Hostname: page-component-5c6d5d7d68-xq9c7 Total loading time: 0 Render date: 2024-08-21T21:44:36.921Z Has data issue: false hasContentIssue false

What Is the Rationale for New Treatment Strategies in Alzheimer's Disease?

Published online by Cambridge University Press:  07 November 2014

Abstract

Alzheimer's disease (AD) is characterized by the abnormal extracellular accumulation of amyloid β-peptide (Aβ) into neuritic plaques and the intraneuronal aggregation of the microtubule-associated protein tau to form neurofibrillary tangles. These molecular events are implicated in the selective damage to neural systems critical for the brain functions that are impaired in AD. Impairment of cholinergic neurotransmission may be an important factor underlying the defects in cognition and memory that characterize AD. Cholinesterase (ChE) inhibitors, such as donepezil, rivastigmine, and galantamine, cause symptomatic improvement by inhibiting the breakdown of the neurotransmitter acetylcholine to increase its synaptic availability and, in the case of galantamine, by also allosterically potentiating nicotinic cholinergic receptors. Other agents, including vitamin E, monoamine oxidase inhibitors, and statins, have shown some benefit in epidemiological studies and clinical trials although compelling evidence of their efficacy is lacking. Memantine, shown to cause cognitive and functional improvement, is not an ChE inhibitor and does not interact with marketed ChE inhibitors. While the mechanism of action of memantine in AD is not known, the principal pharmacologic actions at therapeutic dose are inhibition of ionotropic neurotransmitter receptors, specifically N-methyl-D-aspartate (NMDA), 5-HT3, and nicotinic receptors. Like other NMDA antagonists, memantine causes behavioral activation associated with enhanced cerebral glucose utilization. Studies have shown that memantine can reverse the decreased metabolic activity associated with AD, possibly accounting for its beneficial effects on cognition and global functioning. Memantine also has neuroprotective properties and can inhibit Aβ-induced neurodegeneration.

Type
Academic Supplement
Copyright
Copyright © Cambridge University Press 2004

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

REFERENCES

1.DL, Price, RE, Tanzi, DR, Borchelt, SS, Sisodia. Alzheimer's disease: genetic studies and transgenic models. Annu Rev Genet. 1998;32:461493.Google Scholar
2.DJ, Selkoe. Alzheimer's disease: genes, proteins, and therapy. Physiol Rev. 2001;81:741766.Google Scholar
3.Avila, J, JJ, Lucas, Perez, M, Hernandez, F. Role of tau protein in both physiological and pathological conditions. Physiol Rev. 2004;84:361384.Google Scholar
4.DJ, Selkoe, Schenk, D. Alzheimer's disease: molecular understanding predicts amyloid-hased therapeutics. Annu Rev Pharmacol Toxicol. 2003;43:545584.Google Scholar
5. Giacobini, E. Chotinergic function and Alzheimer's disease. Int J Geriatr Psychiatry. 2003;18(suppl 1):S1S5.Google Scholar
6.GL, Wenk, Danysz, W, SL, Mobley. Investigations of neurotoxicity and neuro-protection within the nucleus basalis of the rat. Brain Res. 1994;655:711.Google Scholar
7.MJ, Knapp, DS, Knopman, PR, Solomon, WW, Pendlebury, CS, Davis, SI, Gracon; The Tacrine Study Group. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer's disease. JAMA. 1994;271;985991.Google Scholar
8.JL, Cummings. Use of cholinesterase inhibitors in clinical practice: evidence-based recommendations. Am J Geriatr Psychiatry. 2003;11:131145.Google Scholar
9.Rogers, SL, Farlow, MR, Doody, RS, Mohs, R, Friedhoff, LT. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer's disease. Donepezil Study Group. Neurology. 1998;50:136145.CrossRefGoogle ScholarPubMed
10.Rosler, M, Anand, R, Cicin-Sain, A, et al.Efficacy and safety of rivastigmine in patients with Alzheimer's disease: international randomised controlled trial. BMJ. 1999;318:633638.Google Scholar
11.Jann, MW, Shirley, KL, Small, GW. Clinical pharmacokinetics and pharmaco-dynamics of cholinesterase inhibitors. Clin Pharmocokinet. 2002;41:719739.Google Scholar
12.Lilienfeld, S. Galantamine—a novel cholinergic drug with a unique dual mode of action for the treatment of patients with Alzheimer's disease. CNS Drug Reviews. 2002;8:159176.CrossRefGoogle ScholarPubMed
13.Samochocki, M, Hoffle, A, Fehrenbacher, A, et al.Galantamine is an allosterically potentiating ligand of neuronal nicotinic but not of muscarinic acetylcholine receptors. J Pharmacol Exp Ther. 2003;305:10241036.Google Scholar
14.Dajas-Bailador, FA, Heimala, K, Wonnacott, S. The allosteric potentiation of nicotinic acetylcholine receptors by galantamine is transduced into cellular responses in neurons: Ca2+ signals and neurotransmitter release. Mol Pharmacol. 2003;64:12171226.Google Scholar
15.Raskind, MA, Peskind, ER, Wessel, T, Yuan, W; the Galantamine USA-1 Study Group. Galantamine in AD: a 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology. 2000;54:22612268.Google Scholar
16.Wilcock, GK, Lilienfeld, S, Gaens, E; Galantamine International-1 Study Group. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer's disease: multicentre randomised controlled trial. BMJ. 2000;321:14451449.Google Scholar
17.Nordberg, A, Winblad, B. Reduced number of [3H]nicotine and [3H]acetylcholine binding sites in the frontal cortex of Alzheimer brains. Neurosci Lett. 1986;72:115119.Google Scholar
18.Kellar, KJ, Whitehouse, PJ, Martino-Barrows, AM, Marcus, K, Price, DL. Muscarinic and nicotinic cholinergic binding sites in Alzheimer's disease cerebral cortex. Brain Res. 1987;436:6268.Google Scholar
19.Nordberg, A, Adem, A, Hardy, J, Winblad, B. Change in nicotinic receptor subtypes in temporal cortex of Alzheimer brains. Neurosci Lett. 1988;86:317321.Google Scholar
20.Maelicke, A, Schrattenholz, A, Samochocki, M, Radina, M, EX, Albuquerque. Allosterically potentiating ligands of nicotinic receptors as a treatment strategy for Alzheimer's disease. Behav Brain Res. 2000;113:199206.Google Scholar
21.Sher, E, Chen, Y, TJ, Sharples, et al.Physiological roles of neuronal nicotinic receptor subtypes: new insights on the nicotinic modulation of neurotransmitter release, synaptic transmission and plasticity. Curr Top Med Chem. 2004;4:283297.Google Scholar
22.Woodruff-Pak, DS, RW, Vogel III, Wenk, GL. Galantamine: effect on nicotinic receptor binding, acetylcholinesterase inhibition, and learning. Proc Natl Acad Sci USA. 2001;98:20892094.Google Scholar
23.Santos, MD, Alkondon, M, Pereira, EF, et al.The nicotinic allosteric potentiating ligand galantamine facilitates synaptic transmission in the mammalian central nervous system. Mol Pharmacol. 2002;61:12221234.Google Scholar
24.Levin, ED, Rezvani, AH. Nicotinic treatment for cognitive dysfunction. Curr Drug Target CNS Neurol Disord. 2002:423431.Google Scholar
25.Rezvani, AH, Levin, ED. Cognitive effects of nicotine. Biol Psychiatry. 2001;49:258267.Google Scholar
26.Ringheim, GE, Conant, K. Neurodegenerative disease and the neuroimmune axis (Alzheimer's and Parkinson's disease, and viral infections). J Neuroimmunol. 2004;147:4349.Google Scholar
27.JCS, Breitner, Welsh, KA, Helms, MJ, et al.Delayed onset of Alzheimer's disease with nonsteroidal anti-inflammatory and histamine H2 blocking drugs. Neurobiol Aging. 1995;16:523530.Google Scholar
28.Stewart, WF, Kawas, C, Corrada, M, Metter, EJ. Risk of Alzheimer's disease and duration of NSAID use. Neurology. 1997;48:626632.Google Scholar
29.Rogers, J, Kirby, LC, Hempelman, SR, et al.Clinical trial of indomethacin in Alzheimer's disease. Neurology. 1993;43:16091611.Google Scholar
30.Aisen, PS, Schafer, KA, Grundman, M, et al.Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;289:28192826.Google Scholar
31.Reines, SA, Block, GA, Morris, JC, et al.Rofecoxib: no effect on Alzheimer's disease in a 1-year, randomized, blinded, controlled study. Neurology. 2004;62:6671.Google Scholar
32.Henderson, VW, Paganini-Hill, A, Miller, BL, et al.Estrogen for Alzheimer's disease in women: randomized, double-blind, placebo-controlled trial. Neurology. 2000;54:295301.Google Scholar
33.Shumaker, SA, Legault, C, Rapp, SR, et al.Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial. JAMA. 2003;289:26512662.Google Scholar
34.Sano, M, Ernesto, C, Thomas, RG, et al.A controlled trial of selegiline, alphatocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. N Engl J Med. 1997;336:12161222.Google Scholar
35.Miller, LJ, Chacko, R. The role of cholesterol and statins in Alzheimer's disease. Ann Pharmacother. 2004;38:9198.Google Scholar
36.Doraiswamy, PM. Non-cholinergic strategies for treating and preventing Alzheimer's disease. CNS Drugs. 2002;6:811824.CrossRefGoogle Scholar
37.Schenk, D. Amyloid- immunotherapy for Alzheimer's disease: the end of the beginning. Nat Rev Neurosci. 2002;3:824828.Google Scholar
38.Morgan, D. Antibody therapy for Alzheimer's disease. Expert Rev Vaccines. 2003;2:5359.Google Scholar
39.Conway, KA, Baxter, EW, Felsenstein, KM, Reitz, AB. Emerging -amyloid therapies for the treatment of Alzheimer's disease. Curr Pharm Des. 2003;9:427447.Google Scholar
40.Rogawski, MA, Wenk, GLThe neuropharmacological basis for the use of memantine in the treatment of Alzheimer's disease. CNS Drug Rev. 2003;9:275308.Google Scholar
41.Potkin, SG, Peskind, ER, Pomara, N, McDonald, S, Xie, Y, Gergel, I. Memantine monotherapy is effective and safe for the treatment of mild to moderate Alzheimer's disease: a randomized controlled trial [Abstract LBS.003]. American Academy of Neurology 56th Annual Meeting; April 24–May 1, 2004; San Francisco, Calif.Google Scholar
42.Wenk, GL, Quack, G, Moebius, HJ, Danysz, W. No interaction of memantine with acetylcholinesterase inhibitors approved for clinical use. Life Sci. 2000;66:10791083.Google Scholar
43.Hartmann, S, Möbius, HJ. Tolerability of memantine in combination with cholinesterase inhibitors in dementia therapy. Int Clin Psychopharmacol. 2003;18:8185.Google Scholar
44.Tariot, PN, Farlow, MR, Grossberg, GT, Graham, SM, McDonald, S, Gergel, I, and the Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291:317324.Google Scholar
45.Rammes, G, Rupprecht, R, Ferrari, U, Zieglgansberger, W, Parsons, CG. The N-methyl-D-aspartate receptor channel blockers memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes antagonise 5-HT3 receptor currents in cultured HEK-293 and N1E-115 cell systems in a non-competitive manner. Neurosci Lett. 2001;306:8184.Google Scholar
46.Danysz, W, Parsons, CG, Möbius, HJ, Stöffler, A, Quack, G. Neuroprotective and symptomalogical action of memantine relevant for Alzheimer's disease: a unified glutamatergic hypothesis on the mechanism of action. Neurotoxicity Res. 1999;2:8597.Google Scholar
47.Dingledine, R, Borges, K, Bowie, D, SF, Traynelis. The glutamate receptor ion channels. Pharmacol Rev. 1999;51:761.Google Scholar
48.Lynch, MA. Long-term potentiation and memory. Physiol Rev. 2004;84:87136.Google Scholar
49.Rogawski, MA. Excitatory amino acids and seizures. In: Stone, TW, ed. CNS Neurotransmitters and Neuromodulators, Volume I: Glutamate. Boca Raton, FI: CRC Press; 1995:2119–237.Google Scholar
50.Albin, RL, Greenamyre, JT. Alternative excitotoxic hypotheses. Neurology. 1992;42:733738.Google Scholar
51.Malinow, R. AMPA receptot trafficking and long-term potentiation. Philos Trans R Soc Land B Biol Sci. 2003;358:707714.Google Scholar
52.Chen, H.-SV, Pellegrini, JW, Aggarwal, SK, et al.Open-channel block of N-methyl-D-aspartate (NMDA) responses by memantine: therapeutic advantage against NMDA receptor-mediated neurotoxicity. J Neurosci. 1992;12:44274436.Google Scholar
53.Rogawski, MA. Low affinity channel blocking (uncompetitive) NMDA receptor antagonists as therapeutic agents—toward an understanding of their favorable tolerability. Amino Acids. 2000;19:133149.Google Scholar
54.Chen, HS, Wang, YF, Rayudu, PV, et al.Neuroprotective concentrations of the N-methyl-D-aspartate open-channel blocker memantine are effective without cytoplasmic vacuolation following post-ischemic administration and do not block maze learning or long-term potentiation. Neurosrience. 1998;86:11211132.Google Scholar
55.Danysz, W, Parsons, CG. The NMDA receptor antagonist memantine as a symptomatological and neuroprotective treatment for Alzheimer's disease: preclinical evidence. Int J Geriatr Psychiatry. 2003;18(suppl 1):S23S32.Google Scholar
56.Korey, SR, Scheinberg, L, Terry, R, Stein, A. Studies in presenile dementia. Trans Am Neurol Assoc. 1961;86:99102.Google Scholar
57.Hyman, BT, Van Hoesen, GW, Damasio, AR. Alzheimer's disease: glutamate depletion in the hippocampal perforant pathway zone. Ann Neurol. 1987;22:3740.Google Scholar
58.Lowe, SL, Bowen, DM, Francis, PT, Neary, D. Ante mortem cerebral amino acid concentrations indicate selective degeneration of glutamate-enriched neurons in Alzheimer's disease. Neuroscience. 1990;38:571577.Google Scholar
59.Ferrarese, C, Aliprandi, A, Tremolizzo, L, et al.Increased glutamate in CSF and plasma of patients with HIV dementia. Neurology. 2001;57:671675.Google Scholar
60.Francis, PT. Glutamatergic systems in Alzheimer's disease. Int J Geriatr Psychiatry. 2003;18:S15S21.Google Scholar
61.Danysz, W, Parsons, CG, Kornhuber, J, Schmidt, WJ, Quack, G. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents–preclinical studies. Neurosci Biobehav Rev. 1997;21:455–68.Google Scholar
62.Schmidt, WJ, Kretschmer, BD. Behavioural pharmacology of glutamate receptors in the basal ganglia. Neurosci Biobehav Rev. 1997;21:381–92.Google Scholar
63.Vollenweider, FX, Leenders, KL, Oye, I, Hell, D, Angst, J. Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET). Eur Neuropsychopharmacol. 1997;7:2538.Google Scholar
64.Vollenweider, FX, Leenders, KL, Scharfetter, C, et al.Metabolic hyperfrontality and psychopathology in the ketamine model of psychosis using positron emission tomography (PET) and [18F]fluorodeoxyglucose (FDG). Eur Neuropsychopharmacol. 1997;7:924.Google Scholar
65.Herholz, K. PET studies in dementia. Ann Nucl Med. 2003;17:7989.Google Scholar
66.Parsons, CG, Danysz, W, Quack, G. Memantine is a clinically well tolerated N-methyl-D-aspartate (NMDA) receptor antagonist—a review of preclinical data. Neuropharmocology. 1999;38:735767.Google Scholar
67.Schulz, JB, Matthews, RT, Henshaw, DR, Beal, MF. Neuroprotective strategies for treatment of lesions produced by mitochondrial toxins: implications for neurodegenerative diseases. Neuroscience. 1996;71:10431048.Google Scholar
68.Wenk, GL, Danysz, W, Mobley, SL. Investigations of neurotoxicity and neuro-protection within the nucleus basalis of the rat. Brain Res. 1994;655:711.Google Scholar
69.Wenk, GL, Danysz, W, Mobley, SL. MK-801, memantine and amantadine show neuroprotective activity in the nucleus basalis magnocellularis. Eur J Pharmacol Environ Toxicol. 1995;293:267270.Google Scholar
70.Wenk, GL, Danysz, W, Roice, DD. The effects of mitochondrial failure upon cholinergic toxicity in the nucleus basalis. Neuroreport. 1996;7:14531456.Google Scholar
71.Mattson, MP. Cellular actions of β-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol Rev. 1997;77:10811132.Google Scholar
72.Klegeris, A, McGeer, PL. β-amyloid protein enhances macrophage production of oxygen free radicals and glutamate. J Neurosci Res. 1997;49:229235.Google Scholar
73.Miguel-Hidalgo, JJ, Alvarez, XA, Cacabelos, R, Quack, G. Neuroprotection by memantine against neurodegeneration induced by β-amyloid(1-40). Brain Res. 2002;958:210221.Google Scholar