parenchymal amyloid in transgenic mouse models led to the initiation of an active anti-A beta vaccination (AN1792) trial in human patients with mild-to-moderate AD, but was prematurely halted when 6% of inoculated patients developed aseptic meningoencephalitis. Autopsy results from the brains of four individuals treated with AN1792 revealed decreased plaque burden in select brain areas, as well as T-cell lymphocytes in three of the patients. Furthermore, antibody responders showed some improvement in memory task measures. These findings indicated that anti-A beta therapy might still be a viable option for the treatment of AD, if potentially harmful proinflammatory processes can be avoided. Over the past 6 years, this target has led to the development of novel experimental immunization strategies, including selective A beta epitope targeting, antibody and adjuvant modifications, as well as alternative routes and mechanisms of vaccine delivery, to generate anti-A beta antibodies that selectively target and remove specific A beta species without evoking autoimmunity. Results from the passive vaccination AD clinical trials that are currently underway will provide invaluable information about both the effectiveness of newly improved anti-A beta vaccines in clinical treatment, as well as the role of the A beta peptide in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Cheryl A Hawkes
- Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
39
|
Spencer B, Rockenstein E, Crews L, Marr R, Masliah E. Novel strategies for Alzheimer's disease treatment. Expert Opin Biol Ther 2007; 7:1853-67. [PMID: 18034651 DOI: 10.1517/14712598.7.12.1853] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Considerable progress has been made in recent years towards better understanding the pathogenesis of Alzheimer's disease (AD), a dementing neurodegenerative disorder that affects > 10 million individuals in the US and Europe combined. Recent studies suggest that alterations in the processing of amyloid precursor protein (APP), resulting in the accumulation of amyloid-beta protein (Abeta) and the formation of oligomers leads to synaptic damage and neurodegeneration. Therefore, strategies for treatment development have been focused on reducing Abeta accumulation using, among other approaches, antiaggregation molecules, regulators of the APP proteolysis and processing, reducing APP production (e.g., small-interfering RNA), and increasing Abeta clearance with antibodies, apolipoprotein E and Abeta-degrading enzymes (e.g., neprilysin). The main focus of this review is on novel treatments for AD with a special emphasis on delivering neuroprotective and antiamyloidogenic molecules by gene therapy and by promoting neurogenesis.
Collapse
Affiliation(s)
- Brian Spencer
- University of California, Department of Neurosciences, San Diego, La Jolla, CA 92093-0624, USA
| | | | | | | | | |
Collapse
|
40
|
Qu BX, Xiang Q, Li L, Johnston SA, Hynan LS, Rosenberg RN. Abeta42 gene vaccine prevents Abeta42 deposition in brain of double transgenic mice. J Neurol Sci 2007; 260:204-13. [PMID: 17574274 PMCID: PMC2587214 DOI: 10.1016/j.jns.2007.05.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2007] [Revised: 05/09/2007] [Accepted: 05/10/2007] [Indexed: 01/10/2023]
Abstract
Abeta42 peptide aggregation and deposition is an important component of the neuropathology of Alzheimer's disease (AD). Gene-gun mediated gene vaccination targeting Abeta42 is a potential method to prevent and treat AD. APPswe/PS1DeltaE9 transgenic (Tg) mice were immunized with an Abeta42 gene construct delivered by the gene gun. The vaccinated mice developed Th2 antibodies (IgG1) against Abeta42. The Abeta42 levels in brain were decreased by 41% and increased in plasma 43% in the vaccinated compared with control mice as assessed by ELISA analysis. Abeta42 plaque deposits in cerebral cortex and hippocampus were reduced by 51% and 52%, respectively, as shown by quantitative immunolabeling. Glial cell activation was also significantly attenuated in vaccinated compared with control mice. One rhesus monkey was vaccinated and developed anti-Abeta42 antibody. These new findings advance significantly our knowledge that gene-gun mediated Abeta42 gene immunization effectively induces a Th2 immune response and reduces the Abeta42 levels in brain in APPswe/PS1DeltaE9 mice. Abeta42 gene vaccination may be safe and efficient immunotherapy for AD.
Collapse
Affiliation(s)
- Bao-Xi Qu
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qun Xiang
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Liping Li
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Stephen Albert Johnston
- The Center for Innovations in Medicine/Biodesign Institute, The Arizona State University, Tempe, Arizona, USA
| | - Linda S. Hynan
- Alzheimer's Disease Center, Departments of Clinical Sciences (Biostatistics) and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Roger N. Rosenberg
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Vasir JK, Labhasetwar V. Biodegradable nanoparticles for cytosolic delivery of therapeutics. Adv Drug Deliv Rev 2007; 59:718-28. [PMID: 17683826 PMCID: PMC2002520 DOI: 10.1016/j.addr.2007.06.003] [Citation(s) in RCA: 351] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Accepted: 06/12/2007] [Indexed: 12/14/2022]
Abstract
Many therapeutics require efficient cytosolic delivery either because the receptors for those drugs are located in the cytosol or their site of action is an intracellular organelle that requires transport through the cytosolic compartment. To achieve efficient cytosolic delivery of therapeutics, different nanomaterials have been developed that consider the diverse physicochemical nature of therapeutics (macromolecule to small molecule; water soluble to water insoluble) and various membrane associated and intracellular barriers that these systems need to overcome to efficiently deliver and retain therapeutics in the cytoplasmic compartment. Our interest is in investigating PLGA and PLA-based nanoparticles for intracellular delivery of drugs and genes. The present review discusses the various aspects of our studies and emphasizes the need for understanding of the molecular mechanisms of intracellular trafficking of nanoparticles in order to develop an efficient cytosolic delivery system.
Collapse
Affiliation(s)
- Jaspreet K Vasir
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
42
|
Abstract
Recent insights into the function and dysfunction of microglia may inform future therapies to combat neurodegeneration. We hypothesise how different aspects of microglial activity including migration, activation, oxidative response, phagocytosis, proteolysis, and replenishment could be targeted by novel therapeutic approaches. A combined approach is suggested, encompassing opsonization and anti-inflammatory strategies in conjunction with an engineering of microglial precursors. Xenoproteases for bioremediation could be used to enhance intracellular and extracellular proteolytic capacity. The capacity of microglial precursors to cross the blood-brain barrier and to home in on sites of neural damage and inflammation might prove to be particularly useful for future therapeutic strategies.
Collapse
Affiliation(s)
- John Schloendorn
- Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA.
| | | | | |
Collapse
|
43
|
Muhs A, Hickman DT, Pihlgren M, Chuard N, Giriens V, Meerschman C, van der Auwera I, van Leuven F, Sugawara M, Weingertner MC, Bechinger B, Greferath R, Kolonko N, Nagel-Steger L, Riesner D, Brady RO, Pfeifer A, Nicolau C. Liposomal vaccines with conformation-specific amyloid peptide antigens define immune response and efficacy in APP transgenic mice. Proc Natl Acad Sci U S A 2007; 104:9810-5. [PMID: 17517595 PMCID: PMC1887581 DOI: 10.1073/pnas.0703137104] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We investigated the therapeutic effects of two different versions of Abeta(1-15 (16)) liposome-based vaccines. Inoculation of APP-V717IxPS-1 (APPxPS-1) double-transgenic mice with tetra-palmitoylated amyloid 1-15 peptide (palmAbeta(1-15)), or with amyloid 1-16 peptide (PEG-Abeta(1-16)) linked to a polyethyleneglycol spacer at each end, and embedded within a liposome membrane, elicited fast immune responses with identical binding epitopes. PalmAbeta(1-15) liposomal vaccine elicited an immune response that restored the memory defect of the mice, whereas that of PEG-Abeta(1-16) had no such effect. Immunoglobulins that were generated were predominantly of the IgG class with palmAbeta(1-15), whereas those elicited by PEG-Abeta(1-16) were primarily of the IgM class. The IgG subclasses of the antibodies generated by both vaccines were mostly IgG2b indicating noninflammatory Th2 isotype. CD and NMR revealed predominantly beta-sheet conformation of palmAbeta(1-15) and random coil of PEG-Abeta(1-16). We conclude that the association with liposomes induced a variation of the immunogenic structures and thereby different immunogenicities. This finding supports the hypothesis that Alzheimer's disease is a "conformational" disease, implying that antibodies against amyloid sequences in the beta-sheet conformation are preferred as potential therapeutic agents.
Collapse
Affiliation(s)
- Andreas Muhs
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - David T. Hickman
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maria Pihlgren
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Nathalie Chuard
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Valérie Giriens
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Carine Meerschman
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Fred van Leuven
- Experimental Genetics Group, KULeuven, B-3000 Leuven, Belgium
| | - Masae Sugawara
- Institut de Science et d'Ingénierie Supramoléculaires, Université Louis Pasteur, F-67083 Strasbourg, France
| | | | - Burkhard Bechinger
- Institut de Science et d'Ingénierie Supramoléculaires, Université Louis Pasteur, F-67083 Strasbourg, France
| | - Ruth Greferath
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Nadine Kolonko
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Luitgard Nagel-Steger
- Institut für Biophysikalische Chemie, Heinrich-Heine Universität, 40225 Düsseldorf, Germany
| | - Detlev Riesner
- Institut für Biophysikalische Chemie, Heinrich-Heine Universität, 40225 Düsseldorf, Germany
| | - Roscoe O. Brady
- Developmental and Metabolic Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892; and
- **To whom correspondence may be addressed. E-mail: or
| | - Andrea Pfeifer
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Claude Nicolau
- *AC Immune, PSE-B, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111
- **To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW In this paper, we review current concepts of Alzheimer's disease, recent progress in diagnosis and treatment and important developments in our understanding of its pathogenesis with a focus on beta-amyloid both as culprit and therapeutic target. RECENT FINDINGS The amyloid cascade hypothesis of Alzheimer's disease pathogenesis continues to predominate with evidence suggesting that small oligomeric forms of Abeta-42 rather than fibrils or senile plaques are the key pathological substrates. The concept of mild cognitive impairment continues to be refined to define better those patients who will progress to Alzheimer's disease. Structural and functional imaging techniques and cerebrospinal fluid biomarkers are gaining acceptance as diagnostic markers of Alzheimer's disease, with a potentially exciting advance being the ability to image amyloid in vivo using novel positron emission tomography ligands. Whilst available treatments afford only symptomatic benefits, disease-modifying treatments may be within reach. Despite the halting of the first amyloid beta-vaccination trial due to adverse effects, amyloid immunotherapy continues to show promise, with new approaches already entering clinical trials. Other therapeutic strategies under investigation include inhibition of beta -and gamma-secretase, key enzymes implicated in Alzheimer's disease pathogenesis. SUMMARY Current research demonstrates the potential for diagnostic strategies and disease modifying treatments to follow from an ever more detailed understanding of the molecular mechanisms underlying the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jonathan M Schott
- Institute of Neurology, Dementia Research Centre, National Hospital for Neurology and Neurosurgery, London, UK
| | | | | |
Collapse
|
45
|
Thanvi B, Robinson T. Sporadic cerebral amyloid angiopathy--an important cause of cerebral haemorrhage in older people. Age Ageing 2006; 35:565-71. [PMID: 16982664 DOI: 10.1093/ageing/afl108] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an important cause of primary intracerebral haemorrhage (PICH) in older people, accounting for approximately 10% of all types of PICH. The amount of amyloid deposition in the vessels and vasculopathic changes determine the propensity to PICH. The risk factors of CAA include advanced age and the presence of certain alleles of apolipoprotein E. There are no specific clinical features of CAA-related PICH, although lobar, recurrent or multiple simultaneous haemorrhages in older patients should raise suspicion of its diagnosis. A definitive diagnosis of CAA requires pathological examination of the affected tissue. However, with modern imaging techniques, it is possible to make a diagnosis of 'probable CAA' in patients presenting with PICH. Gradient-echo magnetic resonance imaging is a sensitive, non-invasive technique for identifying small haemorrhages in life. Currently, there is no specific treatment available for CAA. Recent advances in the immunopathology and pathogenesis of CAA are expected to help in developing specific anti-amyloid therapy.
Collapse
Affiliation(s)
- Bhomraj Thanvi
- Leicester General Hospital, Medicine for the Care of Older People, Leicester, UK.
| | | |
Collapse
|