Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-26T23:01:23.401Z Has data issue: false hasContentIssue false

Aβ-Immunotherapeutic strategies: a wide range of approaches for Alzheimer's disease treatment

Published online by Cambridge University Press:  30 June 2016

Laia Montoliu-Gaya
Affiliation:
Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Facultat de Biociències, 08193 Bellaterra, Barcelona, Spain
Sandra Villegas*
Affiliation:
Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Facultat de Biociències, 08193 Bellaterra, Barcelona, Spain
*
*Corresponding author: Sandra Villegas, Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Facultat de Biociències, 08193 Bellaterra, Barcelona, Spain. E-mail: sandra.villegas@uab.cat

Abstract

Current therapies to treat Alzheimer's disease (AD) are focused on ameliorating symptoms instead of treating the underlying causes of AD. The accumulation of amyloid β (Aβ) oligomers, whether by an increase in production or by a decrease in clearance, has been described as the seed that initiates the pathological cascade in AD. Developing therapies to target these species is a vital step in improving AD treatment. Aβ-immunotherapy, especially passive immunotherapy, is a promising approach to reduce the Aβ burden. Up to now, several monoclonal antibodies (mAbs) have been tested in clinical trials on humans, but none of them have passed Phase III. In all likelihood, these trials failed mainly because patients with mild-to-moderate AD were recruited, and thus treatment may have been too late to be effective. Therefore, many ongoing clinical trials are being conducted in patients at the prodromal stage. New structures based on antibody fragments have been engineered intending to improve efficacy and safety. This review presents the properties of this variety of developing treatments and provides a perspective on state-of-the-art of passive Aβ-immunotherapy in AD.

Type
Review
Copyright
Copyright © Cambridge University Press 2016 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

1. Alzheimer's Disease International. (2014) World Alzheimer Report 2014: Dementia and Risk Reduction Google Scholar
2. Alzheimer's Disease International. (2013) World Alzheimer Report 2013: Journey of Caring.Google Scholar
3. Sperling, R.A. et al. (2011) Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging and the Alzheimer's Association workgroup. Alzheimer's & Dementia 7, 280-292 Google Scholar
4. Albert, M.S. et al. (2011) The diagnosis of mild cognitive impairment due to Alzheimer's disease: recommendations from the National Institute on Aging and Alzheimer's Association workgroup. Alzheimer's & Dementia 7, 270-279 doi:10.1016/j.jalz.2011.03.008 Google Scholar
5. McKhann, G.M. et al. (2011) The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's & Dementia 7, 263-269 Google Scholar
6. Yang, T. et al. (2015) A highly sensitive novel immunoassay specifically detects low levels of soluble Aβ oligomers in human cerebrospinal fluid. Alzheimer's Research and Therapy 7, 14 doi:10.1186/s13195-015-0100-y Google Scholar
7. Terry, R.D. et al. (1991) Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Annals of Neurology 30, 572-580 doi:10.1002/ana.410300410 Google Scholar
8. Benilova, I., Karran, E. and De Strooper, B. (2012) The toxic Aβ oligomer and Alzheimer's disease: an emperor in need of clothes. Nature Neuroscience 15, 349-357 CrossRefGoogle ScholarPubMed
9. Hardy, J. and Selkoe, D.J. (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297, 353-356 doi:10.1126/science.1072994 Google Scholar
10. Mawuenyega, K.G. et al. (2010) Decreased clearance of CNS β-amyloid in Alzheimer's disease. Science 330, 1774 doi:10.1126/science.1197623 Google Scholar
11. Bu, G. (2009) Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy. Nature Reviews Neuroscience 10, 333-344 doi:10.1038/nrn2620 Google Scholar
12. Liu, C.-C. et al. (2013) Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nature Reviews Neurology 9, 106-118 doi:10.1038/nrneurol.2012.263 Google Scholar
13. Kanekiyo, T., Xu, H. and Bu, G. (2014) ApoE and Aβ in Alzheimer's disease: accidental encounters or partners? Neuron 81, 740-754 doi:10.1016/j.neuron.2014.01.045 Google Scholar
14. Huang, Y. and Mucke, L. (2012) Alzheimer mechanisms and therapeutic strategies. Cell 148, 1204-1222 doi:10.1016/j.cell.2012.02.040 Google Scholar
15. Solomon, B. (2009) Immunotherapeutic strategies for Alzheimer's disease treatment. The Scientific World Journal 9, 909-919 Google Scholar
16. Karran, E., Mercken, M. and De Strooper, B. (2011) The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics. Nature Reviews Drug Discovery 10, 698-712 Google Scholar
17. Wolfe, M.S. et al. (1999) Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and γ-secretase activity. Nature 398, 513-517 doi:10.1038/19077 Google Scholar
18. Kojro, E. and Fahrenholz, F. (2005) The non-amyloidogenic pathway: structure and function of α-secretases. Subcellular Biochemistry 38, 105-127 Google Scholar
19. Haass, C. et al. (1993) Beta-Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms. Journal of Biological Chemistry 268, 3021-3024 Google Scholar
20. LaFerla, F.M., Green, K.N. and Oddo, S. (2007) Intracellular amyloid-β in Alzheimer's disease. Nature Reviews Neuroscience 8, 499-509 doi:10.1038/nrn2168 Google Scholar
21. Jarrett, J.T., Berger, E.P. and Lansbury, P.T. (1993) The carboxy terminus of the β amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease. Biochemistry 32, 4693-4697 Google Scholar
22. Younkin, S.G. (1998) The role of Aβ42 in Alzheimer's disease. Journal of Physiology – Paris 92, 289-292 Google Scholar
23. Yamada, M. (2015) Cerebral amyloid angiopathy: emerging concepts. Journal of Stroke 17, 17-30 doi:10.5853/jos.2015.17.1.17 Google Scholar
24. Bentahir, M. et al. (2006) Presenilin clinical mutations can affect γ-secretase activity by different mechanisms. Journal of Neurochemistry 96, 732-742 doi:10.1111/j.1471-4159.2005.03578.x Google Scholar
25. Wang, J. et al. (1999) The levels of soluble versus insoluble brain Aβ distinguish Alzheimer's disease from normal and pathologic aging. Experimental Neurology 158, 328-337 doi:10.1006/exnr.1999.7085 Google Scholar
26. Dubois, B. et al. (2016) Preclinical Alzheimer's disease: definition, natural history, and diagnostic criteria. Alzheimer's & Dementia 12, 292-323 doi:10.1016/j.jalz.2016.02.002 Google Scholar
27. Weksler, M.E. et al. (2002) Patients with Alzheimer disease have lower levels of serum anti-amyloid peptide antibodies than healthy elderly individuals. Experimental Gerontology 37, 943-948 Google Scholar
28. Spencer, B. and Masliah, E. (2014) Immunotherapy for Alzheimer's disease: past, present and future. Frontiers in Aging Neuroscience 6, 114 doi:10.3389/fnagi.2014.00114 Google Scholar
29. Li, Y. et al. (2013) Clinical trials of amyloid-based immunotherapy for Alzheimer's disease: end of beginning or beginning of end? Expert Opinion on Biological Therapy 13, 1515-1522 doi:10.1517/14712598.2013.838555 Google Scholar
30. Lemere, C.A. (2013) Immunotherapy for Alzheimer's disease: hoops and hurdles. Molecular Neurodegeneration 8, 36 Google Scholar
31. Lannfelt, L. et al. (2014) Perspectives on future Alzheimer therapies: amyloid-β protofibrils – a new target for immunotherapy with BAN2401 in Alzheimer's disease. Alzheimer's Research and Therapy 6, 16 doi:10.1186/alzrt246 Google Scholar
32. Gilman, S. et al. (2005) Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology 64, 1553-1562 Google Scholar
33. Sarazin, M. et al. (2013) Immunotherapy in Alzheimer's disease: do we have all the pieces of the puzzle? Biological Psychiatry 74, 329-332 doi:10.1016/j.biopsych.2013.04.011 Google Scholar
34. Schneeberger, A. et al. (2009) Development of AFFITOPE vaccines for Alzheimer's disease (AD) – from concept to clinical testing. Journal of Nutrition Health and Aging 13, 264-267 Google Scholar
35. Schneeberger, A. et al. (2015) 12th International Conference on Alzheimer's and Parkinson's Diseases and Related Neurological Disorders, Nice, FranceGoogle Scholar
36. Winblad, B. et al. (2012) Safety, tolerability, and antibody response of active Aβ immunotherapy with CAD106 in patients with Alzheimer's disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurology 11, 597-604 doi:10.1016/S1474-4422(12)70140-0 Google Scholar
37. Kingwell, K. (2012) Alzheimer disease: amyloid-β immunotherapy CAD106 passes first safety test in patients with Alzheimer disease. Nature Reviews Neurology 8, 414 doi:10.1038/nrneurol.2012.128 Google Scholar
38. Lannfelt, L., Relkin, N.R. and Siemers, E.R. (2014) Amyloid-ß-directed immunotherapy for Alzheimer's disease. Journal of Internal Medicine 275, 284-295 doi:10.1111/joim.12168 Google Scholar
39. Arai, H., Suzuki, H. and Yoshiyama, T. (2015) Vanutide Cridificar and the QS-21 adjuvant in Japanese subjects with mild to moderate Alzheimer's disease: results from two phase 2 studies. Current Alzheimer Research 12, 242-254 Google Scholar
40. Wang, Y.J., Zhou, H.D. and Zhou, X.F. (2010) Modified immunotherapies against Alzheimer's disease: toward safer and effective amyloid-β clearance. Journal of Alzheimer's Disease 21, 1065-1075 Google Scholar
41. Holliger, P. and Hudson, P.J. (2005) Engineered antibody fragments and the rise of single domains. Nature Biotechnology 23, 1126-1136 Google Scholar
42. Waldmann, T.A. (2003) Immunotherapy: past, present and future. Nature Medicine 9, 269-277 doi:10.1038/nm0303-269 Google Scholar
43. Bard, F. et al. (2000) Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nature Medicine 6, 916-919 Google Scholar
44. Salloway, S. et al. (2014) Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. New England Journal of Medicine 370, 322-333 doi:10.1056/NEJMoa1304839 Google Scholar
45. Sperling, R.A. et al. (2011) Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer's Association Research Roundtable Workgroup. Alzheimer's & Dementia 7, 367-385 doi:10.1016/j.jalz.2011.05.2351 Google Scholar
46. Black, R. et al. (2009) Immunotherapy regimes dependent on ApoE status (US Patent) US 20090155256 A1Google Scholar
47. DeMattos, R.B. et al. (2001) Peripheral anti-A beta antibody alters CNS and plasma Aβ clearance and decreases brain Aβburden in a mouse model of Alzheimer's disease. Proceedings of the National Academy of Sciences of the United States of America 98, 8850-8855 doi:10.1073/pnas.151261398 Google Scholar
48. Doody, R.S. et al. (2014) Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. New England Journal of Medicine 370, 311-321 doi:10.1056/NEJMoa1312889 Google Scholar
49. Samadi, H. and Sultzer, D. (2011) Solanezumab for Alzheimer's disease. Expert Opinion on Biological Therapy 11, 787-798 doi:10.1517/14712598.2011.578573 Google Scholar
50. Fleisher, A.S. et al. (2015) Associations between biomarkers and age in the presenilin 1 E280A autosomal dominant Alzheimer disease kindred: a cross-sectional study. JAMA Neurology 72, 316-324 doi:10.1001/jamaneurol.2014.3314 Google Scholar
51. Bohrmann, B. et al. (2012) Gantenerumab: a novel human anti-Aβ antibody demonstrates sustained cerebral amyloid-β binding and elicits cell-mediated removal of human amyloid-β . Journal of Alzheimer's Diseases 28, 49-69 doi:10.3233/JAD-2011-110977 Google Scholar
52. Ostrowitzki, S. et al. (2012) Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Archives of Neurology 69, 198-207 doi:10.1001/archneurol.2011.1538 Google Scholar
53. Adolfsson, O. et al. (2012) An effector-reduced anti-β-amyloid (Aβ) antibody with unique Aβ binding properties promotes neuroprotection and glial engulfment of Aβ . Journal of Neuroscience 32, 9677-9689 doi:10.1523/JNEUROSCI.4742-11.2012 Google Scholar
54. Crespi, G.A.N. et al. (2015) Molecular basis for mid-region amyloid-β capture by leading Alzheimer's disease immunotherapies. Science Report 5, 9649 doi:10.1038/srep09649 Google Scholar
55. La Porte, S.L. et al. (2012) Structural basis of C-terminal β-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer's disease. Journal of Molecular Biology 421, 525-536 doi:10.1016/j.jmb.2011.11.047 Google Scholar
56. Tucker, S. et al. (2015) The murine version of BAN2401 (mAb158) selectively reduces amyloid-β protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. Journal of Alzheimer's Disease 43, 575-588 doi:10.3233/JAD-140741 Google Scholar
57. Lannfelt, L. et al. (2014) Perspectives on future Alzheimer therapies: amyloid-β protofibrils - a new target for immunotherapy with BAN2401 in Alzheimer's disease. Alzheimer's Research and Therapy 6, 16 doi:10.1186/alzrt246 Google Scholar
58. Popugaeva, E. and Bezprozvanny, I. (2014) Can the calcium hypothesis explain synaptic loss in Alzheimer's disease? Neurodegenerative Disease 13, 139-141 doi:10.1159/000354778 Google Scholar
59. Sevigny, J. et al. (2015) 12th International Conference on Alzheimer's and Parkinson's Diseases and Related Neurological Disorders, Nice, FranceGoogle Scholar
60. Underwood, E. (2015) NEUROSCIENCE. Alzheimer's amyloid theory gets modest boost. Science 349, 464 doi:10.1126/science.349.6247.464 Google Scholar
61. Reardon, S. (2015) Antibody drugs for Alzheimer's show glimmers of promise. Nature 523, 509-510 doi:10.1038/nature.2015.18031 CrossRefGoogle ScholarPubMed
62. Loeffler, D.A. (2013) Intravenous immunoglobulin and Alzheimer's disease: what now? Journal of Neuroinflammation 10, 70 Google Scholar
63. Dodel, R. et al. (2002) Human antibodies against amyloid β peptide: a potential treatment for Alzheimer's disease. Annals of Neurology 52, 253-256 Google Scholar
64. Weksler, M.E., Pawelec, G. and Franceschi, C. (2009) Immune therapy for age-related diseases. Trends in Immunology 30, 344-350 Google Scholar
65. Dodel, R. et al. (2011) Naturally occurring autoantibodies against β-amyloid: investigating their role in transgenic animal and in vitro models of Alzheimer's disease. Journal of Neuroscience 31, 5847-5854 doi:10.1523/JNEUROSCI.4401-10.2011 Google Scholar
66. Istrin, G., Bosis, E. and Solomon, B. (2006) Intravenous immunoglobulin enhances the clearance of fibrillar amyloid-β peptide. Journal of Neuroscience Research 84, 434-443 Google Scholar
67. Szabo, P., Relkin, N. and Weksler, M.E. (2008) Natural human antibodies to amyloid beta peptide. Autoimmunity Reviews 7, 415-420 Google Scholar
68. Klaver, A.C. et al. (2010) Antibody concentrations to Aβ1-42 monomer and soluble oligomers in untreated and antibody-antigen-dissociated intravenous immunoglobulin preparations. International Immunopharmacology 10, 115-119 Google Scholar
69. Boada, M. et al. (2009) Amyloid-targeted therapeutics in Alzheimer's disease: use of human albumin in plasma exchange as a novel approach for Aβ mobilization. Drug News & Perspectives 22, 325 doi:10.1358/dnp.2009.22.6.1395256 Google Scholar
70. Dodel, R. et al. (2010) Intravenous immunoglobulins as a treatment for Alzheimer's disease: rationale and current evidence. Drugs 70, 513-528 Google Scholar
71. Magga, J. et al. (2010) Human intravenous immunoglobulin provides protection against Aβ toxicity by multiple mechanisms in a mouse model of Alzheimer's disease. Journal of Neuroinflammation 7, 90 CrossRefGoogle Scholar
72. MacHimoto, T. et al. (2010) Effect of IVIG administration on complement activation and HLA antibody levels: ORIGINAL ARTICLE. Transplant International 23, 1015-1022 Google Scholar
73. Abe, Y. et al. (1994) Anti-cytokine nature of natural human immunoglobulin: one possible mechanism of the clinical effect of intravenous immunoglobulin therapy. Immunological Reviews 139, 5-19 Google Scholar
74. Pigard, N. et al. (2009) Therapeutic activities of intravenous immunoglobulins in multiple sclerosis involve modulation of chemokine expression. Journal of Neuroimmunology 209, 114-120 CrossRefGoogle ScholarPubMed
75. Kessel, A. et al. (2007) Intravenous immunoglobulin therapy affects T regulatory cells by increasing their suppressive function. Journal of Immunology 179, 5571-5575 Google Scholar
76. Schmidt, A.M. et al. (2009) The role of RAGE in amyloid-β peptide-mediated pathology in Alzheimer's disease. Current Opinion on Investigational Drugs 10, 672-680 Google Scholar
77. Deane, R. et al. (2003) RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain. Nature Medicine 9, 907-913 Google Scholar
78. Shibata, M. et al. (2000) Clearance of Alzheimer's amyloid-β1-40 peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. Journal of Clinical Investigation 106, 1489-1499 Google Scholar
79. Deane, R., Sagare, A. and Zlokovic, B.V. (2008) The role of the cell surface LRP and soluble LRP in blood–brain barrier Aβ clearance in Alzheimer's disease. Current Pharmaceutical Design 14, 1601-1605 Google Scholar
80. Sagare, A. et al. (2007) Clearance of amyloid-beta by circulating lipoprotein receptors. Nature Medicine 13, 1029-1031 Google Scholar
81. Tamura, Y. et al. (2005) The F(ab)'2 fragment of an Aβ-specific monoclonal antibody reduces Abeta deposits in the brain. Neurobiology of Disease 20, 541-549 doi:10.1016/j.nbd.2005.04.007 CrossRefGoogle Scholar
82. Robert, R. and Wark, K.L. (2012) Engineered antibody approaches for Alzheimer's disease immunotherapy. Archives of Biochemistry and Biophysics 526, 132-138 Google Scholar
83. Liu, R. et al. (2004) Single chain variable fragments against β-amyloid (Abeta) can inhibit Abeta aggregation and prevent abeta-induced neurotoxicity. Biochemistry 43, 6959-6967 doi:10.1021/bi049933o Google Scholar
84. Frenkel, D., Solomon, B. and Benhar, I. (2000) Modulation of Alzheimer's β-amyloid neurotoxicity by site-directed single-chain antibody. Journal of Neuroimmunology 106, 23-31 Google Scholar
85. Lafaye, P. et al. (2009) Single-domain antibodies recognize selectively small oligomeric forms of amyloid β, prevent Aβ-induced neurotoxicity and inhibit fibril formation. Molecular Immunology 46, 695-704 Google Scholar
86. Robert, R. et al. (2009) Engineered antibody intervention strategies for Alzheimer's disease and related dementias by targeting amyloid and toxic oligomers. Protein Engineering Design and Selection 22, 199-208 Google Scholar
87. Habicht, G. et al. (2007) Directed selection of a conformational antibody domain that prevents mature amyloid fibril formation by stabilizing Aβ protofibrils. Proceedings of the National Academy of Sciences of the United States of America 104, 19232-19237 Google Scholar
88. Meli, G. et al. (2009) Direct in vivo intracellular selection of conformation-sensitive antibody domains targeting Alzheimer's Amyloid-β oligomers. Journal of Molecular Biology 387, 584-606 Google Scholar
89. Paganetti, P. et al. (2005) β-site specific intrabodies to decrease and prevent generation of Alzheimer's Aβ peptide. Journal of Cell Biology 168, 863-868 Google Scholar
90. Rangan, S.K. et al. (2003) Degradation of β-amyloid by proteolytic antibody light chains. Biochemistry 42, 14328-14334 doi:10.1021/bi035038d Google Scholar
91. Taguchi, H. et al. (2008) Exceptional amyloid β peptide hydrolyzing activity of nonphysiological immunoglobulin variable domain scaffolds. Journal of Biological Chemistry 283, 36724-36733 Google Scholar
92. Taguchi, H. et al. (2008) Catalytic antibodies to amyloid β peptide in defense against Alzheimer disease. Autoimmunity Reviews 7, 391-397 Google Scholar
93. Kasturirangan, S. and Sierks, M. (2010) Targeted hydrolysis of β-amyloid with engineered antibody fragment. Current Alzheimer Research 7, 214-222 Google Scholar
94. Giménez-Llort, L. et al. (2013) Early intervention in the 3xTg-AD mice with an amyloid β-antibody fragment ameliorates first hallmarks of Alzheimer disease. MAbs 5, 665-677 Google Scholar
95. Esquerda-Canals, G. et al. (2013) Loss of deep cerebellar nuclei neurons in the 3xTg-AD mice and protection by an anti-amyloid β antibody fragment. MAbs 5, 660-664 doi:10.4161/mabs.25428 Google Scholar
96. Kim, D.Y. et al. (2014) Mutational approaches to improve the biophysical properties of human single-domain antibodies. Biochimica et Biophysica Acta – Proteins and Proteomics 1844, 1983-2001 CrossRefGoogle ScholarPubMed
97. Hamers-Casterman, C. et al. (1993) Naturally occurring antibodies devoid of light chains. Nature 363, 446-448 Google Scholar
98. Muyldermans, S., Cambillau, C. and Wyns, L. (2001) Recognition of antigens by single-domain antibody fragments: the superfluous luxury of paired domains. Trends in Biochemical Sciences 26, 230-235 Google Scholar
99. Paraschiv, G. et al. (2013) Epitope structure and binding affinity of single chain llama anti-β-amyloid antibodies revealed by proteolytic excision affinity-mass spectrometry. Journal of Molecular Recognition 26, 1-9 Google Scholar
100. Hanenberg, M. et al. (2014) Amyloid-β peptide-specific DARPins as a novel class of potential therapeutics for Alzheimer disease. Journal of Biological Chemistry 289, 27080-27089 doi:10.1074/jbc.M114.564013 Google Scholar
101. Binz, H.K. et al. (2004) High-affinity binders selected from designed ankyrin repeat protein libraries. Nature Biotechnology 22, 575-582 doi:10.1038/nbt962 Google Scholar
102. Binz, H.K. et al. (2003) Designing repeat proteins: well-expressed, soluble and stable proteins from combinatorial libraries of consensus ankyrin repeat proteins. Journal of Molecular Biology 332, 489-503 Google Scholar
103. Stumpp, M.T., Binz, H.K. and Amstutz, P. (2008) DARPins: a new generation of protein therapeutics. Drug Discovery Today 13, 695-701 doi:10.1016/j.drudis.2008.04.013 Google Scholar
104. Amstutz, P. et al. (2005) Intracellular kinase inhibitors selected from combinatorial libraries of designed ankyrin repeat proteins. Journal of Biological Chemistry 280, 24715-24722 doi:10.1074/jbc.M501746200 Google Scholar
105. Kohl, A. et al. (2005) Allosteric inhibition of aminoglycoside phosphotransferase by a designed ankyrin repeat protein. Structure 13, 1131-1141 doi:10.1016/j.str.2005.04.020 Google Scholar
106. Zahnd, C. et al. (2010) Efficient tumor targeting with high-affinity designed ankyrin repeat proteins: effects of affinity and molecular size. Cancer Research 70, 1595-1605 doi:10.1158/0008-5472.CAN-09-2724 Google Scholar
107. Pike, C.J. et al. (1991) In vitro aging of β-amyloid protein causes peptide aggregation and neurotoxicity. Brain Research 563, 311-314 Google Scholar
108. Busciglio, J., Lorenzo, A. and Yankner, B.A. (1992) Methodological variables in the assessment of βamyloid neurotoxicity. Neurobiology of Aging 13, 609-612 Google Scholar
109. Cheng, I.H. et al. (2007) Accelerating amyloid-β fibrillization reduces oligomer levels and functional deficits in Alzheimer disease mouse models. Journal of Biological Chemistry 282, 23818-23828 doi:10.1074/jbc.M701078200 Google Scholar
110. Walsh, D.M. and Selkoe, D.J. (2007) Aβ oligomers – a decade of discovery. Journal of Neurochemistry 101, 1172-1184 doi:10.1111/j.1471-4159.2006.04426.x Google Scholar
111. Horikoshi, Y. et al. (2004) Aβ N-terminal-end specific antibody reduced β-amyloid in Alzheimer-model mice. Biochemistry and Biophysical Research Communications 325, 384-387 Google Scholar
112. Bussière, T. et al. (2004) Morphological characterization of Thioflavin-S-positive amyloid plaques in transgenic Alzheimer mice and effect of passive Aβimmunotherapy on their clearance. American Journal of Pathology 165, 987-995 Google Scholar
113. Lu, J.-X. et al. (2013) Molecular structure of β-amyloid fibrils in Alzheimer's disease brain tissue. Cell 154, 1257-1268 doi:10.1016/j.cell.2013.08.035 Google Scholar
114. Schenk, D. et al. (1999) Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400, 173-177 doi:10.1038/22124 Google Scholar
115. Wilcock, D.M. et al. (2004) Passive amyloid immunotherapy clears amyloid and transiently activates microglia in a transgenic mouse model of amyloid deposition. Journal of Neuroscience 24, 6144-6151 doi:10.1523/JNEUROSCI.1090-04.2004 Google Scholar
116. Schlachetzki, F., Zhu, C. and Pardridge, W.M. (2002) Expression of the neonatal Fc receptor (FcRn) at the blood–brain barrier. Journal of Neurochemistry 81, 203-206 Google Scholar
117. Bacskai, B.J. et al. (2002) Non-Fc-mediated mechanisms are involved in clearance of amyloid-β in vivo by immunotherapy. Journal of Neuroscience 22, 7873-7878 Google Scholar
118. Das, P. et al. (2003) Amyloid-β immunization effectively reduces amyloid deposition in FcR gamma-/- knock-out mice. Journal of Neuroscience 23, 8532-8538 Google Scholar
119. Kou, J. et al. (2011) Anti-amyloid-β single-chain antibody brain delivery via aav reduces amyloid load but may increase cerebral hemorrhages in an Alzheimer's disease mouse model. Journal of Alzheimer's Disease 27, 23-28 Google Scholar
120. Wang, Y.J. et al. (2009) Intramuscular delivery of a single chain antibody gene reduces brain Aβ burden in a mouse model of Alzheimer's disease. Neurobiology of Aging 30, 364-376 Google Scholar
121. Boado, R.J. et al. (2013) Pharmacokinetics and brain uptake in the rhesus monkey of a fusion protein of arylsulfatase a and a monoclonal antibody against the human insulin receptor. Biotechnology and Bioengineering 110, 1456-1465 doi:10.1002/bit.24795 Google Scholar
122. Kissel, K. et al. (1998) Immunohistochemical localization of the murine transferrin receptor (TfR) on blood-tissue barriers using a novel anti-TfR monoclonal antibody. Histochemistry and Cell Biology 110, 63-72 CrossRefGoogle ScholarPubMed
123. Jefferies, W.A. et al. (1984) Transferrin receptor on endothelium of brain capillaries. Nature 312, 162-163 CrossRefGoogle ScholarPubMed
124. Fishman, J.B. et al. (1987) Receptor-mediated transcytosis of transferrin across the blood–brain barrier. Journal of Neuroscience Research 18, 299-304 Google Scholar
125. Roberts, R.L., Fine, R.E. and Sandra, A. (1993) Receptor-mediated endocytosis of transferrin at the blood–brain barrier. Journal of Cell Science 104(Pt 2), 521-532 Google Scholar
126. Dufes, C., Al Robaian, M. and Somani, S. (2013) Transferrin and the transferrin receptor for the targeted delivery of therapeutic agents to the brain and cancer cells. Therpeutic Delivery 4(2041-5990 (Print)), 629-640 doi:10.4155/tde.13.21 Google Scholar
127. Yu, Y.J. and Watts, R.J. (2013) Developing therapeutic antibodies for neurodegenerative disease. Neurotherapeutics 10, 459-472 Google Scholar
128. Manich, G. et al. (2013) Study of the transcytosis of an anti-transferrin receptor antibody with a Fab′ cargo across the blood-brain barrier in mice. European Journal of Pharmaceutical Sciences 49, 556-564 Google Scholar
129. Niewoehner, J. et al. (2014) Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron 81, 49-60 Google Scholar
130. Yu, Y.J. et al. (2011) Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Science Translational Medicine 3, 84ra44 CrossRefGoogle ScholarPubMed
131. Shimada, M. et al. (2013) Prophylaxis and treatment of Alzheimer's disease by delivery of an Adeno-Associated virus encoding a monoclonal antibody targeting the amyloid β protein. PLoS ONE 8, e57606 Google Scholar
132. Sudol, K.L. et al. (2009) Generating differentially targeted amyloid-β specific intrabodies as a passive vaccination strategy for Alzheimer's disease. Molecular Therapy 17, 2031-2040 Google Scholar