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

Tau-based therapeutics for Alzheimer’s disease: active and passive immunotherapy

    Francesco Panza

    *Author for correspondence:

    E-mail Address: geriat.dot@geriatria.uniba.it

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    ,
    Vincenzo Solfrizzi

    Geriatric Medicine-Memory Unit & Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy

    Geriatric Medicine-Memory Unit & Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy

    ,
    Davide Seripa

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    ,
    Bruno P Imbimbo

    Research & Development Department, Chiesi Farmaceutici, Parma, Italy

    Research & Development Department, Chiesi Farmaceutici, Parma, Italy

    ,
    Madia Lozupone

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    ,
    Andrea Santamato

    Physical Medicine & Rehabilitation Section, ‘OORR’ Hospital, University of Foggia, Foggia, Italy

    Physical Medicine & Rehabilitation Section, ‘OORR’ Hospital, University of Foggia, Foggia, Italy

    ,
    Rosanna Tortelli

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    ,
    Ilaria Galizia

    Psychiatric Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Psychiatric Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    ,
    Camilla Prete

    Department of OrthoGeriatrics, Rehabilitation & Stabilization, Frailty Area, E.O. Galliera NR-HS Hospital, Genova, Italy

    Department of OrthoGeriatrics, Rehabilitation & Stabilization, Frailty Area, E.O. Galliera NR-HS Hospital, Genova, Italy

    ,
    Antonio Daniele

    Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy

    Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy

    ,
    Alberto Pilotto

    Department of OrthoGeriatrics, Rehabilitation & Stabilization, Frailty Area, E.O. Galliera NR-HS Hospital, Genova, Italy

    Department of OrthoGeriatrics, Rehabilitation & Stabilization, Frailty Area, E.O. Galliera NR-HS Hospital, Genova, Italy

    ,
    Antonio Greco

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    Geriatric Unit & Laboratory of Gerontology & Geriatrics, Department of Medical Sciences, IRCCS ‘Casa Sollievo della Sofferenza,’ San Giovanni Rotondo, Foggia, Italy

    &
    Giancarlo Logroscino

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy

    Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy

    Department of Clinical Research in Neurology, University of Bari Aldo Moro, ‘Pia Fondazione Cardinale G. Panico,’ Tricase, Lecce, Italy

    Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy

    Published Online:https://doi.org/10.2217/imt-2016-0019

    Pharmacological manipulation of tau protein in Alzheimer’s disease included microtubule-stabilizing agents, tau protein kinase inhibitors, tau aggregation inhibitors, active and passive immunotherapies and, more recently, inhibitors of tau acetylation. Animal studies have shown that both active and passive approaches can remove tau pathology and, in some cases, improve cognitive function. Two active vaccines targeting either nonphosphorylated (AAD-vac1) and phosphorylated tau (ACI-35) have entered Phase I testing. Notwithstanding, the recent discontinuation of the monoclonal antibody RG7345 for Alzheimer’s disease, two other antitau antibodies, BMS-986168 and C2N-8E12, are also currently in Phase I testing for progressive supranuclear palsy. After the recent impressive results in animal studies obtained by salsalate, the dimer of salicylic acid, inhibitors of tau acetylation are being actively pursued.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Alzheimer’s Association. 2015 Alzheimer's disease facts and figures. Alzheimers Dement. 11(3), 332–384 (2015).
    • 2 Schneider LS, Mangialasche F, Andreasen N et al. Clinical trials and late-stage drug development for Alzheimer's disease: an appraisal from 1984 to J. Intern. Med. 275(3), 251–283 (2014). • This review article discussed the last three decades of late-stage drug development for Alzheimer’s disease (AD) treatment.
    • 3 Frisardi V, Solfrizzi V, Imbimbo BP et al. Towards disease-modifying treatment of Alzheimer's disease: drugs targeting beta-amyloid. Curr. Alzheimer Res. 7(1), 40–55 (2010).
    • 4 Panza F, Solfrizzi V, Imbimbo BP, Logroscino G. Amyloid-directed monoclonal antibodies for the treatment of Alzheimer's disease: the point of no return? Expert Opin. Biol. Ther. 14(10), 1465–1476 (2014).
    • 5 Wischik CM, Harrington CR, Storey JM. Tau-aggregation inhibitor therapy for Alzheimer's disease. Biochem. Pharmacol. 88(4), 529–539 (2014). • This was a comprehensive review article describing the background of repeated failures of treatment approaches based on the β-amyloid cascade theory of AD, proposing the basis for the development of tau aggregation inhibitors (TAIs) for AD treatment.
    • 6 Martin L, Latypova X, Wilson CM et al. Tau protein kinases: involvement in Alzheimer's disease. Ageing Res. Rev. 12(1), 289–309 (2013).
    • 7 Iqbal K, Gong CX, Liu F. Microtubule-associated protein tau as a therapeutic target in Alzheimer's disease. Expert Opin. Ther. Targets 18(3), 307–318 (2014).
    • 8 Panza F, Frisardi V, Solfrizzi V et al. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 4(2), 213–238 (2012).
    • 9 Pedersen JT, Sigurdsson EM. Tau immunotherapy for Alzheimer's disease. Trends Mol. Med. 21(6), 394–402 (2015). • A comprehensive and very updated review of the principal tau-based active and passive immunization approaches.
    • 10 Buee L, Bussiere T, Buee-Scherrer V, Delacourte A, Hof PR. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Brain Res. Rev. 33(1), 95–130 (2000).
    • 11 Iqbal K, Liu F, Gong CX, Grundke-Iqbal I. Tau in Alzheimer disease and related tauopathies. Curr. Alzheimer Res. 7(8), 656–664 (2010).
    • 12 Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82(4), 239–259 (1991).
    • 13 Braak H, Braak E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol. Aging 18(4), 351–357 (1997).
    • 14 Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 70(11), 960–969 (2011).
    • 15 Mukaetova-Ladinska EB, Garcia-Sierra F, Hurt J et al. Staging of cytoskeletal and β-amyloid changes in human isocortex reveals biphasic synaptic protein response during progression of Alzheimer's disease. Am. J. Pathol. 157(2), 623–636 (2000).
    • 16 Ahmed RM, Paterson RW, Warren JD et al. Biomarkers in dementia: clinical utility and new directions. J. Neurol. Neurosurg. Psychiatry 85(12), 1426–1434 (2014).
    • 17 Grundke-Iqbal I, Iqbal K, Tung YC et al. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl Acad. Sci. USA 83(13), 4913–4917 (1986). •• This study described the discovery of the abnormal hyperphosphorylation of tau in neurofibrillary tangles in AD.
    • 18 Wischik CM, Novak M, Thøgersen HC et al. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer's disease. Proc. Natl Acad. Sci. USA 85(12), 4506–4510 (1988).
    • 19 Wischik CM, Novak M, Edwards PC et al. Structural characterization of the core of the paired helical filament of Alzheimer disease. Proc. Natl Acad. Sci. USA 85(13), 4884–4888 (1988).
    • 20 Guillozet-Bongaarts AL, Garcia-Sierra F, Reynolds MR et al. Tau truncation during neurofibrillary tangle evolution in Alzheimer's disease. Neurobiol. Aging 26(7), 1015–1022 (2005).
    • 21 Yan SD, Chen X, Schmidt AM et al. Glycated tau protein in Alzheimer disease: a mechanism for induction of oxidant stress. Proc. Natl Acad. Sci. USA 91(16), 7787–7791 (1994).
    • 22 Reynolds MR, Berry RW, Binder LI. Site-specific nitration and oxidative dityrosine bridging of the tau protein by peroxynitrite: implications for Alzheimer's disease. Biochemistry 44(5), 1690–1700 (2005).
    • 23 Min SW, Cho SH, Zhou Y et al. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67(6), 953–966 (2010).
    • 24 Liu F, Iqbal K, Grundke-Iqbal I et al. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease. Proc. Natl Acad. Sci. USA 101(29), 10804–10809 (2004). •• This study reported the discovery of global decrease in O-GlcNAcylation and the regulation of phosphorylation of tau by this post-translational modification in AD brain.
    • 25 Perry G, Mulvihill P, Fried VA et al. Immunochemical properties of ubiquitin conjugates in the paired helical filaments of Alzheimer disease. J. Neurochem. 52(5), 1523–1528 (1989).
    • 26 Kolarova M, García-Sierra F, Bartos A et al. Structure and pathology of tau protein in Alzheimer disease. Int. J. Alzheimers Dis. 2012, 731526 (2012).
    • 27 Kimura T, Fukuda T, Sahara N et al. Aggregation of detergent-insoluble tau is involved in neuronal loss but not in synaptic loss. J. Biol. Chem. 285(49), 38692–38699 (2010).
    • 28 Alonso Adel C, Li B, Grundke-Iqbal I, Iqbal K. Polymerization of hyperphosphorylated tau into filaments eliminates its inhibitory activity. Proc. Natl Acad. Sci. USA 103(23), 8864–8869 (2006).
    • 29 Iqbal K, Alonso Adel C, Grundke-Iqbal I. Cytosolic abnormally hyperphosphorylated tau but not paired helical filaments sequester normal MAPs and inhibit microtubule assembly. J. Alzheimers Dis. 14(4), 365–370 (2008).
    • 30 Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 443(7113), 780–786 (2006).
    • 31 Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J. Biol. Chem. 284(19), 12845–12852 (2012).
    • 32 Clavaguera F, Bolmont T, Crowther RA et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell. Biol. 11(7), 909–913 (2009).
    • 33 de Calignon A, Polydoro M, Suárez-Calvet M et al. Propagation of tau pathology in a model of early Alzheimer's disease. Neuron 73(4), 685–697 (2012).
    • 34 Asuni AA, Boutajangout A, Quartermain D, Sigurdsson EM. Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J. Neurosci. 27(34), 9115–9129 (2007). •• This study described the first successful reduction of tau pathology in JNPL3 (P301L) mice immunized with a tau phosphopeptide.
    • 35 Masliah E, Rockenstein E, Adame A et al. Effects of alpha-synuclein immunization in a mouse model of Parkinson's disease. Neuron 46(6), 857–868 (2005).
    • 36 Iqbal K, Liu F, Gong CX et al. Mechanisms of tau-induced neurodegeneration. Acta Neuropathol. 118(1), 53–69 (2009).
    • 37 de Calignon A, Spires-Jones TL, Pitstick R et al. Tangle-bearing neurons survive despite disruption of membrane integrity in a mouse model of tauopathy. J. Neuropathol. Exp. Neurol. 68(7), 757–761 (2009).
    • 38 Roberson ED, Scearce-Levie K, Palop JJ et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science 316(5825), 750–754 (2007).
    • 39 Vossel KA, Zhang K, Brodbeck J et al. Tau reduction prevents A{beta}-induced defects in axonal transport. Science 330(6001), 198 (2010).
    • 40 Rhein V, Song X, Wiesner A et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimer's disease mice. Proc. Natl Acad. Sci. USA 106(47), 20057–20062 (2009).
    • 41 Vershinin M, Carter BC, Razafsky DS et al. Multiple-motor based transport and its regulation by tau. Proc. Natl Acad. Sci. USA 104(1), 87–92 (2007).
    • 42 Ittner LM, Fath T, Ke YD et al. Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia. Proc. Natl Acad. Sci. USA 105(41), 15997–16002 (2008).
    • 43 Ittner LM, Ke YD, Gotz J. Phosphorylated tau interacts with c-Jun N-terminal kinase interacting protein 1 (JIP1) in Alzheimer disease. J. Biol. Chem. 284(31), 20909–20916 (2009).
    • 44 Eckert A, Schulz KL, Rhein V, Götz J. Convergence of Amyloid-beta and tau pathologies on mitochondria in vivo. Mol. Neurobiol. 41(-3), 107–114 (2010).
    • 45 Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease. Neurology 42(3 Pt 1), 631–639 (1992).
    • 46 Anand K, Sabbagh M. Early investigational drugs targeting tau protein for the treatment of Alzheimer's disease. Expert Opin. Investig. Drugs 24(10), 1355–1360 (2015). • A comprehensive review of current drug therapies aimed at targeting the tau protein.
    • 47 Khatoon S, Grundke-Iqbal I, Iqbal K. Brain levels of microtubule-associated protein tau are elevated in Alzheimer's disease: a radioimmuno-slot-blot assay for nanograms of the protein. J. Neurochem. 59(2), 750–753 (1992). •• This study for the first time described that the brain level of tau is approximately four- to five-fold increased in AD and that this increase is in the form of abnormally hyperphosphorylated tau.
    • 48 Wen Y, Planel E, Herman M et al. Interplay between cyclin-dependent kinase 5 and glycogen synthase kinase 3β mediated by neuregulin signaling leads to differential effects on tau phosphorylation and amyloid precursor protein processing. J. Neurosci. 28(10), 2624–2632 (2008).
    • 49 Tolosa E, Litvan I, Höglinger GU et al. A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov. Disord. 29(4), 470–478 (2014). • The negative Tau Restoration on PSP (TAUROS) Phase II trial on the glycogen synthase kinase-3 (GSK-3) inhibitor tideglusib for the treatment of progressive supranuclear palsy (PSP).
    • 50 Del Ser T, Steinwachs KC, Gertz HJ et al. Treatment of Alzheimer's disease with the GSK-3 inhibitor tideglusib: a pilot study. J. Alzheimers Dis. 33(1), 205–215 (2013).
    • 51 Lovestone S, Boada M, Dubois B et al. ARGO investigators. A Phase II trial of tideglusib in Alzheimer's disease. J. Alzheimers Dis. 45(1), 75–88 (2015).
    • 52 Kins S, Crameri A, Evans DR et al. Reduced protein phosphatase 2A activity induces hyperphosphorylation and altered compartmentalization of tau in transgenic mice. J. Biol. Chem. 276(41), 38193–38200 (2001).
    • 53 Voronkov M, Braithwaite SP, Stock JB. Phosphoprotein phosphatase 2A: a novel druggable target for Alzheimer's disease. Future Med. Chem. 3(7), 821–833 (2011).
    • 54 Medina M, Avila J. Further understanding of tau phosphorylation: implications for therapy. Expert Rev. Neurother. 15(1), 115–122 (2015).
    • 55 Wang X, Blanchard J, Kohlbrenner E et al. The carboxy-terminal fragment of inhibitor-2 of protein phosphatase-2A induces Alzheimer disease pathology and cognitive impairment. FASEB J. 24(11), 4420–4321 (2010). • These studies showed that adeno-associated virus vector-mediated expression of I2CTF or I2NTF and I2CTF replicate several features of AD including abnormal hyperphosphorylation and sarkosyl insoluble tau, and cognitive impairment in rats.
    • 56 Landrieu I, Smet-Nocca C, Amniai L et al. Molecular implication of PP2A and Pin1 in the Alzheimer's disease specific hyperphosphorylation of Tau. PLoS ONE 6(6), e21521 (2011).
    • 57 Sontag JM, Nunbhakdi-Craig V, Sontag E. Leucine carboxyl methyltransferase 1 (LCMT1)-dependent methylation regulates the association of protein phosphatases 2A and Tau protein with plasma membrane microdomains in neuroblastoma cells. J. Biol. Chem. 288(38), 27396–27405 (2013).
    • 58 Grundke-Iqbal I, Iqbal K, Quinlan M et al. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J. Biol. Chem. 261(13), 6084–6089 (1986).
    • 59 Iqbal K, Grundke-Iqbal I, Zaidi T et al. Defective brain microtubule assembly in Alzheimer's disease. Lancet 2(8504), 421–426 (1986). • This study described the discovery of the cytosolic abnormally hyperphosphorylated tau in AD and inhibition of in vitro microtubule assembly by it.
    • 60 Butler D, Bendiske J, Michaelis M et al. Microtubule-stabilizing agent prevents protein accumulation-induced loss of synaptic markers. Eur. J. Pharmacol. 562(1–2), 20–27 (2007).
    • 61 Zhang B, Carroll J, Trojanowski JQ et al. The microtubule-stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer-like pathology in an interventional study with aged tau transgenic mice. J. Neurosci. 32(11), 3601–3611 (2012). • This study described the microtubule-stabilizing agent epothilone D in animal tau model of AD.
    • 62 Cortice biosciences announces results from studies evaluating pipeline candidates TPI 287 and CRT 001 in preclinical models of tauopathies and Alzheimer's disease. http://globenewswire.com/newsrelease/2014/11/12/682514/10107850/en/Cortice-Biosciences-Announces-Results-From-Studies-Evaluating-Pipeline-Candidates-TPI-287-and-CRT-001-in-Preclinical-Models-of-Tauopathies-and-Alzheimer-s-Disease.html
    • 63 Bassan M, Zamostiano R, Davidson A et al. Complete sequence of a novel protein containing a femtomolar-activity-dependent neuroprotective peptide. J. Neurochem. 72(3), 1283–1293 (1999).
    • 64 Vulih-Shultzman I, Pinhasov A, Mandel S et al. Activity-dependent neuroprotective protein snippet NAP reduces tau hyperphosphorylation and enhances learning in a novel transgenic mouse model. J. Pharmacol. Exp. Ther. 323(2), 438–449 (2007).
    • 65 Gozes I, Giladi E, Pinhasov A et al. Activity-dependent neurotrophic factor: intranasal administration of femtomolar-acting peptides improve performance in a water maze. J. Pharmacol. Exp. Ther. 293(3), 1091–1098 (2000).
    • 66 Gozes I. NAP (davunetide) provides functional and structural neuroprotection. Curr. Pharm. Des. 17(10), 1040–1044 (2011).
    • 67 Gozes I, Schirer Y, Idan-Feldman A et al. NAP alpha-aminoisobutyric acid (IsoNAP). J. Mol. Neurosci. 52(1), 1–9 (2014).
    • 68 Boxer AL, Lang AE, Grossman M et al. Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled phase 2/3 trial. Lancet Neurol. 13, 676–685 (2014). • The substantially negative Phase II trial of davunetide, a compound able to promote microtubule stability and reduce tau phosphorylation, in patients with PSP.
    • 69 Cisek K, Cooper GL, Huseby CJ, Kuret J. Structure and mechanism of action of tau aggregation inhibitors. Curr. Alzheimer Res. 11(10), 918–927 (2014). • A comprehensive and very updated review of the structures and mechanisms of principal covalent and noncovalent TAIs.
    • 70 Taniguchi S, Suzuki N, Masuda M et al. Inhibition of heparin-induced Tau filament formation by phenothiazines, polyphenols, and porphyrins. J. Biol. Chem. 280(9), 7614–7623 (2005).
    • 71 Wischik CM, Edwards PC, Lai RY et al. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc. Natl Acad. Sci. USA 93(20), 11213–11218 (1996). •• First study in which phenothiazines were proposed as TAIs for AD treatment.
    • 72 Crowe A, James MJ, Lee VM et al. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J. Biol. Chem. 288(16), 11024–11037 (2013).
    • 73 Schafer KN, Cisek K, Huseby CJ et al. Structural determinants of Tau aggregation inhibitor potency. J. Biol. Chem. 288(45), 32599–32611 (2013).
    • 74 Kees F. Dimethyl fumarate: a Janus-faced substance? Expert Opin. Pharmacother. 14(11), 1559–1567 (2013).
    • 75 Wischik CM, Staff RT, Wischik DJ et al. Tau aggregation inhibitor therapy: an exploratory phase 2 study in mild or moderate Alzheimer's disease. J. Alzheimers Dis. 44(2), 705–720 (2015). •• Completed Phase II study regarding derivative of methylthioninium (MT).
    • 76 Harrington CR, Storey JM, Clunas S et al. Cellular models of aggregation-dependent template-directed proteolysis to characterize tau aggregation inhibitors for treatment of Alzheimer disease. J. Biol. Chem. 290(17), 10862–10875 (2015). •• This paper described primary synthesis and characterization of TRx0237, the critical concentration required for dissolution of tau aggregates isolated from AD brain tissues and two cellular models developed to measure the prion-like processing of tau protein in living cell systems and measurement of tau aggregation inhibition activity of novel drug candidates.
    • 77 Melis V, Magbagbeolu M, Rickard JE et al. Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models. Behav. Pharmacol. 26(4), 353–368 (2015). •• An analysis of results from the use of MT and TRx0237 in two transgenic models and defining the brain concentration range for tau aggregation inhibition activity for MT in vivo.
    • 78 Baddeley TC, McCaffrey J, Storey JM et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer's disease. J. Pharmacol. Exp. Ther. 352(1), 110–118 (2015). •• This article presented the four Phase I studies and two preclinical studies that were required to get to the bottom of the bioavailability limitations of the form of MT tested in the Phase II trial and represented the basis for proceeding into Phase III with TRx0237.
    • 79 Wisniewski T, Drummond E. Developing therapeutic vaccines against Alzheimer's disease. Expert Rev. Vaccines 15(3) 401–415 (2016).
    • 80 Götz J, Ittner A, Ittner LM. Tau-targeted treatment strategies in Alzheimer's disease. Br. J. Pharmacol. 165(5), 1246–1259 (2012).
    • 81 Congdon EE, Gu J, Sait HB, Sigurdsson EM. Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J. Biol. Chem. 288(49), 35452–35465 (2013).
    • 82 Gu J, Congdon EE, Sigurdsson EM. Two novel tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce tau pathology. J. Biol. Chem. 288(46), 33081–33095 (2013).
    • 83 Collin L, Bohrmann B, Göpfert U, Oroszlan-Szovik K, Ozmen L, Grüninger F. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer's disease. Brain 137(Pt 10), 2834–2846 (2014).
    • 84 Congdon EE, Krishnaswamy S, Sigurdsson EM. Harnessing the immune system for treatment and detection of tau pathology. J. Alzheimers Dis. 40(Suppl. 1), S113–S121 (2014).
    • 85 Gros-Louis F, Soucy G, Lariviere R, Julien JP. Intracerebroventricular infusion of monoclonal antibody or its derived Fab fragment against misfolded forms of SOD1 mutant delays mortality in a mouse model of ALS. J. Neurochem. 113(5), 1188–1199 (2010).
    • 86 Masliah E, Rockenstein E, Adame A et al. Effects of alpha-synuclein immunization in a mouse model of Parkinson's disease. Neuron 46(6), 857–868 (2005).
    • 87 Tampellini D, Magrane J, Takahashi RH et al. Internalized antibodies to the Abeta domain of APP reduce neuronal Abeta and protect against synaptic alterations. J. Biol. Chem. 282(26), 18895–18906 (2007).
    • 88 Götz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301L tau transgenic mice induced by Abeta 42 fibrils. Science 293(5534), 1491–1495 (2001).
    • 89 Lewis J, Dickson DW, Lin W-L et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant Tau and APP. Science 293(5534), 1487–1491 (2001).
    • 90 Oddo S, Billings L, Kesslak JP, Cribbs DH, LaFerla FM. Abeta immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron 43(3), 321–332 (2004).
    • 91 Kfoury N, Holmes BB, Jiang H, Holtzman DM, Diamond MI. Trans-cellular propagation of Tau aggregation by fibrillar species. J. Biol. Chem. 287(23), 19440–19451 (2012).
    • 92 Yanamandra K, Kfoury N, Jiang H et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 80(2), 402–414 (2013).
    • 93 Boutajangout A, Ingadottir J, Davies P, Sigurdsson EM. Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. J. Neurochem. 118(4), 658–667 (2011). •• First study in which a passive immunization antitau protein was studied in an animal model with decreased tau pathology and functional impairments.
    • 94 Rosenmann H, Grigoriadis N, Karussis D et al. Tauopathy-like abnormalities and neurologic deficits in mice immunized with neuronal tau protein. Arch. Neurol. 63(10), 1459–1467 (2006).
    • 95 Boutajangout A, Quartermain D, Sigurdsson EM. Immunotherapy targeting pathological tau prevents cognitive decline in a new tangle mouse model. J. Neurosci. 30(49), 16559–16566 (2010).
    • 96 Kontsekova E, Zilka N, Kovacech B et al. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer's disease model. Alzheimers Res. Ther. 6(4), 44 (2014). •• First tau vaccine for AD.
    • 97 Paholikova K, Salingova B, Opattova A et al. N-terminal truncation of microtubule associated protein tau dysregulates its cellular localization. J. Alzheimers Dis. 43(3), 915–926 (2015).
    • 98 Encouraging Results of AXON's Tau Vaccine Advance Alzheimer's Therapy - Washington, D.C., July 23, 2015. www.axon-neuroscience.eu/docs/press-release-axon-post-aaic.pdf
    • 99 Theunis C, Crespo-Biel N, Gafner V et al. Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau. P301L mice that model tauopathy. PLoS ONE 8(8), e72301 (2013).
    • 100 Chai X, Wu S, Murray TK et al. Passive immunization with anti-tau antibodies in two transgenic models: reduction of tau pathology and delay of disease progression. J. Biol. Chem. 286(39), 34457–34467 (2011).
    • 101 Congdon EE, Gu J, Sait HB, Sigurdsson EM. Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J. Biol. Chem. 288(49), 35452–35465 (2013).
    • 102 Gu J, Congdon EE, Sigurdsson EM. Two novel Tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce Tau protein pathology. J. Biol. Chem. 288(46), 33081–33095 (2013).
    • 103 Kontsekova E, Ivanovova N, Handzusova M, Novak M. Chaperone-like antibodies in neurodegenerative tauopathies: implication for immunotherapy. Cell. Mol. Neurobiol. 29(6–7), 793–788 (2009).
    • 104 Taniguchi T, Sumida M, Hiraoka S et al. Effects of different anti-tau antibodies on tau fibrillogenesis: RTA-1 and RTA-2 counteract tau aggregation. FEBS Lett. 579(6), 1399–1404 (2005).
    • 105 Zilka N, Kontsekova E, Novak M. Chaperone-like antibodies targeting misfolded tau protein: new vistas in the immunotherapy of neurodegenerative foldopathies. J. Alzheimers Dis. 15, 169–179 (2008).
    • 106 Kontsekova E, Zilka N, Kovacech B et al. Identification of structural determinants on tau protein essential for its pathological function: novel therapeutic target for tau immunotherapy in Alzheimer's disease. Alzheimers Res. Ther. 6(4), 45 (2014).
    • 107 Buée L, Bussière T, Buée-Scherrer V et al. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Brain Res. Rev. 33(1), 95–130 (2000).
    • 108 Collin L, Bohrmann B, Göpfert U et al. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer's disease. Brain 137(Pt 10), 2834–2846 (2014). • Monoclonal antibody for treatment of tau in a mouse model of AD.
    • 109 Troquier L, Caillierez M, Burnouf S et al. Targeting phospho-Ser422 by active Tau immunotherapy in the THY-Tau22 mouse model: a suitable therapeutic approach. Curr. Alzheimer Res. 9(4), 397–405 (2012).
    • 110 Bae EJ, Lee HJ, Rockenstein E et al. Antibody-aided clearance of extracellular alpha-synuclein prevents cell-to-cell aggregate transmission. J. Neurosci. 32(39), 13454–13469 (2012).
    • 111 F. Hoffmann-La Roche Ltd. Product Development Portfolio. Pharma pipeline changes. www.roche.com/research_and_development/who_we_are_how_we_work/pipeline.htm
    • 112 Bright J, Hussain S, Dang V et al. Human secreted tau increases amyloid-beta production. Neurobiol. Aging 36(2), 693–709 (2015).
    • 113 Braak H, Del Tredici K. Alzheimer's pathogenesis: is there neuron-to-neuron propagation?. Acta Neuropathol. 121(5), 589–595 (2011).
    • 114 Yanamandra K, Kfoury N, Jiang H et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 80(2), 402–414 (2013).
    • 115 Yanamandra K, Jiang H, Mahan TE et al. Anti-tau antibody reduces insoluble tau and decreases brain atrophy. Ann. Clin. Transl. Neurol. 2(3), 278–288 (2015).
    • 116 Dai CL, Chen X, Kazim SF et al. Passive immunization targeting the N-terminal projection domain of tau decreases tau pathology and improves cognition in a transgenic mouse model of Alzheimer disease and tauopathies. J. Neural. Transm. (Vienna) 122(4), 607–617 (2015).
    • 117 Wang JZ, Grundke-Iqbal I, Iqbal K. Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration. Eur. J. Neurosci. 25(1), 59–68 (2007). • This study demonstrated that protein phosphatase-2A regulated the abnormal phosphorylation sites of tau and that catalysis by more than one combination of tau protein kinases could lead to the abnormal hyperphosphorylation of tau.
    • 118 Ragupathi G, Gathuru J, Livingston P. Antibody inducing polyvalent cancer vaccines. Cancer Treat. Res. 123, 157–180 (2005).
    • 119 Willet M, Kurup D, Papaneri A et al. Preclinical development of inactivated rabies virus-based polyvalent vaccine against rabies and filoviruses. J. Infect. Dis. 212(Suppl. 2), S414–S424 (2015).
    • 120 Morris M, Knudsen GM, Maeda S et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat. Neurosci. 18(8), 1183–1189 (2015).
    • 121 Keck S, Nitsch R, Grune T, Ullrich O. Proteasome inhibition by paired helical filament-tau in brains of patients with Alzheimer's disease. J. Neurochem. 85(1), 115–122 (2013).
    • 122 Han DH, Na HK, Choi WH et al. Direct cellular delivery of human proteasomes to delay tau aggregation. Nat. Commun. 5, 5633, (2014).
    • 123 Myeku N, Clelland CL, Emrani S et al. Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat. Med. 22(1), 46–53 (2016).
    • 124 Min SW, Chen X, Tracy TE et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat. Med. 21(10), 1154–1162 (2015).
    • 125 Cook C, Carlomagno Y, Gendron TF et al. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum. Mol. Genet. 23(1), 104–116 (2014).
    • 126 Wang JZ, Zhang Y. Configuration-specific immunotherapy targeting cis pThr231-Pro232 tau for Alzheimer disease. J. Neurol. Sci. 348(1–2), 253–255 (2015).
    • 127 Sigurdsson EM. Tau immunotherapy and imaging. Neurodegener. Dis. 13(2), 103–106 (2014).
    • 128 Castillo-Carranza DL, Sengupta U, Guerrero-Muñoz MJ et al. Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J. Neurosci. 34(12), 4260–4272 (2014).