全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Targeting Beta Amyloid: A Clinical Review of Immunotherapeutic Approaches in Alzheimer's Disease

DOI: 10.1155/2012/628070

Full-Text   Cite this paper   Add to My Lib

Abstract:

As the societal and economic burdens of Alzheimer's disease (AD) continue to mount, so does the need for therapies that slow the progression of the illness. Beta amyloid has long been recognized as the pathologic hallmark of AD, and the past decade has seen significant progress in the development of various immunotherapeutic approaches targeting beta amyloid. This paper reviews active and passive approaches aimed at beta amyloid, with a focus on clinical trial data. 1. Introduction Alzheimer’s disease (AD) is by far the most common form of dementia, and the social and economic burdens of AD continue to mount. In 2010, an estimated 36 million people worldwide were living with dementia—a number that is projected to increase to 66 million in 2030, and 115 million in 2050 [1]. According to the World Alzheimer Report, the worldwide cost of dementia is estimated at USD $604 billion for 2010 [1], and according to one model, this cost has increased by 34% between 2005 and 2009 [2]. These statistics must be considered in parallel with the immeasurable emotional and psychological burdens that AD places on patients and families. Significant progress in the treatment of AD has been made since the initial description of the disease by Alois Alzheimer in 1907 [3]. Cholinesterase inhibitors and memantine are potential therapies for the management of many cognitive symptoms of AD, but these neurotransmitter-based approaches do not address the underlying pathology of the illness, and ultimately fail to prevent its progression. The pathologic triad of AD—the accumulation of toxic beta amyloid with the formation of extracellular beta-amyloid-containing plaques, the development of intracellular neurofibrillary tangles, and the degeneration of cerebral neurons—provides numerous potential targets for disease-modifying therapies. Multiple lines of evidence now suggest, however, that it is the production and/or deposition of toxic forms of beta amyloid, along with the slowing of beta-amyloid clearance, that act as the central and primary events in AD pathogenesis, while neurofibrillary tangle formation and neuronal cell death occur downstream in this amyloid cascade [4–6]. Recent in vitro work has demonstrated that beta-amyloid dimers (the major form of soluble oligomers in the human brain) isolated from patients with AD induce both the abnormal phosphorylation of tau that is characteristic of AD and the degeneration of neurites, providing further confirmation of the pivotal role of beta amyloid in the pathogenesis of AD [7]. The search for a disease modifying therapy—one that

References

[1]  A. Wimo and M. Prince, World Alzheimer Report 2010: The Global Economic Impact of Dementia, Alzheimers’ Disease International, London, UK, 2010.
[2]  A. Wimo, B. Winblad, and L. J?nsson, “The worldwide societal costs of dementia: estimates for 2009,” Alzheimer's and Dementia, vol. 6, no. 2, pp. 98–103, 2010.
[3]  A. Alzheimer, “über eine eigenartige Erkrankung der Hirnrinde [On a peculiar disease of the cerebral cortex],” Allgemeine Zeitschrift fur Psychiatrie und Psychisch-Gerichtliche Medizin, vol. 64, pp. 146–148, 1907.
[4]  J. Hardy and D. J. Selkoe, “The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics,” Science, vol. 297, no. 5580, pp. 353–356, 2002.
[5]  A. Caccamo, S. Oddo, M. C. Sugarman, Y. Akbari, and F. M. LaFerla, “Age- and region-dependent alterations in Aβ-degrading enzymes: implications for Aβ-induced disorders,” Neurobiology of Aging, vol. 26, no. 5, pp. 645–654, 2005.
[6]  K. G. Mawuenyega, W. Sigurdson, V. Ovod et al., “Decreased clearance of CNS β-amyloid in Alzheimer's disease,” Science, vol. 330, no. 6012, p. 1774, 2010.
[7]  M. Jin, N. Shepardson, T. Yang, G. Chen, D. Walsh, and D. J. Selkoe, “Soluble amyloid β-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 14, pp. 5819–5824, 2011.
[8]  Biomarkers Definitions Working Group, “Biomarkers and surrogate endpoints: preferred definitions and conceptual framework,” Clinical Pharmacology and Therapeutics, vol. 69, no. 3, pp. 89–95, 2001.
[9]  W. E. Klunk, H. Engler, A. Nordberg et al., “Imaging brain amyloid in Alzheimer's disease with Pittsburgh compound-B,” Annals of Neurology, vol. 55, no. 3, pp. 306–319, 2004.
[10]  C. R. Jack Jr., V. J. Lowe, S. D. Weigand et al., “Serial PIB and MRI in normal, mild cognitive impairment and Alzheimer's disease: implications for sequence of pathological events in Alzheimers disease,” Brain, vol. 132, no. 5, pp. 1355–1365, 2009.
[11]  N. Andreasen and K. Blennow, “CSF biomarkers for mild cognitive impairment and early Alzheimer's disease,” Clinical Neurology and Neurosurgery, vol. 107, no. 3, pp. 165–173, 2005.
[12]  K. Buerger, M. Ewers, T. Pirttil? et al., “CSF phosphorylated tau protein correlates with neocortical neurofibrillary pathology in Alzheimer's disease,” Brain, vol. 129, no. 11, pp. 3035–3041, 2006.
[13]  J. M. Schott, S. J. Crutch, C. Frost, E. K. Warrington, M. N. Rossor, and N. C. Fox, “Neuropsychological correlates of whole brain atrophy in Alzheimer's disease,” Neuropsychologia, vol. 46, no. 6, pp. 1732–1737, 2008.
[14]  T. Wisniewski and E. M. Sigurdsson, “Murine models of Alzheimer's disease and their use in developing immunotherapies,” Biochimica et Biophysica Acta, vol. 1802, no. 10, pp. 847–859, 2010.
[15]  D. Schenk, R. Barbour, W. Dunn et al., “Immunization with amyloid-β attenuates Alzheimer disease-like pathology in the PDAPP mouse,” Nature, vol. 400, no. 6740, pp. 173–177, 1999.
[16]  D. Morgan, D. M. Diamond, P. E. Gottschall et al., “A β peptide vaccination prevents memory loss in an animal model of Alzheimer's disease,” Nature, vol. 408, no. 6815, pp. 982–985, 2000.
[17]  C. Janus, J. Pearson, J. McLaurin et al., “Aβ peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease,” Nature, vol. 408, no. 6815, pp. 979–982, 2000.
[18]  F. Bard, C. Cannon, R. Barbour et al., “Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease,” Nature Medicine, vol. 6, no. 8, pp. 916–919, 2000.
[19]  D. M. Wilcock, A. Rojiani, A. Rosenthal et al., “Passive amyloid immunotherapy clears amyloid and transiently activates microglia in a transgenic mouse model of amyloid deposition,” Journal of Neuroscience, vol. 24, no. 27, pp. 6144–6151, 2004.
[20]  W. Zago, M. Buttini, T. A. Comery, et al., “Neutralization of soluble Aβ species by the N-terminal anti-Aβ antibody 3D6,” in Proceedings of the International Conference on Alzheimer’s & Parkinson’s Diseases, Barcelona, Spain, March 2011, Abstract 1537.
[21]  P. Das, V. Howard, N. Loosbrock, D. Dickson, M. P. Murphy, and T. E. Golde, “Amyloid-β immunization effectively reduces amyloid deposition in FcRγ-/- knock-out mice,” Journal of Neuroscience, vol. 23, no. 24, pp. 8532–8538, 2003.
[22]  B. J. Bacskai, S. T. Kajdasz, M. E. McLellan et al., “Non-Fc-mediated mechanisms are involved in clearance of amyloid-β in vivo by immunotherapy,” Journal of Neuroscience, vol. 22, no. 18, pp. 7873–7878, 2002.
[23]  R. B. DeMattos, K. R. Bales, D. J. Cummins, J. C. Dodart, S. M. Paul, and D. M. Holtzman, “Peripheral anti-Aβ antibody alters CNS and plasma Aβ clearance and decreases brain Aβ burden in a mouse model of Alzheimer's disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 15, pp. 8850–8855, 2001.
[24]  B. Solomon, R. Koppel, E. Hanan, and T. Katzav, “Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer β-amyloid peptide,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 1, pp. 452–455, 1996.
[25]  J. Magga, L. Puli, R. Pihlaja et al., “Human intravenous immunoglobulin provides protection against Aβ toxicity by multiple mechanisms in a mouse model of Alzheimer's disease,” Journal of Neuroinflammation, vol. 7, p. 90, 2010.
[26]  M. Britschgi, C. E. Olin, H. T. Johns et al., “Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer's disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 29, pp. 12145–12150, 2009.
[27]  R. Dodel, F. Neff, C. Noelker et al., “Intravenous immunoglobulins as a treatment for Alzheimer's disease: rationale and current evidence,” Drugs, vol. 70, no. 5, pp. 513–528, 2010.
[28]  A. J. Bayer, R. Bullock, R. W. Jones et al., “Evaluation of the safety and immunogenicity of synthetic Aβ42 (AN1792) in patients with AD,” Neurology, vol. 64, no. 1, pp. 94–101, 2005.
[29]  J. A. R. Nicoll, D. Wilkinson, C. Holmes, P. Steart, H. Markham, and R. O. Weller, “Neuropathology of human Alzheimer disease after immunization with amyloid-β peptide: a case report,” Nature Medicine, vol. 9, no. 4, pp. 448–452, 2003.
[30]  S. Gilman, M. Koller, R. S. Black et al., “Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial,” Neurology, vol. 64, no. 9, pp. 1553–1562, 2005.
[31]  J. M. Orgogozo, S. Gilman, J. F. Dartigues et al., “Subacute meningoencephalitis in a subset of patients with AD after Aβ42 immunization,” Neurology, vol. 61, no. 1, pp. 46–54, 2003.
[32]  B. Vellas, R. Black, L. J. Thal et al., “Long-term follow-up of patients immunized with AN1792: reduced functional decline in antibody responders,” Current Alzheimer Research, vol. 6, no. 2, pp. 144–151, 2009.
[33]  M. Pride, P. Seubert, M. Grundman, M. Hagen, J. Eldridge, and R. S. Black, “Progress in the active immunotherapeutic approach to Alzheimer's disease: clinical investigations into AN1792-associated meningoencephalitis,” Neurodegenerative Diseases, vol. 5, no. 3-4, pp. 194–196, 2008.
[34]  I. Ferrer, M. B. Rovira, M. L. S. Guerra, M. J. Rey, and F. Costa-Jussá, “Neuropathology and pathogenesis of encephalitis following amyloid-β immunization in Alzheimer's disease,” Brain Pathology, vol. 14, no. 1, pp. 11–20, 2004.
[35]  N. C. Fox, R. S. Black, S. Gilman et al., “Effects of Aβ immunization (AN1792) on MRI measures of cerebral volume in Alzheimer disease,” Neurology, vol. 64, no. 9, pp. 1563–1572, 2005.
[36]  J. A. R. Nicoll, E. Barton, D. Boche et al., “Aβ species removal after Aβ42 immunization,” Journal of Neuropathology and Experimental Neurology, vol. 65, no. 11, pp. 1040–1048, 2006.
[37]  E. Masliah, L. Hansen, A. Adame et al., “Aβ vaccination effects on plaque pathology in the absence of encephalitis in Alzheimer disease,” Neurology, vol. 64, no. 1, pp. 129–131, 2005.
[38]  R. L. Patton, W. M. Kalback, C. L. Esh et al., “Amyloid-β peptide remnants in AN-1792-immunized Alzheimer's disease patients: a biochemical analysis,” American Journal of Pathology, vol. 169, no. 3, pp. 1048–1063, 2006.
[39]  A. Serrano-Pozo, C. M. William, I. Ferrer et al., “Beneficial effect of human anti-amyloid-β active immunization on neurite morphology and tau pathology,” Brain, vol. 133, no. 5, pp. 1312–1327, 2010.
[40]  C. Holmes, D. Boche, D. Wilkinson et al., “Long-term effects of Aβ42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial,” The Lancet, vol. 372, no. 9634, pp. 216–223, 2008.
[41]  M. Lee, F. Bard, K. Johnson-Wood et al., “Aβ42 immunization in Alzheimer's disease generates Aβ N-terminal antibodies,” Annals of Neurology, vol. 58, no. 3, pp. 430–435, 2005.
[42]  M. Hagen, G. P. Seubert, S. Jacobsen, et al., “The Aβ peptide conjugate vaccine, ACC-001, generates N-terminal anti-Aβ antibodies in the absence of Aβ directed T-cell responses,” in Proceedings of the Alzheimer’s Association International Conference on Alzheimer’s Disease, Paris, France, July 2011, Abstract P2-461.
[43]  B. Winblad, “Safety, tolerability and immunogenicity of the Aβ immunotherapeutic vaccine CAD106 in a first-in-man study in Alzheimer patients,” Alzheimer's and Dementia, vol. 4, no. 4, p. T128, 2008.
[44]  B. G. Winblad, L. Minthon, A. Floesser, et al., “Results of the first-in-man study with the active Aβ Immunotherapy CAD106 in Alzheimer patients,” Alzheimer's and Dementia, vol. 5, no. 4, pp. P113–P114, 2009.
[45]  A. Graf, N. Andreasen, M. E. Riviere, et al., “Optimization of the treatment regimen with active Aβ immunotherapy CAD106 in alzheimer patients,” Alzheimer's and Dementia, vol. 6, no. 4, p. S532, 2010.
[46]  M. J. Savage, G. Wu, A. McCampbell, et al., “A novel multivalent Abeta peptide vaccine with preclinical evidence of a central immune response that generates antisera recognizing a wide range of abeta peptide species,” Alzheimer's and Dementia, vol. 6, no. 4, p. S142, 2010.
[47]  A. Schneeberger, M. Mandler, O. Otava, W. Zauner, F. Mattner, and W. Schmidt, “Development of AFFITOPE vaccines for Alzheimer's disease (AD)—from concept to clinical testing,” Journal of Nutrition, Health and Aging, vol. 13, no. 3, pp. 264–267, 2009.
[48]  A. Schneeberger, M. Mandler, W. Zauner, et al., “Development of AFFITOPE vaccines for Alzheimer's disease,” Alzheimer's and Dementia, vol. 6, no. 4, pp. S584–S585, 2010.
[49]  R. S. Black, R. A. Sperling, B. Safirstein et al., “A single ascending dose study of bapineuzumab in patients with alzheimer disease,” Alzheimer Disease and Associated Disorders, vol. 24, no. 2, pp. 198–203, 2010.
[50]  R. A. Sperling, C. R. Jack Jr., S. E. Black et al., “Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer's Association Research Roundtable Workgroup,” Alzheimer's and Dementia, vol. 7, no. 4, pp. 367–385, 2011.
[51]  S. Salloway, R. Sperling, S. Gilman et al., “A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease,” Neurology, vol. 73, no. 24, pp. 2061–2070, 2009.
[52]  K. Blennow, H. Zetterberg, J. Wei, E. Liu, R. Black, and M. Grundman, “Immunotherapy with bapineuzumab lowers CSF tau protein levels in patients with Alzheimer's disease,” Alzheimer's and Dementia, vol. 6, no. 4, pp. S134–S135, 2010.
[53]  K. Chalmers, G. K. Wilcock, and S. Love, “APOE ε4 influences the pathological phenotype of Alzheimer's disease by favouring cerebrovascular over parenchymal accumulation of Aβ protein,” Neuropathology and Applied Neurobiology, vol. 29, no. 3, pp. 231–238, 2003.
[54]  S. Crean, A. Ward, C. J. Mercaldi et al., “Apolipoprotein E ε4 prevalence in Alzheimer's disease patients varies across global populations: a systematic literature review and meta-analysis,” Dementia and Geriatric Cognitive Disorders, vol. 31, no. 1, pp. 20–30, 2011.
[55]  C. Kinnecom, M. H. Lev, L. Wendell et al., “Course of cerebral amyloid angiopathy-related inflammation,” Neurology, vol. 68, no. 17, pp. 1411–1416, 2007.
[56]  J. A. Eng, M. P. Frosch, K. Choi, G. W. Rebeck, and S. M. Greenberg, “Clinical manifestations of cerebral amyloid angiopathy-related inflammation,” Annals of Neurology, vol. 55, no. 2, pp. 250–256, 2004.
[57]  J. O. Rinne, D. J. Brooks, M. N. Rossor et al., “11C-PiB PET assessment of change in fibrillar amyloid-β load in patients with Alzheimer's disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study,” The Lancet Neurology, vol. 9, no. 4, pp. 363–372, 2010.
[58]  B. J. Bacskai, M. P. Frosch, S. H. Freeman et al., “Molecular imaging with Pittsburgh compound B confirmed at autopsy: a case report,” Archives of Neurology, vol. 64, no. 3, pp. 431–434, 2007.
[59]  K. A. Johnson, M. Gregas, J. A. Becker et al., “Imaging of amyloid burden and distribution in cerebral amyloid angiopathy,” Annals of Neurology, vol. 62, no. 3, pp. 229–234, 2007.
[60]  E. R. Siemers, S. Friedrich, R. A. Dean et al., “Safety and changes in plasma and cerebrospinal fluid amyloid β after a single administration of an amyloid β monoclonal antibody in subjects with Alzheimer disease,” Clinical Neuropharmacology, vol. 33, no. 2, pp. 67–73, 2010.
[61]  T. Goto, S. Fujikoshi, K. Uenaka, et al., “Solanezumab was safe and well-tolerated for Asian patients with mild-to-moderate Alzheimer's disease in a multicenter, randomized, open-label, multi-dose study,” Alzheimer's and Dementia, vol. 6, no. 4, p. S308, 2010.
[62]  R. B. DeMattos, M. M. Racke, V. Gelfanova, et al., “Identification, characterization, and comparison of amino-terminally truncated Aβ42 peptides in Alzheimer's disease brain tissue and in plasma from Alzheimer's patients receiving solanezumab immunotherapy treatment,” Alzheimer's and Dementia, vol. 5, no. 4, pp. P156–P157, 2009.
[63]  R. B. DeMattos, M. Racke, V. Gelfanova, et al., “Characterization of amino-terminally truncated Abeta-42 peptides in plasma from Alzheimer's patients receiving solanezumab immunotherapy treatment,” Alzheimer's and Dementia, vol. 6, no. 4, p. S542, 2010.
[64]  E. R. Siemers, R. B. Demattos, F. Stuart, et al., “Use of a monoclonal anti-A antibody with biochemical and imaging biomarkers to determine amyloid plaque load in patients with Alzheimer's disease (AD) and control subjects,” in Proceedings of the 61st Annual Meeting of the American Association of Neurology, Seattle, Wash, USA, April 2009, Abstract IN3-2.009.
[65]  E. R. Siemers, R. A. Dean, D. R. Lachno, et al., “Measurement of cerebrospinal fluid total tau and phospho-tau in phase 2 trials of therapies targeting Aβ,” Alzheimer's and Dementia, vol. 5, no. 4, p. P258, 2009.
[66]  E. Siemers, “Biochemical biomarkers as endpoints in clinical trials: applications in phase 1, 2 and 3 studies,” Alzheimer's and Dementia, vol. 5, no. 4, p. P95, 2009.
[67]  A. H. Burstein, Q. Zhao, J. Ross, et al., “Safety and pharmacokinetics following a single 10-minute intravenous infusion of the anti-amyloid mAb ponezumab (PF-04360365) in patients with mild to moderate Alzheimer's disease,” Alzheimer's and Dementia, vol. 6, no. 4, p. S585, 2010.
[68]  J. W. Landen, Q. Zhao, S. Cohen, et al., “Safety and pharmacokinetics following a single infusion of the anti-amyloid monoclonal antibody ponezumab (PF-04360365) in patients with mild-to-moderate Alzheimer's disease: final results,” Alzheimer's and Dementia, vol. 6, no. 4, p. e57, 2010.
[69]  Q. Zhao, J. Landen, A. H. Burstein, et al., “Pharmacokinetics and pharmacodynamics of ponezumab (PF-04360365) following a single-dose intravenous infusion in patients with mild to moderate Alzheimer's disease,” Alzheimer's and Dementia, vol. 6, no. 4, p. S143, 2010.
[70]  T. Nicholas, W. Knebel, M. R. Gastonguay, et al., “Preliminary population pharmacokinetic modeling of PF-04360365, a humanized anti-amyloid monoclonal antibody, in patients with mild-to-moderate Alzheimer's disease,” Alzheimer's and Dementia, vol. 5, no. 4, p. P253, 2009.
[71]  K. M. Wood, F. McCush, J. J. Conboy, et al., “IP/MS analysis of human CSF Aβ following a single dose of the C-terminal anti-Aβ antibody ponezumab (PF-04360365) to Alzheimer patients,” Alzheimer's and Dementia, vol. 6, no. 4, p. S311, 2010.
[72]  Y. Fujimoto, I. Miyoshi, T. Ishibashi, et al., “Safety of the anti-amyloid monoclonal antibody ponezumab (PF-04360365) following a single-dose intravenous infusion in Japanese patients with mild-to-moderate Alzheimer's disease: preliminary results,” Alzheimer's and Dementia, vol. 6, no. 4, p. S310, 2010.
[73]  Q. Zhao, S. Abe, I. Miyoshi, et al., “Pharmacokinetics of ponezumab (PF-04360365) following a single-dose intravenous infusion in Japanese patients with mild-to-moderate Alzheimer's disease: preliminary results,” Alzheimer's and Dementia, vol. 6, no. 4, p. S305, 2010.
[74]  Eisai Co., Ltd., “Eisai announces the start of the first clinical study of BAN2401, a novel monoclonal antibody targeting the neurotoxic protofibrils believed to cause Alzheimer's disease,” [press release], Eisai Co., Ltd., 2010, http://www.eisai.com/news/enews201044pdf.pdf.
[75]  R. C. Dodel, Y. Du, C. Depboylu et al., “Intravenous immunoglobulins containing antibodies against β-amyloid for the treatment of Alzheimer's disease,” Journal of Neurology, Neurosurgery and Psychiatry, vol. 75, no. 10, pp. 1472–1474, 2004.
[76]  N. R. Relkin, P. Szabo, B. Adamiak et al., “18-Month study of intravenous immunoglobulin for treatment of mild Alzheimer disease,” Neurobiology of Aging, vol. 30, no. 11, pp. 1728–1736, 2009.
[77]  H. Fillit, G. Hess, J. Hill, P. Bonnet, and C. Toso, “IV immunoglobulin is associated with a reduced risk of Alzheimer disease and related disorders,” Neurology, vol. 73, no. 3, pp. 180–185, 2009.
[78]  D. Tsakanikas and N. Relkin, “Neuropsychological outcomes following 18-months of uninterrupted intravenous immunoglobulin (IVIg) treatment in patients with Alzheimer's disease (AD),” in Proceedings of the 62nd Annual Meeting of the American Academy of Neurology, Toronto, Ontario, Canada, April 2010, Abstract S34.005.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133