251
|
Engelender S, Isacson O. The Threshold Theory for Parkinson's Disease. Trends Neurosci 2016; 40:4-14. [PMID: 27894611 DOI: 10.1016/j.tins.2016.10.008] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 01/17/2023]
Abstract
Parkinson's disease (PD) is recognized by the accumulation of α-synuclein within neurons. In contrast to the current ascending theory where α-synuclein would propagate from neuron to neuron, we now propose the threshold theory for PD based on evidence of parallel degeneration of both central nervous system (CNS) and peripheral nervous system (PNS) in PD. The functional threshold is lower for the emergence of early symptoms before the classical motor symptoms of PD. This is due to the larger functional reserve of the midbrain dopamine and integrated basal ganglia motor systems to control movement. This threshold theory better accounts for the current neurobiology of PD symptom progression compared to the hypothesis that the disease ascends from the PNS to the CNS as proposed by Braak's hypothesis.
Collapse
Affiliation(s)
- Simone Engelender
- Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital, Belmont, MA 02478, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
252
|
Tatarnikova OG, Orlov MA, Bobkova NV. Beta-Amyloid and Tau-Protein: Structure, Interaction, and Prion-Like Properties. BIOCHEMISTRY (MOSCOW) 2016; 80:1800-19. [PMID: 26878581 DOI: 10.1134/s000629791513012x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
During the last twenty years, molecular genetic investigations of Alzheimer's disease (AD) have significantly broadened our knowledge of basic mechanisms of this disorder. However, still no unambiguous concept on the molecular bases of AD pathogenesis has been elaborated, which significantly impedes the development of AD therapy. In this review, we analyze issues concerning processes of generation of two proteins (β-amyloid peptide and Tau-protein) in the cell, which are believed to play the key role in AD genesis. Until recently, these agents were considered independently of each other, but in light of the latest studies, it becomes clear that it is necessary to study their interaction and combined effects. Studies of mechanisms of toxic action of these endogenous compounds, beginning from their interaction with known receptors of main neurotransmitters to specific peculiarities of functioning of signal intracellular pathways upon development of this pathology, open the way to development of new pharmaceutical substances directed concurrently on key mechanisms of interaction of toxic proteins inside the cell and on the pathways of their propagation in the extracellular space.
Collapse
Affiliation(s)
- O G Tatarnikova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | | | | |
Collapse
|
253
|
Hara H, Ono F, Nakamura S, Matsumoto SE, Jin H, Hattori N, Tabira T. An Oral Aβ Vaccine Using a Recombinant Adeno-Associated Virus Vector in Aged Monkeys: Reduction in Plaque Amyloid and Increase in Aβ Oligomers. J Alzheimers Dis 2016; 54:1047-1059. [DOI: 10.3233/jad-160514] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hideo Hara
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Fumiko Ono
- The Corporation for Production and Research of Laboratory Primates, Tsukuba, Ibaraki, Japan
- Faculty of Risk and Crisis Management, Chiba Institute of Science, Shiomi, Choshi, Chiba, Japan
| | - Shinichiro Nakamura
- The Corporation for Production and Research of Laboratory Primates, Tsukuba, Ibaraki, Japan
- Shiga University of Medical Science, Research Center for Animal Life Science, Seta-Tsukinowa, Otsu, Shiga, Japan
| | - Shin-ei Matsumoto
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Haifeng Jin
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takeshi Tabira
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
254
|
MacLellan A, Breiner A, Tang-Wai DF, Casaubon LK. Clinical Reasoning: A case of subacute cognitive decline in a 76-year-old man. Neurology 2016; 87:e124-8. [PMID: 27647588 DOI: 10.1212/wnl.0000000000003131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Adam MacLellan
- From the Division of Neurology, University Health Network, Toronto, Canada
| | - Ari Breiner
- From the Division of Neurology, University Health Network, Toronto, Canada
| | - David F Tang-Wai
- From the Division of Neurology, University Health Network, Toronto, Canada
| | - Leanne K Casaubon
- From the Division of Neurology, University Health Network, Toronto, Canada.
| |
Collapse
|
255
|
Echeverria V, Yarkov A, Aliev G. Positive modulators of the α7 nicotinic receptor against neuroinflammation and cognitive impairment in Alzheimer's disease. Prog Neurobiol 2016; 144:142-57. [DOI: 10.1016/j.pneurobio.2016.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 11/07/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
|
256
|
Sterner RM, Takahashi PY, Yu Ballard AC. Active Vaccines for Alzheimer Disease Treatment. J Am Med Dir Assoc 2016; 17:862.e11-5. [PMID: 27461865 DOI: 10.1016/j.jamda.2016.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Vaccination against peptides specific to Alzheimer disease may generate an immune response that could help inhibit disease and symptom progression. METHODS PubMed and Scopus were searched for clinical trial articles, review articles, and preclinical studies relevant to the field of active Alzheimer disease vaccines and raw searches yielded articles ranging from 2016 to 1973. ClinicalTrials.gov was searched for active Alzheimer disease vaccine trials. Manual research and cross-referencing from reviews and original articles was performed. RESULTS First generation Aβ42 phase 2a trial in patients with mild to moderate Alzheimer disease resulted in cases of meningoencephalitis in 6% of patients, so next generation vaccines are working to target more specific epitopes to induce a more controlled immune response. Difficulty in developing these vaccines resides in striking a balance between providing a vaccine that induces enough of an immune response to actually clear protein sustainably but not so much of a response that results in excess immune activation and possibly adverse effects such as meningoencephalitis. CONCLUSIONS Although much work still needs to be done in the field to make this a practical possibility, the enticing allure of being able to treat or even prevent the extraordinarily impactful disease that is Alzheimer disease makes the idea of active vaccination for Alzheimer disease very appealing and something worth striving toward.
Collapse
|
257
|
Lee KL, Twyman RM, Fiering S, Steinmetz N. Virus-based nanoparticles as platform technologies for modern vaccines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:554-78. [PMID: 26782096 PMCID: PMC5638654 DOI: 10.1002/wnan.1383] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/11/2015] [Indexed: 12/25/2022]
Abstract
Nanoscale engineering is revolutionizing the development of vaccines and immunotherapies. Viruses have played a key role in this field because they can function as prefabricated nanoscaffolds with unique properties that are easy to modify. Viruses are immunogenic via multiple pathways, and antigens displayed naturally or by engineering on the surface can be used to create vaccines against the cognate virus, other pathogens, specific molecules or cellular targets such as tumors. This review focuses on the development of virus-based nanoparticle systems as vaccines indicated for the prevention or treatment of infectious diseases, chronic diseases, cancer, and addiction. WIREs Nanomed Nanobiotechnol 2016, 8:554-578. doi: 10.1002/wnan.1383 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Karin L. Lee
- Department of Biomedical Engineering, Case Western Reserve University Schools of Engineering and Medicine, Cleveland, OH 44106
| | | | - Steven Fiering
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Nicole Steinmetz
- Departments of Biomedical Engineering, Radiology, Materials Science and Engineering, and Macromolecular Science and Engineering, Case Western Reserve University and Medicine, Cleveland, OH 44106;
| |
Collapse
|
258
|
Amanatkar HR, Papagiannopoulos B, Grossberg GT. Analysis of recent failures of disease modifying therapies in Alzheimer’s disease suggesting a new methodology for future studies. Expert Rev Neurother 2016; 17:7-16. [DOI: 10.1080/14737175.2016.1194203] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Hamid Reza Amanatkar
- Department of Psychiatry, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Bill Papagiannopoulos
- Department of Psychiatry, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | | |
Collapse
|
259
|
Current pharmacotherapy and putative disease-modifying therapy for Alzheimer's disease. Neurol Sci 2016; 37:1403-35. [PMID: 27250365 DOI: 10.1007/s10072-016-2625-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease of the central nervous system correlated with the progressive loss of cognition and memory. β-Amyloid plaques, neurofibrillary tangles and the deficiency in cholinergic neurotransmission constitute the major hallmarks of the AD. Two major hypotheses have been implicated in the pathogenesis of AD namely the cholinergic hypothesis which ascribed the clinical features of dementia to the deficit cholinergic neurotransmission and the amyloid cascade hypothesis which emphasized on the deposition of insoluble peptides formed due to the faulty cleavage of the amyloid precursor protein. Current pharmacotherapy includes mainly the acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor agonist which offer symptomatic therapy and does not address the underlying cause of the disease. The disease-modifying therapy has garnered a lot of research interest for the development of effective pharmacotherapy for AD. β and γ-Secretase constitute attractive targets that are focussed in the disease-modifying approach. Potentiation of α-secretase also seems to be a promising approach towards the development of an effective anti-Alzheimer therapy. Additionally, the ameliorative agents that prevent aggregation of amyloid peptide and also the ones that modulate inflammation and oxidative damage associated with the disease are focussed upon. Development in the area of the vaccines is in progress to combat the characteristic hallmarks of the disease. Use of cholesterol-lowering agents also is a fruitful strategy for the alleviation of the disease as a close association between the cholesterol and AD has been cited. The present review underlines the major therapeutic strategies for AD with focus on the new developments that are on their way to amend the current therapeutic scenario of the disease.
Collapse
|
260
|
Ribarič S. The Rationale for Insulin Therapy in Alzheimer's Disease. Molecules 2016; 21:molecules21060689. [PMID: 27240327 PMCID: PMC6273626 DOI: 10.3390/molecules21060689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a prevalence that increases with age. By 2050, the worldwide number of patients with AD is projected to reach more than 140 million. The prominent signs of AD are progressive memory loss, accompanied by a gradual decline in cognitive function and premature death. AD is the clinical manifestation of altered proteostasis. The initiating step of altered proteostasis in most AD patients is not known. The progression of AD is accelerated by several chronic disorders, among which the contribution of diabetes to AD is well understood at the cell biology level. The pathological mechanisms of AD and diabetes interact and tend to reinforce each other, thus accelerating cognitive impairment. At present, only symptomatic interventions are available for treating AD. To optimise symptomatic treatment, a personalised therapy approach has been suggested. Intranasal insulin administration seems to open the possibility for a safe, and at least in the short term, effective symptomatic intervention that delays loss of cognition in AD patients. This review summarizes the interactions of AD and diabetes from the cell biology to the patient level and the clinical results of intranasal insulin treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
261
|
Longitudinal noninvasive magnetic resonance imaging of brain microhemorrhages in BACE inhibitor-treated APP transgenic mice. Neurobiol Aging 2016; 45:50-60. [PMID: 27459925 DOI: 10.1016/j.neurobiolaging.2016.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/12/2016] [Accepted: 05/10/2016] [Indexed: 12/16/2022]
Abstract
Currently, several immunotherapies and BACE (Beta Site APP Cleaving Enzyme) inhibitor approaches are being tested in the clinic for the treatment of Alzheimer's disease. A crucial mechanism-related safety concern is the exacerbation of microhemorrhages, which are already present in the majority of Alzheimer patients. To investigate potential safety liabilities of long-term BACE inhibitor therapy, we used aged amyloid precursor protein (APP) transgenic mice (APP23), which robustly develop cerebral amyloid angiopathy. T2*-weighted magnetic resonance imaging (MRI), a translational method applicable in preclinical and clinical studies, was used for the detection of microhemorrhages throughout the entire brain, with subsequent histological validation. Three-dimensional reconstruction based on in vivo MRI and serial Perls' stained sections demonstrated a one-to-one matching of the lesions thus allowing for their histopathological characterization. MRI detected small Perls' positive areas with a high spatial resolution. Our data demonstrate that volumetric assessment by noninvasive MRI is well suited to monitor cerebral microhemorrhages in vivo. Furthermore, 3 months treatment of aged APP23 with the potent BACE-inhibitor NB-360 did not exacerbate microhemorrhages in contrast to Aβ-antibody β1. These results substantiate the safe use of BACE inhibitors regarding microhemorrhages in long-term clinical studies for the treatment of Alzheimer's disease.
Collapse
|
262
|
Bigaeva E, van Doorn E, Liu H, Hak E. Meta-Analysis on Randomized Controlled Trials of Vaccines with QS-21 or ISCOMATRIX Adjuvant: Safety and Tolerability. PLoS One 2016; 11:e0154757. [PMID: 27149269 PMCID: PMC4858302 DOI: 10.1371/journal.pone.0154757] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/19/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES QS-21 shows in vitro hemolytic effect and causes side effects in vivo. New saponin adjuvant formulations with better toxicity profiles are needed. This study aims to evaluate the safety and tolerability of QS-21 and the improved saponin adjuvants (ISCOM, ISCOMATRIX and Matrix-M™) from vaccine trials. METHODS A systematic literature search was conducted from MEDLINE, EMBASE, Cochrane library and Clinicaltrials.gov. We selected for the meta-analysis randomized controlled trials (RCTs) of vaccines adjuvanted with QS-21, ISCOM, ISCOMATRIX or Matrix-M™, which included a placebo control group and reported safety outcomes. Pooled risk ratios (RRs) and their 95% confidence intervals (CIs) were calculated using a random-effects model. Jadad scale was used to assess the study quality. RESULTS Nine RCTs were eligible for the meta-analysis: six trials on QS-21-adjuvanted vaccines and three trials on ISCOMATRIX-adjuvanted, with 907 patients in total. There were no studies on ISCOM or Matrix-M™ adjuvanted vaccines matching the inclusion criteria. Meta-analysis identified an increased risk for diarrhea in patients receiving QS21-adjuvanted vaccines (RR 2.55, 95% CI 1.04-6.24). No increase in the incidence of the reported systemic AEs was observed for ISCOMATRIX-adjuvanted vaccines. QS-21- and ISCOMATRIX-adjuvanted vaccines caused a significantly higher incidence of injection site pain (RR 4.11, 95% CI 1.10-15.35 and RR 2.55, 95% CI 1.41-4.59, respectively). ISCOMATRIX-adjuvanted vaccines also increased the incidence of injection site swelling (RR 3.43, 95% CI 1.08-10.97). CONCLUSIONS Our findings suggest that vaccines adjuvanted with either QS-21 or ISCOMATRIX posed no specific safety concern. Furthermore, our results indicate that the use of ISCOMATRIX enables a better systemic tolerability profile when compared to the use of QS-21. However, no better local tolerance was observed for ISCOMATRIX-adjuvanted vaccines in immunized non-healthy subjects. This meta-analysis is limited by the relatively small number of individuals recruited in the included trials, especially in the control groups.
Collapse
Affiliation(s)
- Emilia Bigaeva
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Eva van Doorn
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| | - Heng Liu
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| | - Eelko Hak
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| |
Collapse
|
263
|
Kudrna JJ, Ugen KE. Gene-based vaccines and immunotherapeutic strategies against neurodegenerative diseases: Potential utility and limitations. Hum Vaccin Immunother 2016; 11:1921-6. [PMID: 26125436 DOI: 10.1080/21645515.2015.1065364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
There has been a recent expansion of vaccination and immunotherapeutic strategies from controlling infectious diseases to the targeting of non-infectious conditions including neurodegenerative disorders. In addition to conventional vaccine and immunotherapeutic modalities, gene-based methods that express antigens for presentation to the immune system by either live viral vectors or non-viral naked DNA plasmids have been developed and evaluated. This mini-review/commentary summarizes the advantages and disadvantages, as well as the research findings to date, of both of these gene-based vaccination approaches in terms of how they can be targeted against appropriate antigens within the Alzheimer and Parkinson disease pathogenesis processes as well as potentially against targets in other neurodegenerative diseases. Most recently, the novel utilization of these viral vector and naked DNA gene-based technologies includes the delivery of immunoglobulin genes from established biologically active monoclonal antibodies. This modified passive immunotherapeutic strategy has recently been applied to deliver passive antibody immunotherapy against the pathologically relevant amyloid β protein in Alzheimer disease. The advantages and disadvantages of this technological application of gene-based immune interventions, as well as research findings to date are also summarized. In sum, it is suggested that further evaluation of gene based vaccines and immunotherapies against neurodegenerative diseases are warranted to determine their potential clinical utility.
Collapse
Affiliation(s)
- Jeremy J Kudrna
- a Department of Molecular Medicine ; Morsani College of Medicine; University of South Florida ; Tampa , FL USA
| | | |
Collapse
|
264
|
Structure of amyloid oligomers and their mechanisms of toxicities: Targeting amyloid oligomers using novel therapeutic approaches. Eur J Med Chem 2016; 114:41-58. [DOI: 10.1016/j.ejmech.2016.02.065] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 01/22/2023]
|
265
|
Winblad B, Amouyel P, Andrieu S, Ballard C, Brayne C, Brodaty H, Cedazo-Minguez A, Dubois B, Edvardsson D, Feldman H, Fratiglioni L, Frisoni GB, Gauthier S, Georges J, Graff C, Iqbal K, Jessen F, Johansson G, Jönsson L, Kivipelto M, Knapp M, Mangialasche F, Melis R, Nordberg A, Rikkert MO, Qiu C, Sakmar TP, Scheltens P, Schneider LS, Sperling R, Tjernberg LO, Waldemar G, Wimo A, Zetterberg H. Defeating Alzheimer's disease and other dementias: a priority for European science and society. Lancet Neurol 2016; 15:455-532. [DOI: 10.1016/s1474-4422(16)00062-4] [Citation(s) in RCA: 1001] [Impact Index Per Article: 111.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/06/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
|
266
|
St-Amour I, Cicchetti F, Calon F. Immunotherapies in Alzheimer's disease: Too much, too little, too late or off-target? Acta Neuropathol 2016; 131:481-504. [PMID: 26689922 DOI: 10.1007/s00401-015-1518-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/12/2015] [Accepted: 12/03/2015] [Indexed: 12/19/2022]
Abstract
Years of research have highlighted the importance of the immune system in Alzheimer's disease (AD), a system that, if manipulated during strategic time windows, could potentially be tackled to treat this disorder. However, to minimize adverse effects, it is essential to first grasp which exact aspect of it may be targeted. Several clues have been collected over the years regarding specific immune players strongly modulated during different stages of AD progression. However, the inherent complexity of the immune system as well as conflicting data make it quite challenging to pinpoint a specific immune target in AD. In this review, we discuss immune-related abnormalities observed in the periphery as well as in the brain of AD patients, in relation to known risk factors of AD such as genetics, type-2 diabetes or obesity, aging, physical inactivity and hypertension. Although not investigated yet in clinical trials, C5 complement system component, CD40/CD40L interactions and the CXCR2 pathway are altered in AD patients and may represent potential therapeutic targets. Immunotherapies tested in a clinical context, those aiming to attenuate the innate immune response and those used to facilitate the removal of pathological proteins, are further discussed to try and understand the causes of the limited success reached. The prevailing eagerness to move basic research data to clinic should not overshadow the fact that a careful preclinical characterization of a drug is still required to ultimately improve the chance of clinical success. Finally, specific elements to consider prior to initiate large-scale trials are highlighted and include the replication of preclinical data, the use of small-scale human studies, the sub-typing of AD patients and the determination of pharmacokinetic and pharmacodynamics parameters such as brain bioavailability and target engagement.
Collapse
Affiliation(s)
- Isabelle St-Amour
- Axe Neurosciences, Centre de Recherche du CHU de Québec, 2705, Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
- Département de Psychiatrie & Neurosciences, Faculté de médecine, Université Laval, Quebec, QC, Canada
- Faculté de pharmacie, Université Laval, Quebec, QC, Canada
| | - Francesca Cicchetti
- Axe Neurosciences, Centre de Recherche du CHU de Québec, 2705, Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
- Département de Psychiatrie & Neurosciences, Faculté de médecine, Université Laval, Quebec, QC, Canada
| | - Frédéric Calon
- Axe Neurosciences, Centre de Recherche du CHU de Québec, 2705, Boulevard Laurier, Quebec, QC, G1V 4G2, Canada.
- Faculté de pharmacie, Université Laval, Quebec, QC, Canada.
| |
Collapse
|
267
|
Declercq LD, Vandenberghe R, Van Laere K, Verbruggen A, Bormans G. Drug Development in Alzheimer's Disease: The Contribution of PET and SPECT. Front Pharmacol 2016; 7:88. [PMID: 27065872 PMCID: PMC4814730 DOI: 10.3389/fphar.2016.00088] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
Clinical trials aiming to develop disease-altering drugs for Alzheimer’s disease (AD), a neurodegenerative disorder with devastating consequences, are failing at an alarming rate. Poorly defined inclusion-and outcome criteria, due to a limited amount of objective biomarkers, is one of the major concerns. Non-invasive molecular imaging techniques, positron emission tomography and single photon emission (computed) tomography (PET and SPE(C)T), allow visualization and quantification of a wide variety of (patho)physiological processes and allow early (differential) diagnosis in many disorders. PET and SPECT have the ability to provide biomarkers that permit spatial assessment of pathophysiological molecular changes and therefore objectively evaluate and follow up therapeutic response, especially in the brain. A number of specific PET/SPECT biomarkers used in support of emerging clinical therapies in AD are discussed in this review.
Collapse
Affiliation(s)
- Lieven D Declercq
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven Leuven, Belgium
| | - Alfons Verbruggen
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| |
Collapse
|
268
|
High-affinity Anticalins with aggregation-blocking activity directed against the Alzheimer β-amyloid peptide. Biochem J 2016; 473:1563-78. [PMID: 27029347 PMCID: PMC4888463 DOI: 10.1042/bcj20160114] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 03/30/2016] [Indexed: 01/25/2023]
Abstract
Anticalins engineered for high affinity and specificity towards the central VFFAED epitope in Aβ peptides potently inhibit their aggregation, thus providing novel reagents to study the molecular pathology of Alzheimer's disease (AD) and alternative drug candidates compared with current biopharmaceutical treatments. Amyloid beta (Aβ) peptides, in particular Aβ42 and Aβ40, exert neurotoxic effects and their overproduction leads to amyloid deposits in the brain, thus constituting an important biomolecular target for treatments of Alzheimer's disease (AD). We describe the engineering of cognate Anticalins as a novel type of neutralizing protein reagent based on the human lipocalin scaffold. Phage display selection from a genetic random library comprising variants of the human lipocalin 2 (Lcn2) with mutations targeted at 20 exposed amino acid positions in the four loops that form the natural binding site was performed using both recombinant and synthetic target peptides and resulted in three different Anticalins. Biochemical characterization of the purified proteins produced by periplasmic secretion in Escherichia coli revealed high folding stability in a monomeric state, with Tm values ranging from 53.4°C to 74.5°C, as well as high affinities for Aβ40, between 95 pM and 563 pM, as measured by real-time surface plasmon resonance analysis. The central linear VFFAED epitope within the Aβ sequence was mapped using a synthetic peptide array on membranes and was shared by all three Anticalins, despite up to 13 mutual amino acid differences in their binding sites. All Anticalins had the ability–with varying extent–to inhibit Aβ aggregation in vitro according to the thioflavin-T fluorescence assay and, furthermore, they abolished Aβ42-mediated toxicity in neuronal cell culture. Thus, these Anticalins provide not only useful protein reagents to study the molecular pathology of AD but they also show potential as alternative drug candidates compared with antibodies.
Collapse
|
269
|
Belichenko PV, Madani R, Rey-Bellet L, Pihlgren M, Becker A, Plassard A, Vuillermot S, Giriens V, Nosheny RL, Kleschevnikov AM, Valletta JS, Bengtsson SKS, Linke GR, Maloney MT, Hickman DT, Reis P, Granet A, Mlaki D, Lopez-Deber MP, Do L, Singhal N, Masliah E, Pearn ML, Pfeifer A, Muhs A, Mobley WC. An Anti-β-Amyloid Vaccine for Treating Cognitive Deficits in a Mouse Model of Down Syndrome. PLoS One 2016; 11:e0152471. [PMID: 27023444 PMCID: PMC4811554 DOI: 10.1371/journal.pone.0152471] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/15/2016] [Indexed: 11/18/2022] Open
Abstract
In Down syndrome (DS) or trisomy of chromosome 21, the β-amyloid (Aβ) peptide product of the amyloid precursor protein (APP) is present in excess. Evidence points to increased APP gene dose and Aβ as playing a critical role in cognitive difficulties experienced by people with DS. Particularly, Aβ is linked to the late-life emergence of dementia as associated with neuropathological markers of Alzheimer's disease (AD). At present, no treatment targets Aβ-related pathogenesis in people with DS. Herein we used a vaccine containing the Aβ 1-15 peptide embedded into liposomes together with the adjuvant monophosphoryl lipid A (MPLA). Ts65Dn mice, a model of DS, were immunized with the anti-Aβ vaccine at 5 months of age and were examined for cognitive measures at 8 months of age. The status of basal forebrain cholinergic neurons and brain levels of APP and its proteolytic products were measured. Immunization of Ts65Dn mice resulted in robust anti-Aβ IgG titers, demonstrating the ability of the vaccine to break self-tolerance. The vaccine-induced antibodies reacted with Aβ without detectable binding to either APP or its C-terminal fragments. Vaccination of Ts65Dn mice resulted in a modest, but non-significant reduction in brain Aβ levels relative to vehicle-treated Ts65Dn mice, resulting in similar levels of Aβ as diploid (2N) mice. Importantly, vaccinated Ts65Dn mice showed resolution of memory deficits in the novel object recognition and contextual fear conditioning tests, as well as reduction of cholinergic neuron atrophy. No treatment adverse effects were observed; vaccine did not result in inflammation, cellular infiltration, or hemorrhage. These data are the first to show that an anti-Aβ immunotherapeutic approach may act to target Aβ-related pathology in a mouse model of DS.
Collapse
Affiliation(s)
- Pavel V. Belichenko
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | | | - Ann Becker
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | | | - Rachel L. Nosheny
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Alexander M. Kleschevnikov
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Janice S. Valletta
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sara K. S. Bengtsson
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Gordon R. Linke
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Michael T. Maloney
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, California, United States of America
| | | | | | | | | | | | - Long Do
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Nishant Singhal
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Matthew L. Pearn
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | - William C. Mobley
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
270
|
Carlson C, Siemers E, Hake A, Case M, Hayduk R, Suhy J, Oh J, Barakos J. Amyloid-related imaging abnormalities from trials of solanezumab for Alzheimer's disease. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2016; 2:75-85. [PMID: 27239538 PMCID: PMC4879647 DOI: 10.1016/j.dadm.2016.02.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction Solanezumab, a humanized monoclonal antibody that binds soluble amyloid beta peptide, is being developed for treatment of Alzheimer's disease (AD). Methods Patients (n = 2042) with mild and moderate AD were randomized 1:1 to 400-mg solanezumab or placebo infusion every 4 weeks for 80 weeks and 1457 patients entered an open-label extension. Magnetic resonance imaging scans monitored for amyloid-related imaging abnormalities-edema/effusion (ARIA-E) and amyloid-related imaging abnormalities-hemorrhage/hemosiderin deposition. Results Sixteen patients (solanezumab, n = 11; placebo, n = 5) developed ARIA-E during the double-blind phase, and 7 patients developed ARIA-E during the open-label extension as of July 31, 2014. Unique cases are discussed including solanezumab patients who were given solanezumab, while ARIA-E was present and a patient who developed ARIA-E during placebo treatment and again during solanezumab treatment. Discussion Asymptomatic ARIA-E was detected in solanezumab-treated and placebo-treated AD patients. ARIA-E occurs infrequently during solanezumab and placebo treatments but may occur repeatedly in some patients.
Collapse
Affiliation(s)
- Christopher Carlson
- Lilly Bio-medicines, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Eric Siemers
- Lilly Bio-medicines, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Ann Hake
- Lilly Bio-medicines, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Michael Case
- Lilly Bio-medicines, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Roza Hayduk
- Neurology, CNS Medical Strategy & Science, Therapeutic Science & Strategy Unit, Quintiles, Durham, NC, USA
| | - Joyce Suhy
- Medical Imaging, Neuroscience, Bioclinica, Newark, CA, USA
| | - Joonmi Oh
- Medical Imaging, Neuroscience, Bioclinica, Newark, CA, USA
| | - Jerome Barakos
- Medical Imaging, Neuroscience, Bioclinica, Newark, CA, USA; Department of Radiology, California Pacific Medical Center, San Francisco, CA, USA
| |
Collapse
|
271
|
Ravishankar HN, Dutta A, Lewis AG, Mohan A. Advances in therapies and scope for personalized medicine in Alzheimer's disease. Per Med 2016; 13:189-199. [PMID: 29749899 DOI: 10.2217/pme.15.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease is one of the leading causes of death worldwide and currently does not have any cure. The rate of incidence of Alzheimer's from 2010 is up by 71%, whereas many other diseases have been decreasing in their prevalence. In this review, we have attempted to cover the current landscape of treatment alongside forthcoming advances. We have also covered the present genes identified through genome-wide association studies, which could be used as novel biomarkers and could eventually reduce the cost of treatment through early diagnosis. As this disease is highly polymorphic, applications of personalized medicine have also found its way. All these upcoming developments offer a bright hope in the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Hosakere N Ravishankar
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Abhik Dutta
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Alisha G Lewis
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Amogh Mohan
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| |
Collapse
|
272
|
Schroeder SK, Joly-Amado A, Gordon MN, Morgan D. Tau-Directed Immunotherapy: A Promising Strategy for Treating Alzheimer's Disease and Other Tauopathies. J Neuroimmune Pharmacol 2016; 11:9-25. [PMID: 26538351 PMCID: PMC4746105 DOI: 10.1007/s11481-015-9637-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/16/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy directed against tau is a promising treatment strategy for Alzheimer's Disease (AD) and tauopathies. We review initial studies on tau-directed immunotherapy, and present data from our laboratory testing antibodies using the rTg4510 mouse model, which deposits tau in forebrain neurons. Numerous antibodies have been tested for their efficacy in treating both pathology and cognitive function, in different mouse models, by different routes of administration, and at different ages or durations. We report, here, that the conformation-specific antibody MC-1 produces some degree of improvement to both cognition and pathology in rTg4510. Pathological improvements as measured by Gallyas staining for fully formed tangles and phosphorylated tau appeared 4 days after intracranial injection into the hippocampus. We also examined markers for microglial activation, which did not appear impacted from treatment. Behavioral effects were noted after continuous infusion of antibodies into the lateral ventricle for approximately 2 weeks. We examined basic motor skills, which were not impacted by treatment, but did note cognitive improvements with both novel object and radial arm water maze testing. Our results support earlier reports in the initial review presented here, and collectively show promise for this strategy of treatment. The general absence of extracellular tau deposits may avoid the opsonization and phagocytosis mechanisms activated by antibodies against amyloid, and make anti tau approaches a safer method of immunotherapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Sulana K Schroeder
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Marcia N Gordon
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Dave Morgan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA.
| |
Collapse
|
273
|
Advances in recent patent and clinical trial drug development for Alzheimer's disease. Pharm Pat Anal 2016; 3:429-47. [PMID: 25291315 DOI: 10.4155/ppa.14.22] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, involving a large number of genes, proteins and their complex interactions. Currently, no effective therapeutic agents are available to either stop or reverse the progression of this disease, likely due to its polygenic nature. The complicated pathophysiology of AD remains unresolved. Although it has been hypothesized that the amyloid β cascade and the hyper-phosphorylated tau protein may be primarily involved, other mechanisms, such as oxidative stress, deficiency of central cholinergic neurotransmitter, mitochondrial dysfunction and inflammation have also been implicated. The main focus of this review is to document current therapeutic agents in clinical trials and patented candidate compounds under development based on their main mechanisms of action. It also discusses the relationship between the recent understanding of key targets and the development of potential therapeutic agents for the treatment of AD.
Collapse
|
274
|
Lin X, Bai G, Lin L, Wu H, Cai J, Ugen KE, Cao C. Vaccination induced changes in pro-inflammatory cytokine levels as an early putative biomarker for cognitive improvement in a transgenic mouse model for Alzheimer disease. Hum Vaccin Immunother 2016; 10:2024-31. [PMID: 25424812 DOI: 10.4161/hv.28735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several pieces of experimental evidence suggest that administration of anti-β amyloid (Aβ) vaccines, passive anti-Aβ antibodies or anti-inflammatory drugs can reduce Aβ deposition as well as associated cognitive/behavioral deficits in an Alzheimer disease (AD) transgenic (Tg) mouse model and, as such, may have some efficacy in human AD patients as well. In the investigation reported here an Aβ 1-42 peptide vaccine was administered to 16-month old APP+PS1 transgenic (Tg) mice in which Aβ deposition, cognitive memory deficits as well as levels of several pro-inflammatory cytokines were measured in response to the vaccination regimen. After vaccination, the anti-Aβ 1-42 antibody-producing mice demonstrated a significant reduction in the sera levels of 4 pro-inflammatory cytokines (TNF-α, IL-6, IL-1 α, and IL-12). Importantly, reductions in the cytokine levels of TNF-α and IL-6 were correlated with cognitive/behavioral improvement in the Tg mice. However, no differences in cerebral Aβ deposition in these mice were noted among the different control and experimental groups, i.e., Aβ 1-42 peptide vaccinated, control peptide vaccinated, or non-vaccinated mice. However, decreased levels of pro-inflammatory cytokines as well as improved cognitive performance were noted in mice vaccinated with the control peptide as well as those immunized with the Aβ 1-42 peptide. These findings suggest that reduction in pro-inflammatory cytokine levels in these mice may be utilized as an early biomarker for vaccination/treatment induced amelioration of cognitive deficits and are independent of Aβ deposition and, interestingly, antigen specific Aβ 1-42 vaccination. Since cytokine changes are typically related to T cell activation, the results imply that T cell regulation may have an important role in vaccination or other immunotherapeutic strategies in an AD mouse model and potentially in AD patients. Overall, these cytokine changes may serve as a predictive marker for AD development and progression as well as having potential therapeutic implications.
Collapse
Affiliation(s)
- Xiaoyang Lin
- a Department of Pharmacuetical Sciences College of Pharmacy; University of South Florida; Tampa, FL USA
| | | | | | | | | | | | | |
Collapse
|
275
|
Wang T, Xie XX, Ji M, Wang SW, Zha J, Zhou WW, Yu XL, Wei C, Ma S, Xi ZY, Pang GL, Liu RT. Naturally occurring autoantibodies against Aβ oligomers exhibited more beneficial effects in the treatment of mouse model of Alzheimer's disease than intravenous immunoglobulin. Neuropharmacology 2016; 105:561-576. [PMID: 26907803 DOI: 10.1016/j.neuropharm.2016.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 02/07/2016] [Accepted: 02/15/2016] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) is characterized by memory loss, intracellular neurofibrillary tangles, and extracellular plaque deposits composed of β-amyloid (Aβ). Previous reports showed that naturally occurring autoantibodies, such as intravenous immunoglobulin (IVIG), benefited patients with moderate-stage AD who carried an APOE-ε4 allele. However, the mechanism underlying the role of IVIG remains unclear. In this study, we identified naturally occurring autoantibodies against Aβ oligomers (NAbs-Aβo), which were purified by Aβ42 oligomer or Cibacron Blue affinity chromatography from IVIG and termed as Oli-NAbs and Blue-NAbs, respectively. Oli-NAbs and Blue-NAbs recognized Aβ42 oligomers or both Aβ40 and 42 oligomers, differently. Both antibodies inhibited Aβ42 aggregation and attenuated Aβ42-induced cytotoxicity. Compared with vehicles, Oli-NAbs, Blue-NAbs and IVIG significantly improved the memory and cognition, and reduced the soluble and oligomeric Aβ levels in APPswe/PS1dE9 transgenic mice. Further investigation showed that Blue-NAbs at increased doses effectively decreased plaque burden and insoluble Aβ levels, whereas Oli-NAbs significantly declined the microgliosis and astrogliosis, as well as the production of proinflammatory cytokines in vivo. Therefore, high levels of these antibodies against oligomeric Aβ40 or Aβ42 were required, correspondingly, to achieve the optimal effect. NAbs-Aβo could be condensed to a high concentration by affinity chromatography and its isolation from IVIG may not interfere with the normal function of conventional IVIG as its concentration is very low. Thus, the isolated NAbs-Aβo as an extra product of plasma required low cost and the enriched NAbs-Aβo may be more feasible than IVIG for the treatment of AD.
Collapse
Affiliation(s)
- Teng Wang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Life Science, AnHui Agricultural University, HeFei 230036, China
| | - Xi-Xiu Xie
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Mei Ji
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Shao-Wei Wang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Zha
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei-Wei Zhou
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiao-Lin Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Chen Wei
- National Institutes for Food and Drug Control, Beijing 100050, China
| | - Shan Ma
- Shandong Institute of Biological Products, Taian 2710000, China
| | - Zhi-Ying Xi
- Shandong Institute of Biological Products, Taian 2710000, China
| | - Guang-Li Pang
- Shandong Institute of Biological Products, Taian 2710000, China.
| | - Rui-Tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
276
|
Hayama T, Murakami K, Watanabe T, Maeda R, Kamata M, Kondo S. Single administration of a novel γ-secretase modulator ameliorates cognitive dysfunction in aged C57BL/6J mice. Brain Res 2016; 1633:52-61. [PMID: 26707406 DOI: 10.1016/j.brainres.2015.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
Mutations in presenilin 1 (PS1) and presenilin 2 (PS2) are known to cause early onset of Alzheimer's disease (AD). These proteins comprise the catalytic domain of γ-secretase, which catalyzes the cleavage of β-amyloid (Aβ) from amyloid precursor protein (APP). In recent reports, PS1 and PS2 were linked to the modulation of intracellular calcium ion (Ca(2+)) dynamics, a key regulator of synaptic function. Ca(2+) dysregulation and synaptic dysfunction are leading hypothesis of cognitive dysfunctions during aging and AD progression. Accordingly, manipulations of presenilins by small molecules may have therapeutic potential for the treatment of cognitive dysfunction. In an accompanying report, we showed that chronic treatment with compound-1, a novel γ-secretase modulator (GSM), reduced Aβ production and ameliorated cognitive dysfunction in Tg2576 APP transgenic mice. Accordingly, in the present study we showed that single oral administration of compound-1 at 1 and 3mg/kg ameliorated cognitive dysfunction in aged non-transgenic mice. Moreover, compound-1 enhanced synaptic plasticity in hippocampal slices from aged C57BL/6J mice and increased messenger RNA (mRNA) expression of the immediate early gene c-fos, which has been shown to be related to synaptic plasticity in vivo. Finally, compound-1 modulated Ca(2+) signals through PS1 in mouse embryonic fibroblast cells. Taken together, compound-1 ameliorates both Aβ pathology and age-related cognitive dysfunctions. Hence, compound-1 may have potential as an early intervention for the cognitive declines that are commonly diagnosed in aged subjects, such as mild cognitive impairment (MCI) and prodromal AD.
Collapse
Affiliation(s)
- Tatsuya Hayama
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Koji Murakami
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomomichi Watanabe
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Ryota Maeda
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Makoto Kamata
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shinichi Kondo
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
277
|
Ronsin S, Deiana G, Geraldo AF, Durand-Dubief F, Thomas-Maisonneuve L, Formaglio M, Desestret V, Meyronet D, Nighoghossian N, Berthezène Y, Honnorat J, Ducray F. Pseudotumoral presentation of cerebral amyloid angiopathy-related inflammation. Neurology 2016; 86:912-9. [PMID: 26850981 DOI: 10.1212/wnl.0000000000002444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/09/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To identify the clinical and radiologic features that should raise suspicion for the pseudotumoral presentation of cerebral amyloid angiopathy-related inflammation (CAA-I). METHODS We retrospectively reviewed the characteristics of 5 newly diagnosed and 23 previously reported patients in whom the CAA-I imaging findings were initially interpreted as CNS neoplasms. RESULTS Most cases (85%) occurred in patients >60 years old. The clinical characteristics at presentation included subacute cognitive decline (50%), confusion (32%), focal deficits (32%), seizures (25%), and headaches (21%). Brain MRI demonstrated infiltrative white matter lesions that exhibited a loco-regional mass effect without parenchymal enhancement (93%). In general, these findings were interpreted as low-grade glioma or lymphoma. Eighteen patients (64%) underwent a biopsy, which was nondiagnostic in 4 patients (14%), and 6 patients (21%) underwent a surgical resection. The primary reason for the misinterpretation of the imaging findings was the absence of T2*-weighted gradient recalled echo (T2*-GRE) sequences on initial imaging (89%). When subsequently performed (39%), the T2*-GRE sequences demonstrated multiple characteristic cortical and subcortical microhemorrhages in all cases. Perfusion MRI and magnetic resonance spectroscopy (MRS), which were performed on a subset of patients, indicated markedly reduced relative cerebral blood flow and a normal metabolic ratio. CONCLUSION The identification of one or several nonenhancing space-occupying lesions, especially in elderly patients presenting with cognitive impairment, should raise suspicion for the pseudotumoral presentation of CAA-I and lead to T2*-GRE sequences. Perfusion MRI and MRS appear to be useful techniques for the differential diagnosis of this entity.
Collapse
Affiliation(s)
- Solène Ronsin
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Gianluca Deiana
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Ana Filipa Geraldo
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Françoise Durand-Dubief
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Laure Thomas-Maisonneuve
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Maïté Formaglio
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Virginie Desestret
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - David Meyronet
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Norbert Nighoghossian
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Yves Berthezène
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| | - Jérôme Honnorat
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France.
| | - François Ducray
- From the Neuro-oncology Department (S.R., L.T.-M., J.H., F.D.), Neuro-radiology Department (G.D., A.F.G., Y.B.), Neurology Department A (F.D.-D.), Neurology Department D (M.F., V.D.), Neuropathology Department (D.M.), and Stroke Unit (N.N.), Hôpital Neurologique, Hospices Civils de Lyon; Université de Lyon-Université Claude Bernard Lyon 1 (S.R., G.D., A.F.G., F.D.-D., L.T.-M., M.F., V.D., D.M., N.N., Y.B., J.H., F.D.), France; Neurology Department and Stroke Unit (G.D.), Ospedale San Francesco, Nuoro, Italy; and Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292 (J.H., F.D.), France
| |
Collapse
|
278
|
Awasthi M, Singh S, Pandey VP, Dwivedi UN. Alzheimer's disease: An overview of amyloid beta dependent pathogenesis and its therapeutic implications along with in silico approaches emphasizing the role of natural products. J Neurol Sci 2016; 361:256-71. [DOI: 10.1016/j.jns.2016.01.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 01/02/2016] [Accepted: 01/04/2016] [Indexed: 01/09/2023]
|
279
|
Regulatory T cells delay disease progression in Alzheimer-like pathology. Brain 2016; 139:1237-51. [DOI: 10.1093/brain/awv408] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/23/2015] [Indexed: 01/07/2023] Open
|
280
|
|
281
|
Valera E, Spencer B, Masliah E. Immunotherapeutic Approaches Targeting Amyloid-β, α-Synuclein, and Tau for the Treatment of Neurodegenerative Disorders. Neurotherapeutics 2016; 13:179-89. [PMID: 26494242 PMCID: PMC4720672 DOI: 10.1007/s13311-015-0397-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disease-modifying alternatives are sorely needed for the treatment of neurodegenerative disorders, a group of diseases that afflict approximately 50 million Americans annually. Immunotherapy is one of the most developed approaches in this direction. Vaccination against amyloid-β, α-synuclein, or tau has been extensively explored, specially as the discovery that these proteins may propagate cell-to-cell and be accessible to antibodies when embedded into the plasma membrane or in the extracellular space. Likewise, the use of passive immunization approaches with specific antibodies against abnormal conformations of these proteins has also yielded promising results. The clinical development of immunotherapies for Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia, dementia with Lewy bodies, and other neurodegenerative disorders is a field in constant evolution. Results to date suggest that immunotherapy is a promising therapeutic approach for neurodegenerative diseases that progress with the accumulation and prion-like propagation of toxic protein aggregates. Here we provide an overview of the most novel and relevant immunotherapeutic advances targeting amyloid-β in Alzheimer’s disease, α-synuclein in Alzheimer’s disease and Parkinson’s disease, and tau in Alzheimer’s disease and frontotemporal dementia.
Collapse
Affiliation(s)
- Elvira Valera
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
| | - Brian Spencer
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
| | - Eliezer Masliah
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
- grid.266100.30000000121074242Department of Pathology, University of California, La Jolla, San Diego, CA 92093 USA
| |
Collapse
|
282
|
Yang KC, Chen HH. Probabilistic Cost-Effectiveness Analysis of Vaccination for Mild or Moderate Alzheimer's Disease. Curr Alzheimer Res 2016; 13:809-16. [PMID: 26825097 PMCID: PMC4923704 DOI: 10.2174/1567205013666160129095012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/26/2016] [Accepted: 04/01/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Studies on the immunotherapy for Alzheimer's disease (AD) have increasingly gained attention since 1990s. However, there are pros (preventing of AD) and cons (incurred cost and side effects) regarding the administration of immunotherapy. Up to date, there has been lacking of economic evaluation for immunotherapy of AD. We aimed to assess the cost-effectiveness analysis of the vaccination for AD. METHODS A meta-analysis of randomized control trials after systemic review was conducted to evaluate the efficacy of the vaccine. A Markov decision model was constructed and applied to a 120,000-Taiwanese cohort aged ≥65 years. Person years and quality-adjusted life years (QALY) were computed between the vaccinated group and the the unvaccinated group. Economic evaluation was performed to calculate the incremental cost-effectiveness ratio (ICER) and cost-effectiveness acceptability curve (CEAC). RESULTS Vaccinated group gained an additional 0.84 life years and 0.56 QALYs over 10-years and an additional 0.35 life years and 0.282 QALYs over 5-years of follow-up. The vaccinated group dominated the unvaccinated group by ICER over 5-years of follow-up. The ICERs of 10-year follow-up for the vaccinated group against the unvaccinated group were $13,850 per QALY and $9,038 per life year gained. Given the threshold of $20,000 of willingness to pay (WTP), the CEAC showed the probability of being cost-effective for vaccination with QALY was 70.7% and 92% for life years gained after 10-years of follow-up. The corresponding figures were 87.3% for QALY and 93.5% for life years gained over 5-years follow-up. CONCLUSION The vaccination for AD was cost-effective in gaining QALY and life years compared with no vaccination, under the condition of a reasonable threshold of WTP.
Collapse
Affiliation(s)
| | - Hsiu-Hsi Chen
- Division of Biostatistics, Graduate Institute of Epidemiology and Preventive, Medicine, College of Public Health, National Taiwan University, Room 533, No. 17 Hsuchow Road, Taipei 100, Taiwan.
| |
Collapse
|
283
|
Immunotherapy Against N-Truncated Amyloid-β Oligomers. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3560-4_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
284
|
|
285
|
Murakami K, Watanabe T, Koike T, Kamata M, Igari T, Kondo S. Pharmacological properties of a novel and potent γ-secretase modulator as a therapeutic option for the treatment of Alzheimer's disease. Brain Res 2015; 1633:73-86. [PMID: 26707977 DOI: 10.1016/j.brainres.2015.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 11/17/2022]
Abstract
Previous studies of γ-secretase inhibitors (GSIs) and Notch-sparing GSIs have shown reduced amyloid-β (Aβ) peptide levels but increased Notch-related and -unrelated adverse effects. In this study, we examined the effects of compound-1 on Aβ processing and cognitive function and assessed Notch-related and -unrelated adverse effects. Compound-1 reduced Aβ40 and Aβ42 levels but inversely increased Aβ37 in Neuro2a cells, leading to no net changes in total Aβ levels, indicating that compound-1 is a γ-secretase modulator (GSM). In time-course experiments, compound-1 reduced Aβ40 and Aβ42 levels in tris-soluble fractions, with peak reduction at approximately 3h after oral administration in C57BL mice. Moreover, at >1mg/kg, compound-1 dose dependently reduced Aβ40 and Aβ42 levels in Tg2576 mice. Chronic treatment with compound-1 in Tg2576 mice for 4 months significantly reduced both soluble and insoluble Aβ42 levels and ameliorated cognitive impairments, even after drug withdrawal for 10 days following oral administration for 2 months. In contrast with compound-1, at 100-fold higher doses (100mg/kg), the GSI LY450139 decreased HES1 mRNA expression in thymus tissues and increased the intensity of periodic acid-Schiff (PAS)-positive areas in the intestine. Moreover, the Notch-sparing GSI BMS708163 led to amyloid precursor protein (APP)-β-C-terminal fragment accumulation in mouse primary neurons. BMS708163 significantly hampered cognitive function in normal mice 1 month after administration, whereas compound-1 significantly improved cognitive function. Taken together, the present novel and orally active GSM is a promising molecule for the treatment of pathologies associated with Aβ42 and Aβ40.
Collapse
Affiliation(s)
- Koji Murakami
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomomichi Watanabe
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tatsuki Koike
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Makoto Kamata
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomoko Igari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Shinichi Kondo
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan.
| |
Collapse
|
286
|
Charef S, Leblanc A, Guibourg B, Quintin-Roue I, Ben Salem D, Zagnoli F. [A subacute dementia: Inflammatory cerebral amyloid angiopathy]. Rev Neurol (Paris) 2015; 171:832-40. [PMID: 26612553 DOI: 10.1016/j.neurol.2015.10.003] [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: 05/24/2015] [Revised: 07/30/2015] [Accepted: 10/05/2015] [Indexed: 11/18/2022]
Abstract
We report a case of inflammatory cerebral amyloid angiopathy (CAA) that led to rapid cognitive decline, seizures, visual hallucinations, hyperproteinorrachia and right hemispheric leukopathy. Brain biopsy gave the diagnosis of CAA. Although no inflammatory infiltrate was found in the biopsy sample, corticosteroids led to a regression of the radiological lesions without significant clinical improvement. CAA is a rare disease, defined by lesions of classical cerebral amyloid angiopathy and perivascular infiltrates in contact with the affected vessels. In cases of rapidly progressive dementia associated with leukopathy, inflammatory amyloid angiopathy should be considered as cognitive disorders may improve after immunosuppressive therapy.
Collapse
Affiliation(s)
- S Charef
- CHRU Cavale-Blanche, boulevard Tanguy-Prigent, 29200 Brest, France.
| | - A Leblanc
- CHRU Cavale-Blanche, boulevard Tanguy-Prigent, 29200 Brest, France
| | - B Guibourg
- CHRU Morvan, 2, avenue Floch, 29200 Brest, France
| | | | - D Ben Salem
- CHRU Cavale-Blanche, boulevard Tanguy-Prigent, 29200 Brest, France
| | - F Zagnoli
- CHRU Cavale-Blanche, boulevard Tanguy-Prigent, 29200 Brest, France
| |
Collapse
|
287
|
Novak G, Fox N, Clegg S, Nielsen C, Einstein S, Lu Y, Tudor IC, Gregg K, Di J, Collins P, Wyman BT, Yuen E, Grundman M, Brashear HR, Liu E. Changes in Brain Volume with Bapineuzumab in Mild to Moderate Alzheimer’s Disease. J Alzheimers Dis 2015; 49:1123-34. [DOI: 10.3233/jad-150448] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Gerald Novak
- Janssen Research and Development, Titusville, NJ, USA
| | - Nick Fox
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| | - Shona Clegg
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| | - Casper Nielsen
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| | | | - Yuan Lu
- Jazz Pharmaceuticals, Palo Alto, CA, USA
| | | | - Keith Gregg
- Janssen Alzheimer Immunotherapy, South San Francisco, CA, USA
| | - Jianing Di
- Janssen Research and Development, San Diego, CA, USA
| | | | | | - Eric Yuen
- Janssen Alzheimer Immunotherapy, South San Francisco, CA, USA
| | | | | | - Enchi Liu
- Janssen Research and Development, San Diego, CA, USA
| |
Collapse
|
288
|
Olson KE, Gendelman HE. Immunomodulation as a neuroprotective and therapeutic strategy for Parkinson's disease. Curr Opin Pharmacol 2015; 26:87-95. [PMID: 26571205 DOI: 10.1016/j.coph.2015.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 01/06/2023]
Abstract
While immune control is associated with nigrostriatal neuroprotection for Parkinson's disease, direct cause and effect relationships have not yet been realized, and modulating the immune system for therapeutic gain has been openly debated. Here, we review how innate and adaptive immunity affect disease pathobiology, and how each could be harnessed for treatment. The overarching idea is to employ immunopharmacologics as neuroprotective strategies for disease. The aim of the current work is to review disease-modifying treatments that are currently being developed as neuroprotective strategies for PD in experimental animal models and for human disease translation. The long-term goal of this research is to effectively harness the immune system to slow or prevent PD pathobiology.
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
289
|
Abstract
Use of highly pure antigens to improve vaccine safety has led to reduced vaccine immunogenicity and efficacy. This has led to the need to use adjuvants to improve vaccine immunogenicity. The ideal adjuvant should maximize vaccine immunogenicity without compromising tolerability or safety. Unfortunately, adjuvant research has lagged behind other vaccine areas such as antigen discovery, with the consequence that only a very limited number of adjuvants based on aluminium salts, monophosphoryl lipid A and oil emulsions are currently approved for human use. Recent strategic initiatives to support adjuvant development by the National Institutes of Health should translate into greater adjuvant choices in the future. Mechanistic studies have been valuable for better understanding of adjuvant action, but mechanisms of adjuvant toxicity are less well understood. The inflammatory or danger-signal model of adjuvant action implies that increased vaccine reactogenicity is the inevitable price for improved immunogenicity. Hence, adjuvant reactogenicity may be avoidable only if it is possible to separate inflammation from adjuvant action. The biggest remaining challenge in the adjuvant field is to decipher the potential relationship between adjuvants and rare vaccine adverse reactions, such as narcolepsy, macrophagic myofasciitis or Alzheimer's disease. While existing adjuvants based on aluminium salts have a strong safety record, there are ongoing needs for new adjuvants and more intensive research into adjuvants and their effects.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Department of Endocrinology and Diabetes, Flinders University, Adelaide, SA, 5042, Australia.
- Vaxine Pty Ltd, Adelaide, SA, Australia.
| |
Collapse
|
290
|
Fuller JP, Stavenhagen JB, Christensen S, Kartberg F, Glennie MJ, Teeling JL. Comparing the efficacy and neuroinflammatory potential of three anti-abeta antibodies. Acta Neuropathol 2015; 130:699-711. [PMID: 26433971 PMCID: PMC4612324 DOI: 10.1007/s00401-015-1484-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 11/21/2022]
Abstract
Immunotherapy is a promising strategy for the treatment of Alzheimer's disease (AD). Antibodies directed against Amyloid Beta (Aβ) are able to successfully clear plaques and reverse cognitive deficits in mouse models. Excitement towards this approach has been tempered by high profile failures in the clinic, one key issue has been the development of inflammatory side effects in the brain (ARIAs). New antibodies are entering the clinic for Alzheimer's disease; therefore, it is important to learn all we can from the current generation. In this study, we directly compared 3 clinical candidates in the same pre-clinical model, with the same effector function, for their ability to clear plaques and induce inflammation in the brain. We produced murine versions of the antibodies: Bapineuzumab (3D6), Crenezumab (mC2) and Gantenerumab (chGantenerumab) with an IgG2a constant region. 18-month transgenic APP mice (Tg2576) were injected bilaterally into the hippocampus with 2 µg of each antibody or control. After 7 days, the mice tissue was analysed for clearance of plaques and neuroinflammation by histology and biochemical analysis. 3D6 was the best binder to plaques and in vitro, whilst mC2 bound the least strongly. This translated into 3D6 effectively clearing plaques and reducing the levels of insoluble Aβ, whilst chGantenerumab and mC2 did not. 3D6 caused a significant increase in the levels of pro-inflammatory cytokines IL-1β and TNFα, and an associated increase in microglial expression of CD11B and CD68. chGantenerumab increased pro-inflammatory cytokines and microglial activation, but minimal changes in CD68, as an indicator of phagocytosis. Injection of mC2 did not cause any significant inflammatory changes. Our results demonstrate that the ability of an antibody to clear plaques and induce inflammation is dependent on the epitope and affinity of the antibody.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Amyloid beta-Peptides/genetics
- Amyloid beta-Peptides/immunology
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- CD11b Antigen/metabolism
- Cell Line
- Drug Evaluation, Preclinical
- Female
- Hippocampus/drug effects
- Hippocampus/immunology
- Hippocampus/pathology
- Humans
- Immunologic Factors/pharmacology
- Interleukin-1beta/metabolism
- Mice, Transgenic
- Microglia/drug effects
- Microglia/immunology
- Microglia/pathology
- Neuroimmunomodulation/drug effects
- Neuroimmunomodulation/physiology
- Plaque, Amyloid/drug therapy
- Plaque, Amyloid/immunology
- Plaque, Amyloid/pathology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- James P Fuller
- Centre for Biological Sciences, University of Southampton, Southampton, UK.
| | | | | | | | | | - Jessica L Teeling
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
291
|
Bouter Y, Lopez Noguerola JS, Tucholla P, Crespi GAN, Parker MW, Wiltfang J, Miles LA, Bayer TA. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against N‑truncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol 2015; 130:713-29. [PMID: 26467270 DOI: 10.1007/s00401-015-1489-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/05/2015] [Accepted: 10/05/2015] [Indexed: 01/01/2023]
Abstract
Solanezumab and Crenezumab are two humanized antibodies targeting Amyloid-β (Aβ) which are currently tested in multiple clinical trials for the prevention of Alzheimer's disease. However, there is a scientific discussion ongoing about the target engagement of these antibodies. Here, we report the immunohistochemical staining profiles of biosimilar antibodies of Solanezumab, Crenezumab and Bapineuzumab in human formalin-fixed, paraffin-embedded tissue and human fresh frozen tissue. Furthermore, we performed a direct comparative immunohistochemistry analysis of the biosimilar versions of the humanized antibodies in different mouse models including 5XFAD, Tg4-42, TBA42, APP/PS1KI, 3xTg. The staining pattern with these humanized antibodies revealed a surprisingly similar profile. All three antibodies detected plaques, cerebral amyloid angiopathy and intraneuronal Aβ in a similar fashion. Remarkably, Solanezumab showed a strong binding affinity to plaques. We also reaffirmed that Bapineuzumab does not recognize N-truncated or modified Aβ, while Solanezumab and Crenezumab do detect N-terminally modified Aβ peptides Aβ4-42 and pyroglutamate Aβ3-42. In addition, we compared the results with the staining pattern of the mouse NT4X antibody that recognizes specifically Aβ4-42 and pyroglutamate Aβ3-42, but not full-length Aβ1-42. In contrast to the biosimilar antibodies of Solanezumab, Crenezumab and Bapineuzumab, the murine NT4X antibody shows a unique target engagement. NT4X does barely cross-react with amyloid plaques in human tissue. It does, however, detect cerebral amyloid angiopathy in human tissue. In Alzheimer mouse models, NT4X detects intraneuronal Aβ and plaques comparable to the humanized antibodies. In conclusion, the biosimilar antibodies Solanezumab, Crenezumab and Bapineuzumab strongly react with amyloid plaques, which are in contrast to the NT4X antibody that hardly recognizes plaques in human tissue. Therefore, NT4X is the first of a new class of therapeutic antibodies.
Collapse
|
292
|
Abstract
Since the launch of our journal as Nature Clinical Practice Neurology in 2005, we have seen remarkable progress in many areas of neurology research, but what does the future hold? Will advances in basic research be translated into effective disease-modifying therapies, and will personalized medicine finally become a reality? For this special Viewpoint article, we invited a panel of Advisory Board members and other journal contributors to outline their research priorities and predictions in neurology for the next 10 years.
Collapse
|
293
|
The Peptide Vaccine Combined with Prior Immunization of a Conventional Diphtheria-Tetanus Toxoid Vaccine Induced Amyloid β Binding Antibodies on Cynomolgus Monkeys and Guinea Pigs. J Immunol Res 2015; 2015:786501. [PMID: 26539559 PMCID: PMC4619934 DOI: 10.1155/2015/786501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 07/21/2015] [Indexed: 12/21/2022] Open
Abstract
The reduction of brain amyloid beta (Aβ) peptides by anti-Aβ antibodies is one of the possible therapies for Alzheimer's disease. We previously reported that the Aβ peptide vaccine including the T-cell epitope of diphtheria-tetanus combined toxoid (DT) induced anti-Aβ antibodies, and the prior immunization with conventional DT vaccine enhanced the immunogenicity of the peptide. Cynomolgus monkeys were given the peptide vaccine subcutaneously in combination with the prior DT vaccination. Vaccination with a similar regimen was also performed on guinea pigs. The peptide vaccine induced anti-Aβ antibodies in cynomolgus monkeys and guinea pigs without chemical adjuvants, and excessive immune responses were not observed. Those antibodies could preferentially recognize Aβ40, and Aβ42 compared to Aβ fibrils. The levels of serum anti-Aβ antibodies and plasma Aβ peptides increased in both animals and decreased the brain Aβ40 level of guinea pigs. The peptide vaccine could induce a similar binding profile of anti-Aβ antibodies in cynomolgus monkeys and guinea pigs. The peptide vaccination could be expected to reduce the brain Aβ peptides and their toxic effects via clearance of Aβ peptides by generated antibodies.
Collapse
|
294
|
DiFrancesco JC, Longoni M, Piazza F. Anti-Aβ Autoantibodies in Amyloid Related Imaging Abnormalities (ARIA): Candidate Biomarker for Immunotherapy in Alzheimer's Disease and Cerebral Amyloid Angiopathy. Front Neurol 2015; 6:207. [PMID: 26441825 PMCID: PMC4585101 DOI: 10.3389/fneur.2015.00207] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/08/2015] [Indexed: 11/13/2022] Open
Abstract
Amyloid-related imaging abnormalities (ARIA) represent the major severe side effect of amyloid-beta (Aβ) immunotherapy for Alzheimer’s disease (AD). Early biomarkers of ARIA represent an important challenge to ensure safe and beneficial effects of immunotherapies, given that different promising clinical trials in prodromal and subjects at risk for AD are underway. The recent demonstration that cerebrospinal fluid (CSF) anti-Aβ autoantibodies play a key role in the development of the ARIA-like events characterizing cerebral amyloid angiopathy-related inflammation generated great interest in the field of immunotherapy. Herein, we critically review the growing body of evidence supporting the monitoring of CSF anti-Aβ autoantibody as a promising candidate biomarker for ARIA in clinical trials.
Collapse
Affiliation(s)
- Jacopo C DiFrancesco
- School of Medicine, Milan Center for Neuroscience (NeuroMi), University of Milano-Bicocca , Monza , Italy ; The Inflammatory Cerebral Amyloid Angiopathy and Alzheimer's Disease βiomarkers (iCAβ) International Network , Monza , Italy
| | - Martina Longoni
- School of Medicine, Milan Center for Neuroscience (NeuroMi), University of Milano-Bicocca , Monza , Italy ; The Inflammatory Cerebral Amyloid Angiopathy and Alzheimer's Disease βiomarkers (iCAβ) International Network , Monza , Italy
| | - Fabrizio Piazza
- School of Medicine, Milan Center for Neuroscience (NeuroMi), University of Milano-Bicocca , Monza , Italy ; The Inflammatory Cerebral Amyloid Angiopathy and Alzheimer's Disease βiomarkers (iCAβ) International Network , Monza , Italy ; The iCAβ-ITALY Study Group of the Italian Society for the Study of Dementia (SINdem) , Monza , Italy
| |
Collapse
|
295
|
Sábado J, Casanovas A, Rodrigo H, Arqué G, Esquerda JE. Adverse effects of a SOD1-peptide immunotherapy on SOD1 G93A mouse slow model of amyotrophic lateral sclerosis. Neuroscience 2015; 310:38-50. [PMID: 26384962 DOI: 10.1016/j.neuroscience.2015.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022]
Abstract
Previous reports from our lab had shown that some anti-purinergic receptor P2X4 antibodies cross-reacted with misfolded forms of mutant Cu/Zn superoxide dismutase 1 (SOD1), linked to amyotrophic lateral sclerosis (ALS). Cross-reactivity could be caused by the abnormal exposure of an epitope located in the inner hydrophobic region of SOD1 that shared structural homology with the P2X4-immunizing peptide. We had previously raised antibodies against human SOD1 epitope mimicked by the P2X4 immunizing peptide. One of these antibodies, called AJ10, was able to recognize mutant/misfolded forms of ALS-linked mutant SOD1. Here, we used the AJ10 antigen as a vaccine to target neurotoxic species of mutant SOD1 in a slow mouse model of ALS. However, the obtained results showed no improvement in life span, disease onset or weight loss in treated animals; we observed an increased microglial neuroinflammatory response and high amounts of misfolded SOD1 accumulated within spinal cord neurons after AJ10 immunization. An increase of immunoglobulin G deposits was also found due to the treatment. Finally, a significantly worse clinical evolution was displayed by an impairment on motor function as a consequence of AJ10 peptide immunization.
Collapse
Affiliation(s)
- J Sábado
- Universitat de Lleida, Facultat de Medicina-IRBLLEIDA, Departament de Medicina Experimental, Unitat de Neurobiologia Cel·lular, Avda Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - A Casanovas
- Universitat de Lleida, Facultat de Medicina-IRBLLEIDA, Departament de Medicina Experimental, Unitat de Neurobiologia Cel·lular, Avda Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - H Rodrigo
- Universitat de Lleida, Facultat de Medicina-IRBLLEIDA, Departament de Medicina Experimental, Unitat de Neurobiologia Cel·lular, Avda Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - G Arqué
- Universitat de Lleida, Facultat de Medicina-IRBLLEIDA, Departament de Medicina Experimental, Unitat de Neurobiologia Cel·lular, Avda Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - J E Esquerda
- Universitat de Lleida, Facultat de Medicina-IRBLLEIDA, Departament de Medicina Experimental, Unitat de Neurobiologia Cel·lular, Avda Rovira Roure 80, 25198 Lleida, Catalonia, Spain.
| |
Collapse
|
296
|
Fernandez-Funez P, Zhang Y, Sanchez-Garcia J, de Mena L, Khare S, Golde TE, Levites Y, Rincon-Limas DE. Anti-Aβ single-chain variable fragment antibodies exert synergistic neuroprotective activities in Drosophila models of Alzheimer's disease. Hum Mol Genet 2015; 24:6093-105. [PMID: 26253732 DOI: 10.1093/hmg/ddv321] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/31/2015] [Indexed: 12/16/2022] Open
Abstract
Both active and passive immunotherapy protocols decrease insoluble amyloid-ß42 (Aß42) peptide in animal models, suggesting potential therapeutic applications against the main pathological trigger in Alzheimer's disease (AD). However, recent clinical trials have reported no significant benefits from humanized anti-Aß42 antibodies. Engineered single-chain variable fragment antibodies (scFv) are much smaller and can easily penetrate the brain, but identifying the most effective scFvs in murine AD models is slow and costly. We show here that scFvs against the N- and C-terminus of Aß42 (scFv9 and scFV42.2, respectively) that decrease insoluble Aß42 in CRND mice are neuroprotective in Drosophila models of Aß42 and amyloid precursor protein neurotoxicity. Both scFv9 and scFv42.2 suppress eye toxicity, reduce cell death in brain neurons, protect the structural integrity of dendritic terminals in brain neurons and delay locomotor dysfunction. Additionally, we show for the first time that co-expression of both anti-Aß scFvs display synergistic neuroprotective activities, suggesting that combined therapies targeting distinct Aß42 epitopes can be more effective than targeting a single epitope. Overall, we demonstrate the feasibility of using Drosophila as a first step for characterizing neuroprotective anti-Aß scFvs in vivo and identifying scFv combinations with synergistic neuroprotective activities.
Collapse
Affiliation(s)
- Pedro Fernandez-Funez
- Department of Neurology, McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease and Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Yan Zhang
- Department of Neurology, McKnight Brain Institute
| | | | | | - Swati Khare
- Department of Neurology, McKnight Brain Institute
| | - Todd E Golde
- Department of Neurology, McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease and
| | - Yona Levites
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease and
| | - Diego E Rincon-Limas
- Department of Neurology, McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease and Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
297
|
Li YP, Yang GJ, Jin L, Yang HM, Chen J, Chai GS, Wang L. Erythropoietin attenuates Alzheimer-like memory impairments and pathological changes induced by amyloid β42 in mice. Brain Res 2015; 1618:159-67. [DOI: 10.1016/j.brainres.2015.05.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/23/2015] [Accepted: 05/25/2015] [Indexed: 01/05/2023]
|
298
|
Mishra P, Ayyannan SR, Panda G. Perspectives on Inhibiting β-Amyloid Aggregation through Structure-Based Drug Design. ChemMedChem 2015; 10:1467-74. [DOI: 10.1002/cmdc.201500215] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/30/2015] [Indexed: 12/24/2022]
|
299
|
Agadjanyan MG, Petrovsky N, Ghochikyan A. A fresh perspective from immunologists and vaccine researchers: active vaccination strategies to prevent and reverse Alzheimer's disease. Alzheimers Dement 2015; 11:1246-59. [PMID: 26192465 DOI: 10.1016/j.jalz.2015.06.1884] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/30/2022]
Abstract
Traditional vaccination against infectious diseases relies on generation of cellular and humoral immune responses that act to protect the host from overt disease even though they do not induce sterilizing immunity. More recently, attempts have been made with mixed success to generate therapeutic vaccines against a wide range of noninfectious diseases including neurodegenerative disorders. After the exciting first report of successful vaccine prevention of progression of an Alzheimer's disease (AD) animal model in 1999, various epitope-based vaccines targeting amyloid beta (Aβ) have proceeded to human clinical trials, with varied results. More recently, AD vaccines based on tau protein have advanced into clinical testing too. This review seeks to put perspective to the mixed results obtained so far in clinical trials of AD vaccines and discusses the many pitfalls and misconceptions encountered on the path to a successful AD vaccine, including better standardization of immunologic efficacy measures of antibodies, immunogenicity of platform/carrier and adjuvants.
Collapse
Affiliation(s)
- Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA; The Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders Medical Centre, Adelaide, South Australia; Flinders Medical Centre and Flinders University, Adelaide, South Australia
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
300
|
Karussis D, Petrou P. Acute Disseminated Encephalomyelitis: Idiopathic, Post‐infectious, and Post‐vaccination. VACCINES AND AUTOIMMUNITY 2015:311-330. [DOI: 10.1002/9781118663721.ch33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|