We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Bapineuzumab: anti-β-amyloid monoclonal antibodies for the treatment of Alzheimer’s disease

    ,
    Vincenza Frisardi*

    Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, Bari, Italy

    *Both authors contributed equally

    Search for more papers by this author

    ,
    Bruno P Imbimbo

    Research & Development Department, Chiesi Farmaceutici, Parma, Italy

    ,
    Grazia D’Onofrio

    Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy

    ,
    Giuseppe Pietrarossa

    Department of Neurology, Di Venere Hospital, Bari, Italy

    ,
    Davide Seripa

    Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy

    ,
    Alberto Pilotto

    Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy

    &
    Vincenzo Solfrizzi

    Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, Bari, Italy

    Published Online:https://doi.org/10.2217/imt.10.80

    In the last decade, new therapeutic approaches targeting β-amyloid (Aβ) have been discovered and developed with the hope of modifying the natural history of Alzheimer’s disease (AD). The most revolutionary of these approaches consists in the removal of brain Aβ via anti-Aβ antibodies. After an active vaccine (AN1792) was discontinued in 2002 due to occurrence of meningoencephalitis in approximately 6% of patients, several other second-generation active Aβ vaccines and passive Aβ immunotherapies have been developed and are under clinical investigation with the aim of accelerating Aβ clearance from the brain of AD patients. The most advanced of these immunological approaches is bapineuzumab, which is composed of humanized anti-Aβ monoclonal antibodies that has been tested in two Phase II trials. Bapineuzumab has been shown to reduce Aβ burden in the brain of AD patients. However, its preliminary cognitive efficacy appears uncertain, particularly in ApoE ε4 carriers, and vasogenic edema may limit its clinical use. The results of four ongoing large Phase III trials on bapineuzumab will provide answers regarding whether passive anti-Aβ immunization is able to alter the course of this devastating disease.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Qiu C, De Ronchi D, Fratiglioni L: The epidemiology of the dementias: an update. Curr. Opin. Psychiatry20,380–385 (2007).
    • Alzheimer ’s Association: Alzheimer’s disease facts and figures. Alzheimers Dement.6(2),158–194 (2010).
    • Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM: Forecasting the global burden of Alzheimer’s disease. Alzheimers Dement.3(3),186–191 (2007).
    • Panza F, Solfrizzi V, Frisardi V et al.: Disease-modifying approach to the treatment of Alzheimer’s disease: from α-secretase activators to γ-secretase inhibitors and modulators. Drugs Aging26(7),537–555 (2009).
    • Panza F, Solfrizzi V, Frisardi V et al.: Beyond the neurotransmitter-focused approach in treating Alzheimer’s disease: drugs targeting β-amyloid and tau protein. Aging Clin. Exp. Res.21(6),386–406 (2009).
    • Frisardi V, Solfrizzi V, Imbimbo PB et al.: Towards disease-modifying treatment of Alzheimer’s disease: drugs targeting β-amyloid. Curr. Alzheimer Res.7(1),40–55 (2010).
    • Sabbagh MN: Drug development for Alzheimer’s disease: where are we now and where we are headed? Am. J. Geriatr. Pharmacother.7(3),167–185 (2010).
    • Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M: Alzheimer’s disease: clinical trials and drug development. Lancet Neurol.9(7),702–716 (2010).
    • Citron M: Alzheimer’s disease: strategies for disease modification. Nat. Rev. Drug Discov.9(5),387–398 (2010).
    • 10  Walter J, Kaether C, Steiner H, Haass C: The cell biology of Alzheimer’s disease: uncovering the secrets of secretases. Curr. Opin. Neurobiol.11(5),585–590 (2001).
    • 11  Hardy J, Allsop D: Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci.12(5),383–388 (1991).
    • 12  Wisniewski KE, Wisniewski HM, Wen GY: Occurrence of neuropathological changes and dementia of Alzheimer’s disease in Down’s syndrome. Ann. Neurol.17(3),278–282 (1985).
    • 13  Kawahara M: Neurotoxicity of β-amyloid protein: oligomerization, channel formation, and calcium dyshomeostasis. Curr. Pharm. Des.16(25),2779–2789 (2010).
    • 14  Brion JP, Ando K, Heraud C, Leroy K: Modulation of tau pathology in tau transgenic models. Biochem. Soc. Trans.38(4),996–1000 (2010).
    • 15  McDonald JM, Savva GM, Brayne C et al.: Medical research council cognitive function and ageing study; the presence of sodium dodecyl sulphate-stable Aβ dimers is strongly associated with Alzheimer-type dementia. Brain133(Pt 5),1328–1341 (2010).
    • 16  Krafft GA, Klein WL: ADDLs and the signaling web that leads to Alzheimer’s disease. Neuropharmacology59(4–5),230–242 (2010).
    • 17  Castellani RJ, Lee HG, Zhu X, Perry G, Smith MA: Alzheimer disease pathology as a host response. J. Neuropathol. Exp. Neurol.67(6),523–531 (2008).
    • 18  Scheinin NM, Aalto S, Koikkalainen J et al.: Follow-up of [11C]PIB uptake and brain volume in patients with Alzheimer disease and controls. Neurology73(15),1186–1192 (2009).
    • 19  Knopman DS, Parisi JE, Salviati A et al.: Neuropathology of cognitively normal elderly. J. Neuropathol. Exp. Neurol.62(11),1087–1095 (2003).
    • 20  Morris JC, Roe CM, Grant EA et al.: Pittsburgh compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch. Neurol.66(12),1469–1475 (2009).
    • 21  Vickers JC, Chin D, Edwards AM, Sampson V, Harper C, Morrison J: Dystrophic neurite formation associated with age-related β-amyloid deposition in the neocortex: clues to the genesis of neurofibrillary pathology. Exp. Neurol.141(1),1–11 (1996).
    • 22  Imbimbo BP, Solfrizzi V, Panza F: Are NSAIDs useful to treat Alzheimer’s disease or mild cognitive impairment? Front. Aging Neurosci.2,19 (2010).
    • 23  Hüll M, Berger M, Heneka M: Disease-modifying therapies in Alzheimer’s disease: how far have we come? Drugs66(16),2075–2093 (2006).
    • 24  Panza F, D’Introno A, Colacicco AM et al.: Lipid metabolism in cognitive decline and dementia. Brain Res. Rev.51(2),275–292 (2006).
    • 25  Martenyi F, Lowe S, Dean RA et al.: Central and peripheral pharmacokinetics and pharmacodynamic effects of the β-site APP cleavage enzyme (BACE1) inhibitor LY2811376 in humans. Presented at: ICAD 2010. Honolulu, HI, USA, 10–15 July 2010.
    • 26  Panza F, Frisardi V, Imbimbo BP, Capurso C et al.: γ-secretase inhibitors for the treatment of Alzheimer’s disease: the current state. CNS Neurosci. Ther. DOI: 10.1111/j.1755–5949.2010.00164.x (2010) (Epub ahead of print).
    • 27  Faux NG, Ritchie CW, Gunn A et al.: PBT2 rapidly improves cognition in Alzheimer’s disease: additional Phase II analyses. J. Alzheimers Dis.20(2),509–516 (2010).▪ Post-hoc analysis of the Phase II trial examining PBT2, confirming the positive cognitive effects of this zinc/copper ionophore in Alzheimer’s disease (AD) patients.
    • 28  Wisniewski T, Konietzko U: amyloid-β immunisation for Alzheimer’s disease. Lancet Neurol.7(9),805–811 (2008).▪ Comprehensive review discussing β-amyloid (Aβ) immunization in AD, with a special focus on possible mechanisms of Aβ-directed immunomodulation.
    • 29  Cribbs DH: Aβ DNA vaccination for Alzheimer’s disease: focus on disease prevention. CNS Neurol. Disord. Drug Targets9(2),207–216 (2010).
    • 30  Solomon B, Koppel R, Hanan E, Katzav T: Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer β-amyloid peptide. Proc. Natl Acad. Sci. USA93(1),452–455 (1996).▪ To the best of our knowledge, this was the first report in which it was found that monoclonal antibodies raised against Aβ peptide inhibited its aggregation.
    • 31  Schenk D, Barbour R, Dunn W et al.: Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature400(6740),173–177 (1999).▪ First description of Aβ vaccination and plaque removal in amyloid precursor protein transgenic mice, an animal model of AD.
    • 32  Kennedy GJ, Golde TE, Tariot PN, Cummings JL: amyloid-based interventions in Alzheimer’s disease. CNS Spectr.12(12 Suppl. 1),1–14 (2007).
    • 33  Bacskai BJ, Kajdasz ST, Christie RH et al.: Imaging of amyloid-β deposits in brains of living mice permits direct observation of clearance of plaques with immunotherapy. Nat. Med.7(3),369–372 (2001).
    • 34  Solomon B, Koppel R, Hanan E, Katzav T: Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer β-amyloid peptide. Proc. Natl Acad. Sci. USA93(1),452–455 (1996).
    • 35  Bard F, Cannon C, Barbour R et al.: Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat. Med.6(8),916–919 (2000).
    • 36  Bacskai BJ, Kajdasz ST, McLellan ME et al.: Non-Fc-mediated mechanisms are involved in clearance of amyloid-β in vivo by immunotherapy. J. Neurosci.22(18),7873–7878 (2002).
    • 37  Das P, Howard V, Loosbrock N, Dickson D, Murphy MP, Golde TE: amyloid-β immunization effectively reduces amyloid deposition in FcRg-/- knock-out mice. J. Neurosci.23(24),8532–8538 (2003).
    • 38  DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM: Peripheral anti-Aβ antibody alters CNS and plasma Aβ clearance and decreases brain Aβ burden in a mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA98,8850–8855 (2001).▪ To the best of our knowledge, this was the first study showing that antibodies could prevent Aβ deposition with the creation of a peripheral sink effect, in which the removal of excess circulatory soluble Aβ draws soluble Aβ from the brain.
    • 39  Sigurdsson EM, Scholtzova H, Mehta P, Frangione B, Wisniewski T: Immunization with a non-toxic/non-fibrillar amyloid-β homologous peptide reduces Alzheimer’s disease associated pathology in transgenic mice. Am. J. Pathol.159(2),439–447 (2001).
    • 40  Sigurdsson EM, Knudsen E, Asuni A et al.: An attenuated immune response is sufficient to enhance cognition in an Alzheimer’s disease mouse model immunized with amyloid-β derivatives. J. Neurosci.24(28),6277–6282 (2004).
    • 41  Brody DL, Holtzman DM: Active and passive immunotherapy for neurodegenerative disorders. Annu. Rev. Neurosci.31,175–193 (2008).
    • 42  Bayer AJ, Bullock R, Jones RW et al.: Evaluation of the safety and immunogenicity of synthetic Aβ42 (AN1792) in patients with AD. Neurology64(1),94–101 (2005).▪▪ Phase I trial of AN1792 on 80 mild-to-moderate AD patients, and the first published account of Aβ immunization in humans.
    • 43  Gilman S, Koller M, Black RS et al.: Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology64(9),1553–1562 (2005).▪▪ Phase II trial of AN1792, interrupted for the development of meningoencephalitis in 6% of AD patients.
    • 44  Robinson SR, Bishop GM, Lee HG, Munch G: Lessons from the AN1792 Alzheimer vaccine: lest we forget. Neurobiol. Aging25(5),609–615 (2004).
    • 45  Pride M, Seubert P, Grundman M, Hagen M, Eldridge J, Black RS: Progress in the active immunotherapeutic approach to Alzheimer’s disease: clinical investigations into AN1792-associated meningoencephalitis. Neurodegener. Dis.5(3–4),194–196 (2008).
    • 46  Bombois S, Maurage CA, Gompel M et al.: Absence of β-amyloid deposits after immunization in Alzheimer disease with Lewy body dementia. Arch. Neurol.64(4),583–587 (2007).
    • 47  Ferrer I, Boada Rovira M, Sánchez Guerra ML, Rey MJ, Costa-Jussá F: Neuropathology and pathogenesis of encephalitis following amyloid-β immunization in Alzheimer’s disease. Brain Pathol.14(1),11–20 (2004).▪ Helped to define the pathology of meningoencephalitis in AN1792-treated patients.
    • 48  Masliah E, Hansen L, Adame A et al.: Aβ vaccination effects on plaque pathology in the absence of encephalitis in Alzheimer disease. Neurology64(1),129–131 (2005).▪▪ To the best of our knowledge, this was the first report of brain pathology in an AN1792-treated patient who did not develop meningoencephalitis, suggesting that Aβ clearance could occur in the absence of a symptomatic neuroinflammatory reaction.
    • 49  Hock C, Konietzko U, Streffer JR et al.: Antibodies against β-amyloid slow cognitive decline in Alzheimer’s disease. Neuron38(4),547–554 (2003).
    • 50  Holmes C, Boche D, Wilkinson D et al.: Long-term effects of Aβ42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled Phase I trial. Lancet372(9634),216–223 (2008).▪▪ This case series demonstrated that the burden of Aβ correlated inversely with antibody titer. Although this was not the first postmortem study of an AN1792-treated patient, it strongly suggested that Aβ immunization facilitated plaque clearance in humans; however, doing so had no effect on survival or progression of dementia.
    • 51  Frautschy SA, Baird A, Cole GM: Effects of injected Alzheimer β-amyloid cores in rat brain. Proc. Natl Acad. Sci. USA88(19),8362–8366 (1991).
    • 52  Sigurdsson EM, Lee JM, Dong XW, Hejna MJ, Lorens SA: Bilateral injections of amyloid-β 25–35 into the amygdala of young Fischer rats: behavioral, neurochemical, and time dependent histopathological effects. Neurobiol. Aging18(6),591–608 (1997).
    • 53  Götz J, Chen F, van Dorpe J, Nitsch RM: Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Aβ42 fibrils. Science293(5534),1491–1495 (2001).
    • 54  Oddo S, Billings L, Kesslak JP, Cribbs DH, LaFerla FM: Aβ immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron43(3),321–332 (2004).
    • 55  Boche D, Donald J, Love S, Neal JW, Holmes C, Nicoll J: Aβ42 immunisation reduces both tau and Aβ deposits in Alzheimer’s disease. Presented at: ICAD 2010. Honolulu, HI, USA, 10–15 July 2010.
    • 56  Lemere CA, Masliah E: Can Alzheimer disease be prevented by amyloid-β immunotherapy? Nat. Rev. Neurol.6(2),108–119 (2010).▪ Influential review article that provided an update on the active and passive Aβ immunotherapies currently in clinical trials.
    • 57  Tabira T: Immunization therapy for Alzheimer disease: a comprehensive review of active immunization strategies. Tohoku J. Exp. Med.220(2),95–106 (2010).
    • 58  Muhs A, Hickman DT, Pihlgren M et al.: Liposomal vaccines with conformation-specific amyloid peptide antigens define immune response and efficacy in APP transgenic mice. Proc. Natl Acad. Sci. USA104(23),9810–9815 (2007).
    • 59  Frenkel D, Dewachter I, Van Leuven F, Solomon B: Reduction of β-amyloid plaques in brain of transgenic mouse model of Alzheimer’s disease by EFRH-phage immunization. Vaccine21(11–12),1060–1065 (2003).
    • 60  Agadjanyan MG, Ghochikyan A, Petrushina I et al.: Prototype Alzheimer’s disease vaccine using the immunodominant B cell epitope from β-amyloid and promiscuous T cell epitope pan HLA DR-binding peptide. J. Immunol.174(3),1580–1586 (2005).
    • 61  Petrushina I, Ghochikyan A, Mktrichyan M et al.: Alzheimer’s disease peptide epitope vaccine reduces insoluble but not soluble/oligomeric Aβ species in amyloid precursor protein transgenic mice. J. Neurosci.27(46),12721–12731 (2007).
    • 62  Nikolic WV, Bai Y, Obregon D et al.: Transcutaneous β-amyloid immunization reduces cerebral β-amyloid deposits without T cell infiltration and microhemorrhage. Proc. Natl Acad. Sci. USA104(7),2507–2512 (2007).
    • 63  Bowers WJ, Mastrangelo MA, Stanley HA, Casey AE, Milo LJ Jr, Federoff HJ: HSV amplicon-mediated Aβ vaccination in Tg2576 mice: differential antigenspecific immune responses. Neurobiol. Aging26(4),393–407 (2005).
    • 64  Youm JW, Kim H, Han JH et al.: Transgenic potato expressing Aβ reduce Aβ burden in Alzheimer’s disease mouse model. FEBS Lett.579(30),6737–6744 (2005).
    • 65  Youm JW, Jeon JH, Kim H et al.: Transgenic tomatoes expressing human β-amyloid for use as a vaccine against Alzheimer’s disease. Biotechnol. Lett.30(10),1839–1645 (2008).
    • 66  Okura Y, Miyakoshi A, Kohyama K, Park IK, Staufenbiel M, Matsumoto Y: Nonviral Aβ DNA vaccine therapy against Alzheimer’s disease: long-term effects and safety. Proc. Natl Acad. Sci. USA103(25),9619–9624 (2006).
    • 67  Movsesyan N, Ghochikyan A, Mkrtichyan M et al.: Reducing AD-like pathology in 3xTg-AD mouse model by DNA epitope vaccine – a novel immunotherapeutic strategy. PLoS One3,E2124 (2008).
    • 68  Bach P, Tschäpe JA, Kopietz F et al.: Vaccination with Aβ-displaying virus-like particles reduces soluble and insoluble cerebral Aβ and lowers plaque burden in APP transgenic mice. J. Immunol.182(12),7613–7624 (2008).
    • 69  Lemere CA, Maron R, Selkoe DJ, Weiner HL: Nasal vaccination with β-amyloid peptide for the treatment of Alzheimer’s disease. DNA Cell. Biol.20(11),705–711 (2001).
    • 70  Zhang J, Wu X, Qin C et al.: A novel recombinant adeno-associated virus vaccine reduces behavioral impairment and β-amyloid plaques in a mouse model of Alzheimer’s disease. Neurobiol. Dis.14(3),365–379 (2003).
    • 71  Weiner HL, Lemere CA, Maron R et al.: Nasal administration of amyloid-β peptide decreases cerebral amyloid burden in a mouse model of Alzheimer’s disease. Ann. Neurol.48(4),567–579 (2000).
    • 72  Jicha GA: Is passive immunization for Alzheimer’s disease ‘alive and well’ or ‘dead and buried’? Expert Opin. Biol. Ther.9(4),481–491 (2009).
    • 73  Morgan D: Mechanisms of Aβ-plaque clearance following passive Aβ-immunization. Neurodegener. Dis.2(5),261–266 (2005).
    • 74  Lichtlen P, Mohajeri MH: Antibody-based approaches in Alzheimer’s research: safety, pharmacokinetics, metabolism, and analytical tools. J. Neurochem.104(4),859–874 (2008).
    • 75  Walsh DM, Selkoe DJ: Oligomers on the brain: the emerging role of soluble protein aggregates in neurodegeneration. Protein Pept. Lett.11(3),213–228 (2004).
    • 76  Kerchner GA, Boxer AL: Bapineuzumab. Expert Opin. Biol. Ther.10(7),1121–1130 (2010).
    • 77  Seubert P, Vigo-Pelfrey C, Esch F et al.: Isolation and quantification of soluble Alzheimer’s β-peptide from biological fluids. Nature359(6393),325–327 (1992).
    • 78  Siemers ER, Friedrich S, Dean RA 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. Clin. Neuropharmacol.33(2),67–73 (2010).
    • 79  DeMattos RB, Racke MM, Gelfanova V 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. Alzheimers Dement.5,P156–P157 (2009).
    • 80  Seubert P, Barbour R, Khan K et al.: Antibody capture of soluble Aβ does not reduce cortical Aβ amyloidosis in the PDAPP mouse. Neurodegener Dis.5(2),65–71 (2008).
    • 81  Yamada K, Yabuki C, Seubert P et al.: Aβ immunotherapy: intracerebral sequestration of Aβ by an anti-Aβ monoclonal antibody 266 with high affinity to soluble Aβ. J. Neurosci.29(36),11393–11398 (2009).▪ Questioned the peripheral sink hypothesis, suggesting that solanezumab may be effective by stabilizing Aβ monomers and preventing formation of Aβ oligomers.
    • 82  Siemers ER, Friedrich S, Dean RA et al.: P4–346: safety, tolerability and biomarker effects of an Aβ monoclonal antibody administered to patients with Alzheimer’s disease. Alzheimers Dement.4,T774 (2008).
    • 83  Racke MM, Boone LI, Hepburn DL et al.: Exacerbation of cerebral amyloid angiopathy-associated microhemorrhage in amyloid precursor protein transgenic mice by immunotherapy is dependent on antibody recognition of deposited forms of amyloid b. J. Neurosci.25(3),629–636 (2005).
    • 84  Nicholas T, Knebel W, Gastonguay MR et al.: Preliminary population pharmacokinetic modeling of PF-04360365, a humanized anti-amyloid monoclonal antibody, in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Dement.5,P253 (2009).
    • 85  Watts RJ, Chen M, Atwal J et al.: Selection of an anti-Aβ antibody that binds various forms of Aβ and blocks toxicity both in vitro and in vivo. Alzheimers Dement.5,P426 (2009).
    • 86  Relkin NR: Natural human antibodies targeting amyloid aggregates in intravenous immunoglobulin. Alzheimers Dement.4,T101 (2008).
    • 87  Du Y, Wei X, Dodel R et al.: Human anti-β-amyloid antibodies block β-amyloid fibril formation and prevent β-amyloid-induced neurotoxicity. Brain126(Pt 9),1935–1939 (2003).
    • 88  Ma QL, Lim GP, Harris-White ME et al.: Antibodies against β-amyloid reduce Aβ oligomers, glycogen synthase kinase-3b activation and tau phosphorylation in vivo and in vitro. J. Neurosci. Res.83(3),374–384 (2006).
    • 89  Istrin G, Bosis E, Solomon B: Intravenous immunoglobulin enhances the clearance of fibrillar amyloid-β peptide. J. Neurosci. Res.84(2),434–443 (2006).
    • 90  Dodel RC, Du Y, Depboylu C et al.: Intravenous immunoglobulins containing antibodies against β-amyloid for the treatment of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry75(10),1472–1474 (2004).
    • 91  Relkin NR, Szabo P, Adamiak B et al.: 18-month study of intravenous immunoglobulin for treatment of mild Alzheimer disease. Neurobiol. Aging30(11),1728–1736 (2009).▪ After the trial of Dodel and colleagues [90], this was the second small trial of intravenous immunoglobulin infusions for treatment of AD. In particular, this first American trial was longer, slightly larger and showed a possible benefit on the Mini Mental Scale Examination, forming the basis of Phase II and III trials by the same group in collaboration with Baxter and the NIH-sponsored Alzheimer’s Disease Cooperative Study.
    • 92  Johnson-Wood K, Lee M, Motter R et al.: Amyloid precursor protein processing and Aβ42 deposition in a transgenic mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA94(4),1550–1555 (1997).
    • 93  Black RS, Sperling RA, Safirstein B et al.: A single ascending dose study of bapineuzumab in patients with Alzheimer disease. Alzheimer Dis. Assoc. Disord.24(2),198–203 (2010).▪ Phase I, third-party unblinded, single ascending dose trial that examined 30 patients with mild-to-moderate AD receiving bapineuzumab infusion of 0.5, 1.5 or 5 mg/kg, or placebo.
    • 94  Salloway S, Sperling R, Gilman S et al.: A Phase II multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology73(24),2061–2070 (2009).▪▪ Main Phase II trial of bapineuzumab.
    • 95  Rinne JO, Brooks DJ, Rossor MN 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. Lancet Neurol.9(4),363–372 (2010).▪▪ Phase II trial of bapineuzumab in British and Finnish cohorts that demonstrated convincing in vivo evidence that the monoclonal antibody reduced cerebral Aβ levels, using 11C-PiB retention on PET as the primary outcome measure.
    • 96  Rabinovici GD, Jagust WJ: Amyloid imaging in aging and dementia: testing the amyloid hypothesis in vivo. Behav. Neurol.21(1),117–128 (2009).
    • 97  Blennow K, Zetterberg H, Wei J, Liu E, Black R, Grundman M: Immunotherapy with bapineuzumab lowers CSF tau protein levels in patients with Alzheimer’s disease. Alzheimers Dement.6(Suppl.),S134–S135 (2010).
    • 98  Laskowitz DT, Vitek MP: Apolipoprotein E and neurological disease: therapeutic potential and pharmacogenomic interactions. Pharmacogenomics8(8),959–969 (2008).
    • 99  Kim J, Basak JM, Holtzman DM: The role of apolipoprotein E in Alzheimer’s disease. Neuron63(3) 287–303 (2009).
    • 100  Schroeter S, Khan K, Barbour R et al.: Immunotherapy reduces vascular amyloid-β in PDAPP mice. J. Neurosci.28(27),6787–6793 (2008).▪ Recent passive immunization study with murine bapineuzumab demonstrated increased incidence of microhemorrhage despite low-vascular amyloid deposition in PDGF promoter expressing amyloid precursor protein mice at 12 months of age, suggesting no direct association between vascular amyloid burden and microhemorrhage.
    • 101  Laskowitz DT, Thekdi AD, Thekdi SD et al.: Downregulation of microglial activation by apolipoprotein E and apoE-mimetic peptides. Exp. Neurol.167(1),74–85 (2001).
    • 102  Atwood CS, Bishop GM, Perry G, Smith MA: amyloid-β: a vascular sealant that protects against hemorrhage? J. Neurosci. Res.70(3),356 (2002).
    • 103  Atwood CS, Bowen RL, Smith MA, Perry G: Cerebrovascular requirement for sealant, anti-coagulant and remodeling molecules that allow for the maintenance of vascular integrity and blood supply. Brain Res. Brain Res. Rev.43(1),164–178 (2003).
    • 104  Zhang-Nunes SX, Maat-Schieman ML, Van Duinen SG et al.: The cerebral β-amyloid angiopathies: hereditary and sporadic. Brain Pathol.16(1),30–39 (2006).
    • 105  Pfeifer M, Boncristiano S, Bondolfi L et al.: Cerebral hemorrhage after passive anti-Aβ immunotherapy. Science298(5597),1379 (2002).
    • 106  Racke MM, Boone LI, Hepburn DL et al.: Exacerbation of cerebral amyloid angiopathy-associated microhemorrhages in amyloid precursor protein transgenic mice by immunotherapy is dependent on antibody recognition of deposited forms of amyoid β. J. Neurosci.25(3),629–636 (2005).
    • 107  Wilcock DM, Jantzen PT, Li Q, Morgan D, Gordon MN: amyloid-β vaccination, but not nitro-nonsteroidal anti-inflammatory drug treatment, increases vascular amyloid and microhemorrhage while both reduce parenchymal amyloid. Neuroscience144(3),950–960 (2007).
    • 108  Patton RL, Kalback WM, Esh CL et al.: amyloid-β peptide remnants in AN1792-immunized Alzheimer’s disease patients: a biochemical analysis. Am. J. Pathol.169(3),1048–1063 (2006).
    • 109  Vellas B, Black R, Thal LJ et al.; AN1792 (QS-21)-251 Study Team: Long-term follow-up of patients immunized with AN1792: reduced functional decline in antibody responders. Curr. Alzheimer Res.6(2),144–151 (2009).
    • 110  Castellani RJ, Lee HG, Zhu X, Nunomura A, Perry G, Smith MA: Neuropathology of Alzheimer disease: pathognomonic but not pathogenic. Acta Neuropathol.111(6),503–509 (2006).
    • 111  Lee HG, Zhu X, Nunomura A, Perry G, Smith MA: Amyloid b: the alternate hypothesis. Curr. Alzheimer Res.3(6),75–80 (2006).
    • 112  Rottkamp CA, Raina AK, Zhu X et al.: Redox-active iron mediates amyloid-β toxicity. Free Radic. Biol. Med.30(4),447–450 (2001).
    • 113  Mura E, Lanni C, Preda S et al.: β-amyloid: a disease target or a synaptic regulator affecting age-related neurotransmitter changes? Curr. Pharm. Des.16(6),672–683 (2010).
    • 114  Vossel KA, Zhang K, Brodbeck J et al.: Tau reduction prevents A β-induced defects in axonal transport. Science330(6001),198 (2010).
    • 115  Duce JA, Tsatsanis A, Cater MA et al.: Iron-export ferroxidase activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell142(6),857–867 (2010).
    • 201  Eli Lilly and Company. Press release: 17 August, 2010. Lilly halts development of semagacestat for Alzheimer’s disease based on preliminary results of Phase III clinical trials http://newsroom.lilly.com/releasedetail.cfm?releaseid=499794
    • 202  A Phase 3 Study Evaluating Safety and Effectiveness of Immune Globulin Intravenous (IGIV 10%) for the Treatment of Mild to Moderate Alzheimer’s Disease http://clinicaltrials.gov/ct2/show/NCT00818662
    • 203  Study of Octagam 10% on the Treatment of Mild to Moderate Alzheimer’s Patients http://clinicaltrials.gov/ct2/show/NCT00812565
    • 204  Bapineuzumab in Patients With Mild to Moderate Alzheimer’s Disease (ApoE4 Carrier) http://clinicaltrials.gov/ct2/show/NCT00575055
    • 205  Study Evaluating the Safety and Efficacy of Bapineuzumab in Alzheimer Disease Patients http://clinicaltrials.gov/ct2/show/NCT00676143
    • 206  Bapineuzumab in Patients With Mild to Moderate Alzheimer’s Disease (ApoE4 Non-Carrier) http://clinicaltrials.gov/ct2/show/NCT00574132
    • 207  Study Evaluating the Efficacy and Safety of Bapineuzumab in Alzheimer Disease Patients http://clinicaltrials.gov/ct2/show/NCT00667810
    • 208  Elan and Wyeth plan to amend bapineuzumab Phase 3 protocols. Press release http://newsroom.elan.com/phoenix.zhtml?c=88326&p=irol-newsArticle&ID=1272546&highlight=(2009
    • 209  Study Evaluating the Long-term Safety and Efficacy of Subcutaneous Bapineuzumab http://clinicaltrials.gov/ct2/show/NCT00916617
    • 210  Study Evaluating Bapineuzumab In Alzheimer Disease Subjects http://clinicaltrials.gov/ct2/show/NCT00663026
    • 301  Kline EL, McMichael J: Method and composition for treatment of central nervous systems disease states associated with abnormal amyloid protein. WO91/16819 (1991).
    • 302  Schenk D: Prevention and treatment of amyloidogenic disease. WO99/27944 (1998).