Acessibilidade / Reportar erro

Alzheimer's disease: is a vaccine possible?

Abstract

The cause of Alzheimer's disease is still unknown, but the disease is distinctively characterized by the accumulation of β-amyloid plaques and neurofibrillary tangles in the brain. These features have become the primary focus of much of the research looking for new treatments for the disease, including immunotherapy and vaccines targeting β-amyloid in the brain. Adverse effects observed in a clinical trial based on the β-amyloid protein were attributed to the presence of the target antigen and emphasized the relevance of finding safer antigen candidates for active immunization. For this kind of approach, different vaccine formulations using DNA, peptide, and heterologous prime-boost immunization regimens have been proposed. Promising results are expected from different vaccine candidates encompassing B-cell epitopes of the β-amyloid protein. In addition, recent results indicate that targeting another protein involved in the etiology of the disease has opened new perspectives for the effective prevention of the illness. Collectively, the evidence indicates that the idea of finding an effective vaccine for the control of Alzheimer's disease, although not without challenges, is a possibility.

Alzheimer's disease; β-amyloid; Vaccine; Active immunization


Introduction

Alzheimer's disease (AD), the most common form of dementia in elderly people, is characterized by a progressive decline of brain functions, including memory, language, spatial orientation, and behavior, finally resulting in death. This disease was first described by the German physician Alois Alzheimer in 1906 (published in 1907), based on his studies of a 51-year-old female patient who presented symptoms of dementia, beginning with changes in personality and progressive memory loss, and a life prognosis of 4 to 5 years after the initial symptoms. After necropsy, the physician described cerebral atrophy, deposits of fibrous structures in neurons in the cortical area of the brain, and extracellular plaque-like lesions (11. Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer's 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde”. Clin Anat 1995; 8: 429-431, doi: 10.1002/ca.980080612.
https://doi.org/10.1002/ca.980080612...
). The features he described are currently recognized as typical of AD, whose pathology is characterized by gliosis and tissue atrophy mainly caused by the loss of synapses, especially pronounced in the cortex and hippocampus regions of the brain (22. Braak H, Braak E. Staging of Alzheimer's disease-related neurofibrillary changes. Neurobiol Aging 1995; 16: 271-278, doi: 10.1016/0197-4580(95)00021-6.
https://doi.org/10.1016/0197-4580(95)000...
).

AD is the sixth leading cause of death in the United States and the fifth leading cause of death among the elderly population worldwide (33. Minião AM, Xu J, Kochanek KD. Deaths: preliminary data for 2008. NVSR (National Vital Statistics Reports) Publication PHS 2011-1120. Washington: Centers for Disease Control and Prevention, National Center for Health Statistics. U.S. Government Printing Office; 2010.). Compared to the total world population, the percentage of people suffering from AD is relatively low. A census carried out in the United States in 2000 estimated that there were 4.5 million patients in the country (44. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol 2003; 60: 1119-1122, doi: 10.1001/archneur.60.8.1119.
https://doi.org/10.1001/archneur.60.8.11...
), and in 2010, there were approximately 35.9 million AD patients in the world (55. Alzheimer's Disease International. World Alzheimer Report 2010. London: Alzheimer's Disease International; 2010.). However, both estimates anticipated that these numbers would increase by more than 300% by 2050. It is interesting to note that the mortality rate of AD tends to increase over the years, unlike other major causes of death, such as heart disease and cancer. The explanation for this phenomenon could be the trend toward aging of the human population, and the association of AD with this specific age group. The increases in both numbers are directly proportional (66. United Nations PD. World population ageing: 1950-2050. New York: Department of Economic and Social Affairs; 2002.). In 2010, it was estimated that the total cost of dementia worldwide was more than 600 billion US dollars (55. Alzheimer's Disease International. World Alzheimer Report 2010. London: Alzheimer's Disease International; 2010.). With the tendency for the size of the elderly population to increase, it is expected that costs related to AD, the most common form of dementia, will also increase. Today, owing to the small numbers of patients, little is known about AD in the general population regarding its importance to both society and the economy. However, based on existing data, it is possible that in less than 50 years AD will become a serious public health problem, with a significant socioeconomic impact.

Molecular aspects of Alzheimer's disease

AD presents two well-characterized pathological markers, β-amyloid (Aβ) plaques and neurofibrillary tangles (Figure 1). The Aβ plaques, or extracellular senile plaques, are formed by a 42-amino acid peptide known as Aβ peptide (77. Glenner GG, Wong CW. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984; 120: 885-890, doi: 10.1016/S0006-291X(84)80190-4.
https://doi.org/10.1016/S0006-291X(84)80...
,88. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 1985; 82: 4245-4249, doi: 10.1073/pnas.82.12.4245.
https://doi.org/10.1073/pnas.82.12.4245...
) that is produced after cleavage of a precursor protein known as amyloid precursor protein. The released peptides form the amyloid plaques, which are insoluble, cytotoxic aggregates. These plaques are neurotoxic and result in apoptosis of neurons, local inflammation, disruption of calcium homeostasis, oxidative stress and complement activation, which are responsible for the clinical manifestations of the disease. The neurofibrillary tangles are formed by the hyperphosphorylation of the tau protein, which plays an essential role in inducing Aβ toxicity as well as mitochondrial dysfunction in AD (99. Kurz A, Perneczky R. Amyloid clearance as a treatment target against Alzheimer's disease. J Alzheimers Dis 2011; 24 (Suppl 2): 61-73.

10. Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A. Tau is essential to beta-amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A 2002; 99: 6364-6369, doi: 10.1073/pnas.092136199.
https://doi.org/10.1073/pnas.092136199...
-1111. Pritchard SM, Dolan PJ, Vitkus A, Johnson GV. The toxicity of tau in Alzheimer disease: turnover, targets and potential therapeutics. J Cell Mol Med 2011; 15: 1621-1635, doi: 10.1111/j.1582-4934.2011.01273.x.
https://doi.org/10.1111/j.1582-4934.2011...
).

Figure 1
Alzheimer's disease (AD) in its molecular aspects. AD presents two characterized pathological markers: β-amyloid (Aβ) plaques and neurofibrillary tangles. The β-cleavage of the amyloid precursor protein (APP) results in a soluble form of this protein (sAPPβ). The β-cleavage followed by a γ-cleavage of the precursor protein results in an insoluble form of Aβ peptides, which aggregate and form plaques, causing an inflammatory response that leads to neuronal death and symptoms of dementia. The neurofibrillary tangles are formed by hyperphosphorylation of the Tau protein, which plays a role in inducing Aβ toxicity as well as mitochondrial dysfunction in AD.

Currently available treatments

Currently, there are few treatments available for AD, and they can be classified as pharmacological, psychological and immunological approaches. In the case of pharmacological treatments, there are the acetylcholinesterase inhibitors, which aim to increase the concentration of acetylcholine in the brain, covering the decrease of this neurotransmitter caused by the death of neurons (1212. Giacobini E. Modulation of brain acetylcholine levels with cholinesterase inhibitors as a treatment of Alzheimer disease. Keio J Med 1987; 36: 381-391, doi: 10.2302/kjm.36.381.
https://doi.org/10.2302/kjm.36.381...
). Another pharmacological approach is the use of glutamate receptor inhibitors such as N-methyl-d-aspartate receptors, whose overstimulation leads to cytotoxicity (1313. Greenamyre JT, Young AB. Excitatory amino acids and Alzheimer's disease. Neurobiol Aging 1989; 10: 593-602, doi: 10.1016/0197-4580(89)90143-7.
https://doi.org/10.1016/0197-4580(89)901...
). The psychological treatments involve cognitive stimulation and physical exercises, such as cognitive rehabilitation, which help to deal with the limitations caused by the disease and aim to improve the patient's quality of life (1414. Dröes RM, Van Mierlo LD, Van der Roest HG, Meiland FJM. Focus and effectiveness of psychosocial interventions for people with dementia in institutional care settings from the perspective of coping with the disease. Non Pharm Ther Dement 2010; 1: 139-161.). Passive immunization with anti-Aβ monoclonal antibodies, e.g., bapineuzumab and solanezumab, has been considered an alternative AD treatment, and various monoclonal antibodies are currently being evaluated in clinical trials (1515. Moreth J, Mavoungou C, Schindowski K. Passive anti-amyloid immunotherapy in Alzheimer's disease: What are the most promising targets? Immun Ageing 2013; 10: 18, doi: 10.1186/1742-4933-10-18.
https://doi.org/10.1186/1742-4933-10-18...
).

Despite the availability of different treatment options, each therapeutic approach has specific limitations. The pharmacological treatments are expensive, require permanent use, and serve only to control the symptoms, not aiming to cure or reduce the progression of the disease. The psychosocial interventions improve quality of life but are not effective against the prognosis of the disease. Presently, the immunological treatments with anti-Aβ monoclonal antibodies seem to be the most promising option. However, once approved for human use, their high costs and lifelong use will pose severe limitations on the widespread use of these compounds. In addition to these disadvantages, monoclonal antibodies do not always offer the expected results. Recently, clinical trials of bapineuzumab were halted in phase 3 owing to failure to demonstrate a significant improvement in cognitive and functional activities (1616. Khorassani F, Hilas O. Bapineuzumab, an Investigational Agent For Alzheimer's Disease. P T 2013; 38: 89-91, PMC3628177.).

New treatments against AD are expected to be better and cheaper than the current options by showing long-term therapeutic effects with no or reduced adverse effects. On the other hand, the possibility of preventing the disease using a vaccine approach would be preferable to the treatment of affected subjects. Accordingly, the following question can be asked: is it possible to develop a vaccine against AD that would meet these requirements?

Vaccines against Alzheimer's disease: what has been done so far?

Identifying a target for the control of the disease

Although the exact cause of AD is not yet known, there are common features observed among affected patients, the presence of Aβ plaques being one of them and arguably the most common target for immunotherapeutic approaches. Experiments carried out by Schenk and colleagues in 1999 were the first to demonstrate that immunization with the Aβ peptide could reduce the deposit of plaques in the brain of mice genetically modified to develop AD with symptoms similar to those observed in humans (1717. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999; 400: 173-177, doi: 10.1038/22124.
https://doi.org/10.1038/22124...
).

Noting that the generation of Aβ-specific antibodies can reduce AD symptoms, three different hypotheses were formulated to explain the effects of removal of excess Aβ from the brain (Figure 2). Firstly, the antibodies would bind directly to the peptides in the senile plaque, destabilizing the interactions of the Aβ molecule and disrupting them (1818. Solomon B, Koppel R, Frankel D, Hanan-Aharon E. Disaggregation of Alzheimer beta-amyloid by site-directed mAb. Proc Natl Acad Sci U S A 1997; 94: 4109-4112, doi: 10.1073/pnas.94.8.4109.
https://doi.org/10.1073/pnas.94.8.4109...
). Secondly, the Aβ-specific antibodies would bind to the plaque and promote their phagocytosis by microglial cells mediated by Fc receptors (1919. Wilcock DM, Munireddy SK, Rosenthal A, Ugen KE, Gordon MN, Morgan D. Microglial activation facilitates Abeta plaque removal following intracranial anti-Abeta antibody administration. Neurobiol Dis 2004; 15: 11-20, doi: 10.1016/j.nbd.2003.09.015.
https://doi.org/10.1016/j.nbd.2003.09.01...
). Finally, the antibodies would not cross the blood-brain barrier but would bind to the circulating Aβ molecules present in the plasma of the affected subject, thereby leading to a concentration gradient that ultimately would result in the efflux of Aβ from the brain into the blood and plasma, a mechanism known as the peripheral sink model (2020. DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2001; 98: 8850-8855, doi: 10.1073/pnas.151261398.
https://doi.org/10.1073/pnas.151261398...
). Based on these facts, much of the research related to vaccines against AD has focused on the reduction of senile plaques in the brain by generating antibodies specific to the Aβ peptide through active immunization (2121. Hock C, Konietzko U, Papassotiropoulos A, Wollmer A, Streffer J, von Rotz RC, et al. Generation of antibodies specific for beta-amyloid by vaccination of patients with Alzheimer disease. Nat Med 2002; 8: 1270-1275, doi: 10.1038/nm783.
https://doi.org/10.1038/nm783...
,2222. Weiner HL, Frenkel D. Immunology and immunotherapy of Alzheimer's disease. Nat Rev Immunol 2006; 6: 404-416, doi: 10.1038/nri1843.
https://doi.org/10.1038/nri1843...
).

Figure 2
Mechanism of β-amyloid (Aβ) removal via Aβ-specific antibodies. There are three hypotheses for the mechanism of action of anti-Aβ. The first involves the direct action of the antibody against the Aβ plaques, where the binding of the antibody destabilizes the plaques. The second involves the action of microglia, which leads to the phagocytosis of Aβ mediated by Fc-receptors (FcR). Lastly, there is the peripheral sink mechanism hypothesis, in which the antibody binds to and removes Aβ present in the plasma, generating a net efflux of Aβ from the brain to the plasma.

Vaccine approaches based on Aβ

Several vaccine approaches have been proposed with the Aβ peptide as the target antigen, employing different murine models to evaluate specific humoral responses and provide a prognosis of disease progression (Table 1). Formulations based on DNA vaccines have in common the idea of employing in-tandem fusions with immunomodulatory sequences, such as the PADRE sequence (pan human leukocyte antigen DR-binding peptide), a promiscuous nonself T-cell epitope that has been used by itself or in association with another immune modulator (2323. Movsesyan N, Ghochikyan A, Mkrtichyan M, Petrushina I, Davtyan H, Olkhanud PB, et al. Reducing AD-like pathology in 3xTg-AD mouse model by DNA epitope vaccine - a novel immunotherapeutic strategy. PLoS One 2008; 3: e2124, doi: 10.1371/journal.pone.0002124.
https://doi.org/10.1371/journal.pone.000...

24. Evans CF, Davtyan H, Petrushina I, Hovakimyan A, Davtyan A, Hannaman D, et al. Epitope-based DNA vaccine for Alzheimer's disease: Translational study in macaques. Alzheimers Dement 2013; pii: S1552-5260(13)00656-0.

25. Guo W, Sha S, Xing X, Jiang T, Cao Y. Reduction of cerebral Abeta burden and improvement in cognitive function in Tg-APPswe/PSEN1dE9 mice following vaccination with a multivalent Abeta3-10 DNA vaccine. Neurosci Lett 2013; 549: 109-115, doi: 10.1016/j.neulet.2013.06.018.
https://doi.org/10.1016/j.neulet.2013.06...

26. Yu YZ, Wang S, Bai JY, Zhao M, Chen A, Wang WB, et al. Effective DNA epitope chimeric vaccines for Alzheimer's disease using a toxin-derived carrier protein as a molecular adjuvant. Clin Immunol 2013; 149: 11-24, doi: 10.1016/j.clim.2013.05.016.
https://doi.org/10.1016/j.clim.2013.05.0...

27. Ghochikyan A, Davtyan H, Petrushina I, Hovakimyan A, Movsesyan N, Davtyan A, et al. Refinement of a DNA based Alzheimer's disease epitope vaccine in rabbits. Hum Vaccin Immunother 2013; 9: 1002-1010, doi: 10.4161/hv.23875.
https://doi.org/10.4161/hv.23875...

28. Matsumoto Y, Niimi N, Kohyama K. Development of a new DNA vaccine for Alzheimer disease targeting a wide range of abeta species and amyloidogenic peptides. PLoS One 2013; 8: e75203, doi: 10.1371/journal.pone.0075203.
https://doi.org/10.1371/journal.pone.007...
-2929. Davtyan H, Ghochikyan A, Hovakimyan A, Petrushina I, Yu J, Flyer D, et al. Immunostimulant patches containing Escherichia coliLT enhance immune responses to DNA- and recombinant protein-based Alzheimer's disease vaccines. J Neuroimmunol 2014; 268: 50-57, doi: 10.1016/j.jneuroim.2014.01.002.
https://doi.org/10.1016/j.jneuroim.2014....
). These kinds of vaccine formulations have been shown to generate immune responses, evidenced by the production of antibodies specific to Aβ but without cytotoxic cellular responses.

Approaches using the Aβ1-11 peptide derived from the fusion of Aβ with immunomodulator sequences such as PADRE, and associated with adjuvants or integrated into chimeric vaccines, such as virus-like particles, have been shown to be highly immunogenic, as seen by the induced humoral immune response with an indicative profile of T-helper 2 (Th2) cell modulation (2929. Davtyan H, Ghochikyan A, Hovakimyan A, Petrushina I, Yu J, Flyer D, et al. Immunostimulant patches containing Escherichia coliLT enhance immune responses to DNA- and recombinant protein-based Alzheimer's disease vaccines. J Neuroimmunol 2014; 268: 50-57, doi: 10.1016/j.jneuroim.2014.01.002.
https://doi.org/10.1016/j.jneuroim.2014....

30. Petrushina I, Ghochikyan A, Mktrichyan M, Mamikonyan G, Movsesyan N, Davtyan H, et al. Alzheimer's disease peptide epitope vaccine reduces insoluble but not soluble/oligomeric Abeta species in amyloid precursor protein transgenic mice. J Neurosci 2007; 27: 12721-12731, doi: 10.1523/JNEUROSCI.3201-07.2007.
https://doi.org/10.1523/JNEUROSCI.3201-0...

31. Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, et al. Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Abeta3-10 reduces AD-like pathology and cognitive impairment in Tg-APPswe/PSEN1dE9 mice. J Neuroimmunol 2012; 249: 16-26, doi: 10.1016/j.jneuroim.2012.04.014.
https://doi.org/10.1016/j.jneuroim.2012....

32. Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, et al. The second-generation active Abeta immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci 2011; 31: 9323-9331, doi: 10.1523/JNEUROSCI.0293-11.2011.
https://doi.org/10.1523/JNEUROSCI.0293-1...

33. Bach P, Tschape JA, Kopietz F, Braun G, Baade JK, Wiederhold KH, et al. Vaccination with Abeta-displaying virus-like particles reduces soluble and insoluble cerebral Abeta and lowers plaque burden in APP transgenic mice. J Immunol 2009; 182: 7613-7624, doi: 10.4049/jimmunol.0803366.
https://doi.org/10.4049/jimmunol.0803366...
-3434. Davtyan H, Ghochikyan A, Petrushina I, Hovakimyan A, Davtyan A, Poghosyan A, et al. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer's disease: prelude to a clinical trial. J Neurosci 2013; 33: 4923-4934, doi: 10.1523/JNEUROSCI.4672-12.2013.
https://doi.org/10.1523/JNEUROSCI.4672-1...
). The same applies for vaccines based on recombinant viruses, which code for epitopes of Aβ-specific to B cells, but this kind of vaccine approach remains expensive, may result in the generation of antibodies with altered epitope specificities, and carry a significant risk of inducing adverse effects (3535. Zou J, Yao Z, Zhang G, Wang H, Xu J, Yew DT, et al. Vaccination of Alzheimer's model mice with adenovirus vector containing quadrivalent foldable Abeta(1-15) reduces Abeta burden and behavioral impairment without Abeta-specific T cell response. J Neurol Sci 2008; 272: 87-98, doi: 10.1016/j.jns.2008.05.003.
https://doi.org/10.1016/j.jns.2008.05.00...
).

Vaccine strategies using DNA or peptides against AD based on various approaches have usually induced poor immune responses (the case with DNA) or antibodies with modest avidity for the target protein (the case with peptides). In attempts to enhance the magnitude of the antibody responses, heterologous prime-boost regimens have been tested, in which the first priming dose is followed by a boost based on a different delivery approach that promotes the expansion and selection of B cells with a high degree of avidity for the target antigen (3636. Kim HD, Tahara K, Maxwell JA, Lalonde R, Fukuiwa T, Fujihashi K, et al. Nasal inoculation of an adenovirus vector encoding 11 tandem repeats of Abeta1-6 upregulates IL-10 expression and reduces amyloid load in a Mo/Hu APPswe PS1dE9 mouse model of Alzheimer's disease. J Gene Med 2007; 9: 88-98, doi: 10.1002/jgm.993.
https://doi.org/10.1002/jgm.993...
,3737. Lambracht-Washington D, Qu BX, Fu M, Anderson LD Jr, Eagar TN, Stuve O, et al. A peptide prime-DNA boost immunization protocol provides significant benefits as a new generation Abeta42 DNA vaccine for Alzheimer disease. J Neuroimmunol 2013; 254: 63-68, doi: 10.1016/j.jneuroim.2012.09.008.
https://doi.org/10.1016/j.jneuroim.2012....
). The immune responses achieved with such a vaccine regimen were particularly promising, especially that proposed by Lambracht-Washington and colleagues (3737. Lambracht-Washington D, Qu BX, Fu M, Anderson LD Jr, Eagar TN, Stuve O, et al. A peptide prime-DNA boost immunization protocol provides significant benefits as a new generation Abeta42 DNA vaccine for Alzheimer disease. J Neuroimmunol 2013; 254: 63-68, doi: 10.1016/j.jneuroim.2012.09.008.
https://doi.org/10.1016/j.jneuroim.2012....
) based on a prime-boost immunization strategy in which a shortened Aβ peptide (Aβ1-42) containing both B- and T-cell epitopes was used. The volunteers were initially primed with a synthetic peptide, and then boosted with a DNA vaccine encoding the same target antigen. What is interesting in this approach is that although T cells were present in the initial stage of vaccination, the T cell level later decreased, indicating that the boost with the DNA vaccine promoted the activation of Treg cells, which were responsible for the low reactivity of Aβ-specific T cells (3737. Lambracht-Washington D, Qu BX, Fu M, Anderson LD Jr, Eagar TN, Stuve O, et al. A peptide prime-DNA boost immunization protocol provides significant benefits as a new generation Abeta42 DNA vaccine for Alzheimer disease. J Neuroimmunol 2013; 254: 63-68, doi: 10.1016/j.jneuroim.2012.09.008.
https://doi.org/10.1016/j.jneuroim.2012....
).

Another vaccine approach against AD has been based on the other pathological marker of the disease as the target antigen, the neurofibrillary tangles produced by hyperphosphorylation of the Tau protein (1111. Pritchard SM, Dolan PJ, Vitkus A, Johnson GV. The toxicity of tau in Alzheimer disease: turnover, targets and potential therapeutics. J Cell Mol Med 2011; 15: 1621-1635, doi: 10.1111/j.1582-4934.2011.01273.x.
https://doi.org/10.1111/j.1582-4934.2011...
). This approach is not without challenges: there has been at least one report of neuroinflammation in mice as a result of repeated immunization with phosphorylated Tau-derived peptides, raising concerns about the safety of this kind of vaccine (3838. Rozenstein-Tsalkovich L, Grigoriadis N, Lourbopoulos A, Nousiopoulou E, Kassis I, Abramsky O, et al. Repeated immunization of mice with phosphorylated-tau peptides causes neuroinflammation. Exp Neurol 2013; 248: 451-456, doi: 10.1016/j.expneurol.2013.07.006.
https://doi.org/10.1016/j.expneurol.2013...
).

Clinical studies: past, present, and future

The first clinical trial involving a vaccine against AD was carried out in 2000 with the aggregated human Aβ1-42 peptide combined with a saponin-based adjuvant (AN1792) (3939. Bayer AJ, Bullock R, Jones RW, Wilkinson D, Paterson KR, Jenkins L, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology 2005; 64: 94-101, doi: 10.1212/01.WNL.0000148604.77591.67.
https://doi.org/10.1212/01.WNL.000014860...
). The results of this phase I trial provided evidence of the safety and tolerability of the vaccine based on a multiple-dose regimen. However, adverse inflammatory effects, leading to subacute meningoencephalitis, were observed in nearly 6% of the volunteers enrolled in a phase II trial with the AN1792 vaccine, which ended dramatically after the death of one patient despite improvements in the clinical symptoms and reduction of senile plaques in several other patients (4040. Ferrer I, Boada RM, Sanchez Guerra ML, Rey MJ, Costa-Jussa F. Neuropathology and pathogenesis of encephalitis following amyloid-beta immunization in Alzheimer's disease. Brain Pathol 2004; 14: 11-20, doi: 10.1111/j.1750-3639.2004.tb00493.x.
https://doi.org/10.1111/j.1750-3639.2004...

41. Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller RO. Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med 2003; 9: 448-452, doi: 10.1038/nm840.
https://doi.org/10.1038/nm840...

42. Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003; 61: 46-54, doi: 10.1212/01.WNL.0000073623.84147.A8.
https://doi.org/10.1212/01.WNL.000007362...
-4343. Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005; 64: 1553-1562, doi: 10.1212/01.WNL.0000159740.16984.3C.
https://doi.org/10.1212/01.WNL.000015974...
). Subsequent studies showed that the adverse effects observed in the AN1792 trial could be ascribed to the toxicity mediated by activated T cells reacting with self-antigens, resulting in an inflammatory autoimmune response (4040. Ferrer I, Boada RM, Sanchez Guerra ML, Rey MJ, Costa-Jussa F. Neuropathology and pathogenesis of encephalitis following amyloid-beta immunization in Alzheimer's disease. Brain Pathol 2004; 14: 11-20, doi: 10.1111/j.1750-3639.2004.tb00493.x.
https://doi.org/10.1111/j.1750-3639.2004...
,4242. Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003; 61: 46-54, doi: 10.1212/01.WNL.0000073623.84147.A8.
https://doi.org/10.1212/01.WNL.000007362...
). The Aβ1-42 T-cell epitopes were located in the central region and carboxyl-terminus of that Aβ peptide (4444. Cribbs DH, Ghochikyan A, Vasilevko V, Tran M, Petrushina I, Sadzikava N, et al. Adjuvant-dependent modulation of Th1 and Th2 responses to immunization with beta-amyloid. Int Immunol 2003; 15: 505-514, doi: 10.1093/intimm/dxg049.
https://doi.org/10.1093/intimm/dxg049...
).

In order to avoid undesirable inflammatory effects, the amino-terminus of the Aβ peptide, in which the B-cell epitopes were located, was subsequently used as an antigen target for anti-AD vaccines (4545. Bard F, Barbour R, Cannon C, Carretto R, Fox M, Games D, et al. Epitope and isotype specificities of antibodies to beta-amyloid peptide for protection against Alzheimer's disease-like neuropathology. Proc Natl Acad Sci U S A 2003; 100: 2023-2028, doi: 10.1073/pnas.0436286100.
https://doi.org/10.1073/pnas.0436286100...
,4646. Mamikonyan G, Necula M, Mkrtichyan M, Ghochikyan A, Petrushina I, Movsesyan N, et al. Anti-A beta 1-11 antibody binds to different beta-amyloid species, inhibits fibril formation, and disaggregates preformed fibrils but not the most toxic oligomers. J Biol Chem 2007; 282: 22376-22386, doi: 10.1074/jbc.M700088200.
https://doi.org/10.1074/jbc.M700088200...
). CAD106, a vaccine candidate composed of a B-cell epitope (Aβ1-6) is currently being tested in a clinical trial. In this vaccine, the peptide was genetically fused to the bacteriophage Qβ coat protein to generate virus-like particles, each containing 180 copies of the coat protein of the phage. Phase I trials were performed to evaluate the safety, tolerability, and immunogenicity of this vaccine. The absence of adverse effects, e.g., autoimmune inflammation, allowed the start of phase 2 trials (4747. Winblad B, Andreasen N, Minthon L, Floesser A, Imbert G, Dumortier T, et al. Safety, tolerability, and antibody response of active Abeta immunotherapy with CAD106 in patients with Alzheimer's disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol 2012; 11: 597-604, doi: 10.1016/S1474-4422(12)70140-0.
https://doi.org/10.1016/S1474-4422(12)70...
). Another example that will soon have clinical trials initiated is the Lu AF20513 vaccine, composed of three B-cell epitopes (Aβ1-12) fused to two Th-cell epitopes derived from the tetanus toxoid, P2 and P30 (3434. Davtyan H, Ghochikyan A, Petrushina I, Hovakimyan A, Davtyan A, Poghosyan A, et al. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer's disease: prelude to a clinical trial. J Neurosci 2013; 33: 4923-4934, doi: 10.1523/JNEUROSCI.4672-12.2013.
https://doi.org/10.1523/JNEUROSCI.4672-1...
). The major purpose of this vaccination is the activation of memory Th cells, which are preexistent in the general segment of the population that has been vaccinated with a conventional vaccine against tetanus, facilitating the quick response against Aβ even in the elderly population (3434. Davtyan H, Ghochikyan A, Petrushina I, Hovakimyan A, Davtyan A, Poghosyan A, et al. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer's disease: prelude to a clinical trial. J Neurosci 2013; 33: 4923-4934, doi: 10.1523/JNEUROSCI.4672-12.2013.
https://doi.org/10.1523/JNEUROSCI.4672-1...
).

Conclusion: is a vaccine capable of preventing or treating AD feasible?

Studies performed during the last century allowed the identification of distinct features of AD, such as the accumulation of Aβ plaques in the brain, and the relationship of these deposits with the clinical manifestations. These observations opened perspectives for new therapeutic interventions for the control of the disease, particularly during the last decade. Studies focused on vaccines have advanced significantly and now represent a promising therapeutic alternative for disease control, based on the generation of antibodies against the Aβ peptide. These advances were accompanied by retreats, as in the case of the first clinical trials, that provided important lessons for researchers, who have deepened their knowledge and developed alternatives for the design of safer and more effective vaccines for the control of AD. Recently, the possibility of targeting proteins other than Aβ has been tested, and promising results are expected to be seen with the Tau protein, but clinical data are still lacking, and should be pursued in this kind of approach.

There has been some debate as to whether targeting Aβ would be sufficient for an immunologically based therapy, because the role of senile plaques in the clinical picture appears to be just the tip of the iceberg. Indeed, the fact that promising results generated in animal models have not been reproduced in clinical trials suggests that Aβ alone might not be the only target antigen for active immunization. The finding that the Tau protein could play a role as a target antigen for the control of AD adds further expectations regarding new vaccine formulations with better performance in human beings.

The multifactorial nature of the AD pathology makes it difficult to propose a “perfect target” for the development of drugs or immunotherapy. In the face of these difficulties, the contribution of passive immunotherapy based on monoclonal antibodies might find a more promising role in the treatment of AD. Nonetheless, recent results based on active immunization suggest that, in addition to a direct therapeutic effect in subjects already affected by the onset of disease, immunization should also be considered as a conventional prophylactic approach. Testing vaccines that are able to induce specific antibodies prior to the manifestation of symptoms may be an alternative to prevent amyloid being deposited in the senile plaques, according to the peripheral sink hypothesis. So far, such a preventive approach has not been experimentally proven but deserves future effort and support.

References

  • 1
    Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer's 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde”. Clin Anat 1995; 8: 429-431, doi: 10.1002/ca.980080612.
    » https://doi.org/10.1002/ca.980080612
  • 2
    Braak H, Braak E. Staging of Alzheimer's disease-related neurofibrillary changes. Neurobiol Aging 1995; 16: 271-278, doi: 10.1016/0197-4580(95)00021-6.
    » https://doi.org/10.1016/0197-4580(95)00021-6
  • 3
    Minião AM, Xu J, Kochanek KD. Deaths: preliminary data for 2008. NVSR (National Vital Statistics Reports) Publication PHS 2011-1120. Washington: Centers for Disease Control and Prevention, National Center for Health Statistics. U.S. Government Printing Office; 2010.
  • 4
    Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol 2003; 60: 1119-1122, doi: 10.1001/archneur.60.8.1119.
    » https://doi.org/10.1001/archneur.60.8.1119
  • 5
    Alzheimer's Disease International. World Alzheimer Report 2010. London: Alzheimer's Disease International; 2010.
  • 6
    United Nations PD. World population ageing: 1950-2050. New York: Department of Economic and Social Affairs; 2002.
  • 7
    Glenner GG, Wong CW. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984; 120: 885-890, doi: 10.1016/S0006-291X(84)80190-4.
    » https://doi.org/10.1016/S0006-291X(84)80190-4
  • 8
    Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 1985; 82: 4245-4249, doi: 10.1073/pnas.82.12.4245.
    » https://doi.org/10.1073/pnas.82.12.4245
  • 9
    Kurz A, Perneczky R. Amyloid clearance as a treatment target against Alzheimer's disease. J Alzheimers Dis 2011; 24 (Suppl 2): 61-73.
  • 10
    Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A. Tau is essential to beta-amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A 2002; 99: 6364-6369, doi: 10.1073/pnas.092136199.
    » https://doi.org/10.1073/pnas.092136199
  • 11
    Pritchard SM, Dolan PJ, Vitkus A, Johnson GV. The toxicity of tau in Alzheimer disease: turnover, targets and potential therapeutics. J Cell Mol Med 2011; 15: 1621-1635, doi: 10.1111/j.1582-4934.2011.01273.x.
    » https://doi.org/10.1111/j.1582-4934.2011.01273.x
  • 12
    Giacobini E. Modulation of brain acetylcholine levels with cholinesterase inhibitors as a treatment of Alzheimer disease. Keio J Med 1987; 36: 381-391, doi: 10.2302/kjm.36.381.
    » https://doi.org/10.2302/kjm.36.381
  • 13
    Greenamyre JT, Young AB. Excitatory amino acids and Alzheimer's disease. Neurobiol Aging 1989; 10: 593-602, doi: 10.1016/0197-4580(89)90143-7.
    » https://doi.org/10.1016/0197-4580(89)90143-7
  • 14
    Dröes RM, Van Mierlo LD, Van der Roest HG, Meiland FJM. Focus and effectiveness of psychosocial interventions for people with dementia in institutional care settings from the perspective of coping with the disease. Non Pharm Ther Dement 2010; 1: 139-161.
  • 15
    Moreth J, Mavoungou C, Schindowski K. Passive anti-amyloid immunotherapy in Alzheimer's disease: What are the most promising targets? Immun Ageing 2013; 10: 18, doi: 10.1186/1742-4933-10-18.
    » https://doi.org/10.1186/1742-4933-10-18
  • 16
    Khorassani F, Hilas O. Bapineuzumab, an Investigational Agent For Alzheimer's Disease. P T 2013; 38: 89-91, PMC3628177.
  • 17
    Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999; 400: 173-177, doi: 10.1038/22124.
    » https://doi.org/10.1038/22124
  • 18
    Solomon B, Koppel R, Frankel D, Hanan-Aharon E. Disaggregation of Alzheimer beta-amyloid by site-directed mAb. Proc Natl Acad Sci U S A 1997; 94: 4109-4112, doi: 10.1073/pnas.94.8.4109.
    » https://doi.org/10.1073/pnas.94.8.4109
  • 19
    Wilcock DM, Munireddy SK, Rosenthal A, Ugen KE, Gordon MN, Morgan D. Microglial activation facilitates Abeta plaque removal following intracranial anti-Abeta antibody administration. Neurobiol Dis 2004; 15: 11-20, doi: 10.1016/j.nbd.2003.09.015.
    » https://doi.org/10.1016/j.nbd.2003.09.015
  • 20
    DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2001; 98: 8850-8855, doi: 10.1073/pnas.151261398.
    » https://doi.org/10.1073/pnas.151261398
  • 21
    Hock C, Konietzko U, Papassotiropoulos A, Wollmer A, Streffer J, von Rotz RC, et al. Generation of antibodies specific for beta-amyloid by vaccination of patients with Alzheimer disease. Nat Med 2002; 8: 1270-1275, doi: 10.1038/nm783.
    » https://doi.org/10.1038/nm783
  • 22
    Weiner HL, Frenkel D. Immunology and immunotherapy of Alzheimer's disease. Nat Rev Immunol 2006; 6: 404-416, doi: 10.1038/nri1843.
    » https://doi.org/10.1038/nri1843
  • 23
    Movsesyan N, Ghochikyan A, Mkrtichyan M, Petrushina I, Davtyan H, Olkhanud PB, et al. Reducing AD-like pathology in 3xTg-AD mouse model by DNA epitope vaccine - a novel immunotherapeutic strategy. PLoS One 2008; 3: e2124, doi: 10.1371/journal.pone.0002124.
    » https://doi.org/10.1371/journal.pone.0002124
  • 24
    Evans CF, Davtyan H, Petrushina I, Hovakimyan A, Davtyan A, Hannaman D, et al. Epitope-based DNA vaccine for Alzheimer's disease: Translational study in macaques. Alzheimers Dement 2013; pii: S1552-5260(13)00656-0.
  • 25
    Guo W, Sha S, Xing X, Jiang T, Cao Y. Reduction of cerebral Abeta burden and improvement in cognitive function in Tg-APPswe/PSEN1dE9 mice following vaccination with a multivalent Abeta3-10 DNA vaccine. Neurosci Lett 2013; 549: 109-115, doi: 10.1016/j.neulet.2013.06.018.
    » https://doi.org/10.1016/j.neulet.2013.06.018
  • 26
    Yu YZ, Wang S, Bai JY, Zhao M, Chen A, Wang WB, et al. Effective DNA epitope chimeric vaccines for Alzheimer's disease using a toxin-derived carrier protein as a molecular adjuvant. Clin Immunol 2013; 149: 11-24, doi: 10.1016/j.clim.2013.05.016.
    » https://doi.org/10.1016/j.clim.2013.05.016
  • 27
    Ghochikyan A, Davtyan H, Petrushina I, Hovakimyan A, Movsesyan N, Davtyan A, et al. Refinement of a DNA based Alzheimer's disease epitope vaccine in rabbits. Hum Vaccin Immunother 2013; 9: 1002-1010, doi: 10.4161/hv.23875.
    » https://doi.org/10.4161/hv.23875
  • 28
    Matsumoto Y, Niimi N, Kohyama K. Development of a new DNA vaccine for Alzheimer disease targeting a wide range of abeta species and amyloidogenic peptides. PLoS One 2013; 8: e75203, doi: 10.1371/journal.pone.0075203.
    » https://doi.org/10.1371/journal.pone.0075203
  • 29
    Davtyan H, Ghochikyan A, Hovakimyan A, Petrushina I, Yu J, Flyer D, et al. Immunostimulant patches containing Escherichia coliLT enhance immune responses to DNA- and recombinant protein-based Alzheimer's disease vaccines. J Neuroimmunol 2014; 268: 50-57, doi: 10.1016/j.jneuroim.2014.01.002.
    » https://doi.org/10.1016/j.jneuroim.2014.01.002
  • 30
    Petrushina I, Ghochikyan A, Mktrichyan M, Mamikonyan G, Movsesyan N, Davtyan H, et al. Alzheimer's disease peptide epitope vaccine reduces insoluble but not soluble/oligomeric Abeta species in amyloid precursor protein transgenic mice. J Neurosci 2007; 27: 12721-12731, doi: 10.1523/JNEUROSCI.3201-07.2007.
    » https://doi.org/10.1523/JNEUROSCI.3201-07.2007
  • 31
    Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, et al. Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Abeta3-10 reduces AD-like pathology and cognitive impairment in Tg-APPswe/PSEN1dE9 mice. J Neuroimmunol 2012; 249: 16-26, doi: 10.1016/j.jneuroim.2012.04.014.
    » https://doi.org/10.1016/j.jneuroim.2012.04.014
  • 32
    Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, et al. The second-generation active Abeta immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci 2011; 31: 9323-9331, doi: 10.1523/JNEUROSCI.0293-11.2011.
    » https://doi.org/10.1523/JNEUROSCI.0293-11.2011
  • 33
    Bach P, Tschape JA, Kopietz F, Braun G, Baade JK, Wiederhold KH, et al. Vaccination with Abeta-displaying virus-like particles reduces soluble and insoluble cerebral Abeta and lowers plaque burden in APP transgenic mice. J Immunol 2009; 182: 7613-7624, doi: 10.4049/jimmunol.0803366.
    » https://doi.org/10.4049/jimmunol.0803366
  • 34
    Davtyan H, Ghochikyan A, Petrushina I, Hovakimyan A, Davtyan A, Poghosyan A, et al. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer's disease: prelude to a clinical trial. J Neurosci 2013; 33: 4923-4934, doi: 10.1523/JNEUROSCI.4672-12.2013.
    » https://doi.org/10.1523/JNEUROSCI.4672-12.2013
  • 35
    Zou J, Yao Z, Zhang G, Wang H, Xu J, Yew DT, et al. Vaccination of Alzheimer's model mice with adenovirus vector containing quadrivalent foldable Abeta(1-15) reduces Abeta burden and behavioral impairment without Abeta-specific T cell response. J Neurol Sci 2008; 272: 87-98, doi: 10.1016/j.jns.2008.05.003.
    » https://doi.org/10.1016/j.jns.2008.05.003
  • 36
    Kim HD, Tahara K, Maxwell JA, Lalonde R, Fukuiwa T, Fujihashi K, et al. Nasal inoculation of an adenovirus vector encoding 11 tandem repeats of Abeta1-6 upregulates IL-10 expression and reduces amyloid load in a Mo/Hu APPswe PS1dE9 mouse model of Alzheimer's disease. J Gene Med 2007; 9: 88-98, doi: 10.1002/jgm.993.
    » https://doi.org/10.1002/jgm.993
  • 37
    Lambracht-Washington D, Qu BX, Fu M, Anderson LD Jr, Eagar TN, Stuve O, et al. A peptide prime-DNA boost immunization protocol provides significant benefits as a new generation Abeta42 DNA vaccine for Alzheimer disease. J Neuroimmunol 2013; 254: 63-68, doi: 10.1016/j.jneuroim.2012.09.008.
    » https://doi.org/10.1016/j.jneuroim.2012.09.008
  • 38
    Rozenstein-Tsalkovich L, Grigoriadis N, Lourbopoulos A, Nousiopoulou E, Kassis I, Abramsky O, et al. Repeated immunization of mice with phosphorylated-tau peptides causes neuroinflammation. Exp Neurol 2013; 248: 451-456, doi: 10.1016/j.expneurol.2013.07.006.
    » https://doi.org/10.1016/j.expneurol.2013.07.006
  • 39
    Bayer AJ, Bullock R, Jones RW, Wilkinson D, Paterson KR, Jenkins L, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology 2005; 64: 94-101, doi: 10.1212/01.WNL.0000148604.77591.67.
    » https://doi.org/10.1212/01.WNL.0000148604.77591.67
  • 40
    Ferrer I, Boada RM, Sanchez Guerra ML, Rey MJ, Costa-Jussa F. Neuropathology and pathogenesis of encephalitis following amyloid-beta immunization in Alzheimer's disease. Brain Pathol 2004; 14: 11-20, doi: 10.1111/j.1750-3639.2004.tb00493.x.
    » https://doi.org/10.1111/j.1750-3639.2004.tb00493.x
  • 41
    Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller RO. Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med 2003; 9: 448-452, doi: 10.1038/nm840.
    » https://doi.org/10.1038/nm840
  • 42
    Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003; 61: 46-54, doi: 10.1212/01.WNL.0000073623.84147.A8.
    » https://doi.org/10.1212/01.WNL.0000073623.84147.A8
  • 43
    Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005; 64: 1553-1562, doi: 10.1212/01.WNL.0000159740.16984.3C.
    » https://doi.org/10.1212/01.WNL.0000159740.16984.3C
  • 44
    Cribbs DH, Ghochikyan A, Vasilevko V, Tran M, Petrushina I, Sadzikava N, et al. Adjuvant-dependent modulation of Th1 and Th2 responses to immunization with beta-amyloid. Int Immunol 2003; 15: 505-514, doi: 10.1093/intimm/dxg049.
    » https://doi.org/10.1093/intimm/dxg049
  • 45
    Bard F, Barbour R, Cannon C, Carretto R, Fox M, Games D, et al. Epitope and isotype specificities of antibodies to beta-amyloid peptide for protection against Alzheimer's disease-like neuropathology. Proc Natl Acad Sci U S A 2003; 100: 2023-2028, doi: 10.1073/pnas.0436286100.
    » https://doi.org/10.1073/pnas.0436286100
  • 46
    Mamikonyan G, Necula M, Mkrtichyan M, Ghochikyan A, Petrushina I, Movsesyan N, et al. Anti-A beta 1-11 antibody binds to different beta-amyloid species, inhibits fibril formation, and disaggregates preformed fibrils but not the most toxic oligomers. J Biol Chem 2007; 282: 22376-22386, doi: 10.1074/jbc.M700088200.
    » https://doi.org/10.1074/jbc.M700088200
  • 47
    Winblad B, Andreasen N, Minthon L, Floesser A, Imbert G, Dumortier T, et al. Safety, tolerability, and antibody response of active Abeta immunotherapy with CAD106 in patients with Alzheimer's disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol 2012; 11: 597-604, doi: 10.1016/S1474-4422(12)70140-0.
    » https://doi.org/10.1016/S1474-4422(12)70140-0
  • First published online May 23, 2014.

Publication Dates

  • Publication in this collection
    June 2014

History

  • Received
    17 Aug 2013
  • Accepted
    31 Mar 2014
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br