Skip to main content
Top
Published in: CNS Drugs 5/2001

01-05-2001 | Review Article

Pharmacodynamic-Tolerability Relationships of Cholinesterase Inhibitors for Alzheimer’s Disease

Author: Dr Bruno Pietro Imbimbo

Published in: CNS Drugs | Issue 5/2001

Login to get access

Abstract

According to the cholinergic hypothesis, the impairment of cognitive function and the behavioural disturbances that affect patients with Alzheimer’s disease are mainly due to cortical deficiencies in cholinergic transmission. Numerous cholinesterase inhibitors have been investigated for treatment of this disease, the rationale being to support the cholinergic system by blocking the degradation of acetylcholine released from presynaptic neurons. These drugs can be classified as reversible (tacrine, donepezil and galantamine), pseudo-reversible (physostigmine, eptastigmine and rivastigmine) or irreversible (metrifonate) enzyme inhibitors.
This article reviews efficacy and tolerability results from 6-month placebo-controlled studies of 7 cholinesterase inhibitors: tacrine (80 to 160 mg/day), donepezil (5 to 10 mg/day), rivastigmine (1 to 12 mg/day), metrifonate (30 to 80 mg/day), eptastigmine (30 to 60 mg/day), physostigmine (30 to 36 mg/day) and galantamine (8 to 32 mg/day). All these agents have demonstrated a statistically significant, although modest, effect versus placebo on the cognitive and global performance of patients with Alzheimer’s disease. Dramatic clinical response hasbeen seen in only 3 to 5% of patients. There are no major differences in terms of efficacy between the different drugs. The mean difference between drug and placebo effects on standardised psychometric scales is about 2 to 4 points on the cognitive subscale of the Alzheimer’s Disease Assessment Scale (ADAS-Cog; a 70-point cognitive scale) and 0.2 to 0.5 points on the Clinician’s Interview-Based Impression of Change with Caregiver Input (CIBIC-Plus; a 7-point global scale), or 5 to 14% of the average value of the scales.
The most common adverse effects observed after administration of cholinesterase inhibitors are nausea, vomiting, diarrhoea, dizziness, asthenia and anorexia, all symptoms linked to cholinergic overstimulation. These effects are dose related and largely depend on the degree of cholinesterase inhibition. Also important is the rate of onset of cholinesterase inhibition, which depends on the kinetics of enzyme inhibition, the presence and rate of titration, and the pharmacodynamic peak-to-trough fluctuations. A model predicting the incidence of nausea based on acetylcholinesterase inhibition and the half-life of acetylcholinesterase recovery is proposed.
In conclusion, cholinesterase inhibitors are the only pharmacological agents proved to be effective for the treatment of Alzheimer’s disease in large, long term, double-blind, placebo-controlled trials. While the efficacy of different cholinesterase inhibitors is similar, their tolerability profiles differ. For example, the incidence of nausea (in excess of that seen with placebo) at cognitively effective dosages ranges from 1% with eptastigmine 60 mg/day to 53% with physostigmine 30 mg/day. Differences in tolerability profile may be due to the extent of peripheral acetylcholinesterase inhibition needed to reach clinical efficacy. Other contributing pharmacodynamic factors are the rate of onset of and fluctuations in acetylcholinesterase inhibition at steady state.
Footnotes
1
1 This adverse effect of metrifonate is not considered to be cholinergic in nature, rather it has been linked to the delayed neurotoxicity seen with other organophosphates. [57]
 
Literature
1.
go back to reference McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA work group under the auspices of the Department of Health and Human Services task force on Alzheimer’s disease. Neurology 1984; 34: 939–44PubMedCrossRef McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA work group under the auspices of the Department of Health and Human Services task force on Alzheimer’s disease. Neurology 1984; 34: 939–44PubMedCrossRef
2.
go back to reference Davies P, Maloney AJF. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976; 2: 1403PubMedCrossRef Davies P, Maloney AJF. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976; 2: 1403PubMedCrossRef
3.
go back to reference Whitehouse PJ, Price DL, Clark AW, et al. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol 1981; 10: 122–6PubMedCrossRef Whitehouse PJ, Price DL, Clark AW, et al. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol 1981; 10: 122–6PubMedCrossRef
4.
5.
go back to reference Cummings JL. Cholinesterase inhibitors: a new class of psychotropic compounds. Am J Psychiatry 2000; 157: 4–15PubMed Cummings JL. Cholinesterase inhibitors: a new class of psychotropic compounds. Am J Psychiatry 2000; 157: 4–15PubMed
6.
go back to reference Svensson AL, Giacobini E. Cholinesterase inhibitors do more than inhibit cholinesterase. In: Giacobini E, editor. Cholines-terases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 227–35 Svensson AL, Giacobini E. Cholinesterase inhibitors do more than inhibit cholinesterase. In: Giacobini E, editor. Cholines-terases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 227–35
7.
go back to reference Thal LJ, Ferguson JM, Mintzer J, et al. A 24-week randomized trial of controlled-release physostigmine in patients with Alzheimer’s disease. Neurology 1999; 52: 1146–52PubMedCrossRef Thal LJ, Ferguson JM, Mintzer J, et al. A 24-week randomized trial of controlled-release physostigmine in patients with Alzheimer’s disease. Neurology 1999; 52: 1146–52PubMedCrossRef
8.
go back to reference Eagger SA, Harvey RJ. Clinical heterogeneity: responders to cholinergic therapy. Alzheimer Dis AssocDisord 1995; 9Suppl. 2: 37–42CrossRef Eagger SA, Harvey RJ. Clinical heterogeneity: responders to cholinergic therapy. Alzheimer Dis AssocDisord 1995; 9Suppl. 2: 37–42CrossRef
10.
go back to reference Appleyard ME. Non-cholinergic functions of acetylcholinesterase. Biochem Soc Trans 1994; 22: 749–55PubMed Appleyard ME. Non-cholinergic functions of acetylcholinesterase. Biochem Soc Trans 1994; 22: 749–55PubMed
11.
go back to reference Massoulie J, Sussman J, Bon S, et al. Structure and functions of acetylcholinesterase and butyrylcholinesterase. Prog Brain Res 1993; 98: 139–46PubMedCrossRef Massoulie J, Sussman J, Bon S, et al. Structure and functions of acetylcholinesterase and butyrylcholinesterase. Prog Brain Res 1993; 98: 139–46PubMedCrossRef
12.
go back to reference Chatonnet A, Lockridge O. Comparison of butyrylcholinesterase and acetylcholinesterase. Biochem J 1989; 260: 625–34PubMed Chatonnet A, Lockridge O. Comparison of butyrylcholinesterase and acetylcholinesterase. Biochem J 1989; 260: 625–34PubMed
13.
go back to reference Mack A, Robitzki A. The key role of butyrylcholinesterase during neurogenesis and neural disorders: an antisense-5′butyryl-cholinesterase-DNA study. Prog Neurobiol 2000; 60: 607–28PubMedCrossRef Mack A, Robitzki A. The key role of butyrylcholinesterase during neurogenesis and neural disorders: an antisense-5′butyryl-cholinesterase-DNA study. Prog Neurobiol 2000; 60: 607–28PubMedCrossRef
14.
go back to reference Guillozet AL, Smiley JF, Mash DC, et al. Butyrylcholinesterase in the life cycle of amyloid plaques. Ann Neurol 1997; 42: 909–18PubMedCrossRef Guillozet AL, Smiley JF, Mash DC, et al. Butyrylcholinesterase in the life cycle of amyloid plaques. Ann Neurol 1997; 42: 909–18PubMedCrossRef
15.
go back to reference Reiner E, Radic Z. Mechanism of action of cholinesterase inhibitors. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 103–19 Reiner E, Radic Z. Mechanism of action of cholinesterase inhibitors. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 103–19
16.
go back to reference al-Jafari AA, Kamal MA, Alhomida AS. Sensitivity of bovine retinal acetylcholinesterase (E.C. 3.1.1.7) toward tacrine: kinetic characterization. J Biochem Mol Toxicol 1998; 12: 245–51PubMedCrossRef al-Jafari AA, Kamal MA, Alhomida AS. Sensitivity of bovine retinal acetylcholinesterase (E.C. 3.1.1.7) toward tacrine: kinetic characterization. J Biochem Mol Toxicol 1998; 12: 245–51PubMedCrossRef
17.
go back to reference Kryger G, Silman I, Sussman JL. Three-dimensional structure of a complex of E2020 with acetylcholinesterase from Torpedo californica. J Physiol Paris 1998; 92: 191–4PubMedCrossRef Kryger G, Silman I, Sussman JL. Three-dimensional structure of a complex of E2020 with acetylcholinesterase from Torpedo californica. J Physiol Paris 1998; 92: 191–4PubMedCrossRef
18.
go back to reference Greenblatt HM, Kryger G, Lewis T, et al. Structure of acetylcholinesterase complexed with (−)-galanthamine at 2.3 A resolution. FEBS Lett 1999; 463: 321–6PubMedCrossRef Greenblatt HM, Kryger G, Lewis T, et al. Structure of acetylcholinesterase complexed with (−)-galanthamine at 2.3 A resolution. FEBS Lett 1999; 463: 321–6PubMedCrossRef
19.
go back to reference Thomsen T, Kaden B, Fischer JP, et al. Inhibition of acetylcholinesterase activity in human brain tissue and erythrocytes by galanthamine, physostigmine and tacrine. Eur J Clin Chem Clin Biochem 1991; 29: 487–92PubMed Thomsen T, Kaden B, Fischer JP, et al. Inhibition of acetylcholinesterase activity in human brain tissue and erythrocytes by galanthamine, physostigmine and tacrine. Eur J Clin Chem Clin Biochem 1991; 29: 487–92PubMed
20.
go back to reference Yu QS, Atack JR, Rapoport SI, et al. Carbamate analogues of (−)-physostigmine: in vitro inhibition of acetyl- and butyrylcholinesterase. FEBS Lett 1988; 234: 127–30PubMedCrossRef Yu QS, Atack JR, Rapoport SI, et al. Carbamate analogues of (−)-physostigmine: in vitro inhibition of acetyl- and butyrylcholinesterase. FEBS Lett 1988; 234: 127–30PubMedCrossRef
21.
go back to reference Brossi A, Schonenberger B, Clark OE, et al. Inhibition of acetylcholinesterase from electric eel by (−)-and (+)-physostigmine and related compounds. FEBS Lett 1986 9; 201: 190–2PubMedCrossRef Brossi A, Schonenberger B, Clark OE, et al. Inhibition of acetylcholinesterase from electric eel by (−)-and (+)-physostigmine and related compounds. FEBS Lett 1986 9; 201: 190–2PubMedCrossRef
22.
go back to reference Brufani M, Marta M, Pomponi M. Anticholinesterase activity of a new carbamate, heptylphysostigmine, in view of its use in patients with Alzheimer-type dementia. Eur J Biochem 1986; 157: 115–20PubMedCrossRef Brufani M, Marta M, Pomponi M. Anticholinesterase activity of a new carbamate, heptylphysostigmine, in view of its use in patients with Alzheimer-type dementia. Eur J Biochem 1986; 157: 115–20PubMedCrossRef
23.
go back to reference Weinstock M, Razin M, Chorev M, et al. Pharmacological evaluation of phenyl-carbamates as CNS-selective acetylcholinesterase inhibitors. J Neural Transm Suppl 1994; 43: 219–25PubMed Weinstock M, Razin M, Chorev M, et al. Pharmacological evaluation of phenyl-carbamates as CNS-selective acetylcholinesterase inhibitors. J Neural Transm Suppl 1994; 43: 219–25PubMed
24.
go back to reference Hinz VC, Grewig S, Schmidt BH. Metrifonate induces cholinesterase inhibition exclusively via slow release of dichlorvos. Neurochem Res 1996; 21: 331–7PubMedCrossRef Hinz VC, Grewig S, Schmidt BH. Metrifonate induces cholinesterase inhibition exclusively via slow release of dichlorvos. Neurochem Res 1996; 21: 331–7PubMedCrossRef
25.
go back to reference Giacobini E. Cholinesterase inhibitors: from the Calabar bean to Alzheimer therapy. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 181–226 Giacobini E. Cholinesterase inhibitors: from the Calabar bean to Alzheimer therapy. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 181–226
26.
go back to reference Tago H, Maeda T, McGeer PL, et al. Butyrylcholinesterase-rich neurons in rat brain demonstrated by a sensitive histochemical method. J Comp Neurol 1992; 325: 301–12PubMedCrossRef Tago H, Maeda T, McGeer PL, et al. Butyrylcholinesterase-rich neurons in rat brain demonstrated by a sensitive histochemical method. J Comp Neurol 1992; 325: 301–12PubMedCrossRef
27.
go back to reference Wright CI, Geula C, Mesulam MM. Neurological cholinesterases in the normal brain and in Alzheimer’s disease: relationship to plaques, tangles, and patterns of selective vulnerability. Ann Neurol 1993; 34: 373–84PubMedCrossRef Wright CI, Geula C, Mesulam MM. Neurological cholinesterases in the normal brain and in Alzheimer’s disease: relationship to plaques, tangles, and patterns of selective vulnerability. Ann Neurol 1993; 34: 373–84PubMedCrossRef
28.
go back to reference Rogers SL, Farlow MR, Doody RS, et al. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer’s disease. Neurology 1998; 50: 136–45PubMedCrossRef Rogers SL, Farlow MR, Doody RS, et al. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer’s disease. Neurology 1998; 50: 136–45PubMedCrossRef
29.
go back to reference Imbimbo BP, Martelli P, Troetel WM, et al. Efficacy and safety of eptastigmine for the treatment of patients with Alzheimer’s disease. Neurology 1999; 52: 700–8PubMedCrossRef Imbimbo BP, Martelli P, Troetel WM, et al. Efficacy and safety of eptastigmine for the treatment of patients with Alzheimer’s disease. Neurology 1999; 52: 700–8PubMedCrossRef
30.
go back to reference Morris JC, Cyrus PA, Orazem J, et al. Metrifonate benefits cognitive, behavioral, and global function in patients with Alzheimer’s disease. Neurology 1998; 50: 1222–30PubMedCrossRef Morris JC, Cyrus PA, Orazem J, et al. Metrifonate benefits cognitive, behavioral, and global function in patients with Alzheimer’s disease. Neurology 1998; 50: 1222–30PubMedCrossRef
31.
go back to reference Crismon ML. Pharmacokinetics and drug interactions of cholinesterase inhibitors administered in Alzheimer’s disease. Pharmacotherapy 1998; 18 (2 Pt 2): 47–54PubMed Crismon ML. Pharmacokinetics and drug interactions of cholinesterase inhibitors administered in Alzheimer’s disease. Pharmacotherapy 1998; 18 (2 Pt 2): 47–54PubMed
32.
go back to reference Lotti M. Cholinesteras inhibition: complexities in interpretation. Clin Chem 1995; 41: 1814–8PubMed Lotti M. Cholinesteras inhibition: complexities in interpretation. Clin Chem 1995; 41: 1814–8PubMed
33.
34.
go back to reference Rogers SL, Doody RS, Mohs RC, et al. Donepezil improves cognition and global function in Alzheimer’s disease: a 15-week, double-blind, placebo-controlled study. Donepezil Study Group. Arch Intern Med 1998; 158: 1021–31PubMedCrossRef Rogers SL, Doody RS, Mohs RC, et al. Donepezil improves cognition and global function in Alzheimer’s disease: a 15-week, double-blind, placebo-controlled study. Donepezil Study Group. Arch Intern Med 1998; 158: 1021–31PubMedCrossRef
35.
go back to reference Cummings JL, Cyrus PA, Bieber F, et al. Metrifonate treatment of the cognitive deficits of Alzheimer’s disease. Metrifonate Study Group. Neurology 1998; 50: 1214–21PubMedCrossRef Cummings JL, Cyrus PA, Bieber F, et al. Metrifonate treatment of the cognitive deficits of Alzheimer’s disease. Metrifonate Study Group. Neurology 1998; 50: 1214–21PubMedCrossRef
36.
go back to reference Imbimbo BP, Troetel WM, Martelli P, et al. A 6-month, double-blind, placebo-controlled trial of eptastigmine in Alzheimer’s disease. Dement Geriatr Cogn Disord 2000; 11: 17–23PubMedCrossRef Imbimbo BP, Troetel WM, Martelli P, et al. A 6-month, double-blind, placebo-controlled trial of eptastigmine in Alzheimer’s disease. Dement Geriatr Cogn Disord 2000; 11: 17–23PubMedCrossRef
37.
go back to reference Wilcock G, Wilkinson D. Galantamine hydrobromide: interim results of a group comparative, placebo-controlled study of efficacy and safety in patients with a diagnosis of senile dementia of the Alzheimer type. In: Iqbal K, Winblad B, Nishimura T, et al, editors. Alzheimer’s disease: biology, diagnosis and therapeutics. New York (NY): John Wiley & Sons Ltd, 1997: 661–4 Wilcock G, Wilkinson D. Galantamine hydrobromide: interim results of a group comparative, placebo-controlled study of efficacy and safety in patients with a diagnosis of senile dementia of the Alzheimer type. In: Iqbal K, Winblad B, Nishimura T, et al, editors. Alzheimer’s disease: biology, diagnosis and therapeutics. New York (NY): John Wiley & Sons Ltd, 1997: 661–4
38.
go back to reference Knapp MJ, Knopman DS, Solomon PR, et al. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer’s disease. The Tacrine Study Group. JAMA 1994; 271:985–91PubMedCrossRef Knapp MJ, Knopman DS, Solomon PR, et al. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer’s disease. The Tacrine Study Group. JAMA 1994; 271:985–91PubMedCrossRef
39.
go back to reference Burns A, Rossor M, Hecker J, et al. The effects of donepezil in Alzheimer’s disease — results from a multinational trial. Dement Geriatr Cogn Disord 1999; 10: 237–44PubMedCrossRef Burns A, Rossor M, Hecker J, et al. The effects of donepezil in Alzheimer’s disease — results from a multinational trial. Dement Geriatr Cogn Disord 1999; 10: 237–44PubMedCrossRef
40.
go back to reference Rosier 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: 633–8 Rosier 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: 633–8
41.
go back to reference Corey-Bloom J, Anand R, Veach J. A randomized trial evaluating the efficacy and safety of ENA 713 (rivastigmine tartrate), a new acetylcholinesterase inhibitor, in patients with mild to moderately severe Alzheimer’s disease. Int J Geriat Psychopharmacol 1998; 1: 55–65 Corey-Bloom J, Anand R, Veach J. A randomized trial evaluating the efficacy and safety of ENA 713 (rivastigmine tartrate), a new acetylcholinesterase inhibitor, in patients with mild to moderately severe Alzheimer’s disease. Int J Geriat Psychopharmacol 1998; 1: 55–65
42.
go back to reference Dubois B, McKeith I, Orgogozo JM, et al. A multicentre, randomized, double-blind, placebo-controlled study to evaluate the efficacy, tolerability and safety of two doses of metrifonate in patients with mild-to-moderate Alzheimer’s disease: the MALT study. Int J Geriatr Psychiatry 1999; 14: 973–82PubMedCrossRef Dubois B, McKeith I, Orgogozo JM, et al. A multicentre, randomized, double-blind, placebo-controlled study to evaluate the efficacy, tolerability and safety of two doses of metrifonate in patients with mild-to-moderate Alzheimer’s disease: the MALT study. Int J Geriatr Psychiatry 1999; 14: 973–82PubMedCrossRef
43.
go back to reference Tariot PN, Solomon PR, Morris JC, et al. A 5-month, randomized, placebo-controlled trial of galantamine in AD. Neurology 2000; 54: 2269–76PubMedCrossRef Tariot PN, Solomon PR, Morris JC, et al. A 5-month, randomized, placebo-controlled trial of galantamine in AD. Neurology 2000; 54: 2269–76PubMedCrossRef
44.
go back to reference Raskind MA, Peskind ER, Wessel T, et al. Galantamine in AD: a 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology 2000; 54: 2261–8PubMedCrossRef Raskind MA, Peskind ER, Wessel T, et al. Galantamine in AD: a 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology 2000; 54: 2261–8PubMedCrossRef
45.
go back to reference Wilcock GK, Lilienfled S, Gaens E. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer’s disease: multicentre randomised controlled trial. BMJ 2000; 321: 1445–9PubMedCrossRef Wilcock GK, Lilienfled S, Gaens E. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer’s disease: multicentre randomised controlled trial. BMJ 2000; 321: 1445–9PubMedCrossRef
46.
go back to reference Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of tacrine administration in patients with Alzheimer’s disease. JAMA 1994; 271: 992–8PubMedCrossRef Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of tacrine administration in patients with Alzheimer’s disease. JAMA 1994; 271: 992–8PubMedCrossRef
47.
go back to reference Cultler NR, Sramek JJ. The role of bridging studies in the development of cholinesterase inhibitors for Alzheimer’s disease. CNS Drugs 1998; 10: 355–64CrossRef Cultler NR, Sramek JJ. The role of bridging studies in the development of cholinesterase inhibitors for Alzheimer’s disease. CNS Drugs 1998; 10: 355–64CrossRef
48.
go back to reference Nordberg A, Svensson AL. Cholinesterase inhibitors in the treatment of Alzheimer’s disease: a comparison of tolerability and pharmacology. Drug Safety 1998; 19: 465–80PubMedCrossRef Nordberg A, Svensson AL. Cholinesterase inhibitors in the treatment of Alzheimer’s disease: a comparison of tolerability and pharmacology. Drug Safety 1998; 19: 465–80PubMedCrossRef
49.
50.
go back to reference Gracon SI, Knapp MJ, Berghoff WG, et al. Safety of tacrine: clinical trials, treatment IND, and postmarketing experience. Alzheimer Dis Assoc Disord 1998; 12: 93–101PubMedCrossRef Gracon SI, Knapp MJ, Berghoff WG, et al. Safety of tacrine: clinical trials, treatment IND, and postmarketing experience. Alzheimer Dis Assoc Disord 1998; 12: 93–101PubMedCrossRef
51.
go back to reference Hashimoto M, Imamura T, Tanimukai S, et al. Urinary incontinence: an unrecognised adverse effect with donepezil. Lancet 2000; 356: 568PubMedCrossRef Hashimoto M, Imamura T, Tanimukai S, et al. Urinary incontinence: an unrecognised adverse effect with donepezil. Lancet 2000; 356: 568PubMedCrossRef
52.
go back to reference Bourke D, Druckenbrod RW. Possible association between donepezil and worsening Parkinson’s disease. Ann Pharmacother 1998; 32: 610–1PubMedCrossRef Bourke D, Druckenbrod RW. Possible association between donepezil and worsening Parkinson’s disease. Ann Pharmacother 1998; 32: 610–1PubMedCrossRef
53.
go back to reference Ross JS, Shua-Haim JR. Aricept-induced nightmares in Alzheimer’s disease: 2 case reports. J Am Geriatr Soc 1998; 46: 119–20PubMed Ross JS, Shua-Haim JR. Aricept-induced nightmares in Alzheimer’s disease: 2 case reports. J Am Geriatr Soc 1998; 46: 119–20PubMed
54.
go back to reference Bryant CA, Ouldred E, Jackson SHD, et al. Purpuric rash with donepezil treatment. BMJ 1998; 317: 787PubMedCrossRef Bryant CA, Ouldred E, Jackson SHD, et al. Purpuric rash with donepezil treatment. BMJ 1998; 317: 787PubMedCrossRef
55.
go back to reference Lang IM. Noxious stimulation of emesis. Dig Dis Sci 1999; 44(8 Suppl.): 58S–63SPubMed Lang IM. Noxious stimulation of emesis. Dig Dis Sci 1999; 44(8 Suppl.): 58S–63SPubMed
56.
go back to reference Stewart JJ, Burks TF, Weisbrodt NW. Intestinal myoelectric activity after activation of central emetic mechanism. Am J Physiol 1977; 233: E131–7PubMed Stewart JJ, Burks TF, Weisbrodt NW. Intestinal myoelectric activity after activation of central emetic mechanism. Am J Physiol 1977; 233: E131–7PubMed
57.
go back to reference Furman JM, Whitney SL. Central causes of dizziness. Phys Ther 2000; 80: 179–87PubMed Furman JM, Whitney SL. Central causes of dizziness. Phys Ther 2000; 80: 179–87PubMed
58.
go back to reference Lazartigues E, Freslon JL, Tellioglu T, et al. Pressor and brady-cardic effects of tacrine and other acetylcholinesterase inhibitors in the rat. Eur J Pharmacol 1998; 361: 61–71PubMedCrossRef Lazartigues E, Freslon JL, Tellioglu T, et al. Pressor and brady-cardic effects of tacrine and other acetylcholinesterase inhibitors in the rat. Eur J Pharmacol 1998; 361: 61–71PubMedCrossRef
59.
go back to reference Karczmar AG. Synaptic, behavioral, and toxicological effects of cholinesterase inhibitors in animals and humans. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 157–80 Karczmar AG. Synaptic, behavioral, and toxicological effects of cholinesterase inhibitors in animals and humans. In: Giacobini E, editor. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz, 2000: 157–80
60.
go back to reference Enz A, Amstutz R, Boddeke H, et al. Brain selective inhibition of acetylcholinesterase: a novel approach to therapy for Alz-heimer’s disease. Prog Brain Res 1993; 98: 431–8PubMedCrossRef Enz A, Amstutz R, Boddeke H, et al. Brain selective inhibition of acetylcholinesterase: a novel approach to therapy for Alz-heimer’s disease. Prog Brain Res 1993; 98: 431–8PubMedCrossRef
61.
go back to reference Imbimbo BP, Lucca U, Lucchelli F, et al. A 25-week placebocontrolled study of eptastigmine in patients with Alzheimer’s disease. Alzheimer Dis Assoc Disord 1998; 12: 313–22PubMedCrossRef Imbimbo BP, Lucca U, Lucchelli F, et al. A 25-week placebocontrolled study of eptastigmine in patients with Alzheimer’s disease. Alzheimer Dis Assoc Disord 1998; 12: 313–22PubMedCrossRef
62.
go back to reference Knapp S, Wardlow L, Albert K, et al. Correlation between plasma physostigmine concentrations and percentage of acetylcholinesterase over time after controlled release of physostigmine in volunteer subjects. Drug Metab Dispos 1991; 19: 400–4PubMed Knapp S, Wardlow L, Albert K, et al. Correlation between plasma physostigmine concentrations and percentage of acetylcholinesterase over time after controlled release of physostigmine in volunteer subjects. Drug Metab Dispos 1991; 19: 400–4PubMed
63.
go back to reference Bickel U, Thomsen T, Weber W, et al. Pharmacokinetics of galanthamine in humans and corresponding cholinesterase inhibition. Clin Pharmacol Ther 1991; 50: 420–8PubMedCrossRef Bickel U, Thomsen T, Weber W, et al. Pharmacokinetics of galanthamine in humans and corresponding cholinesterase inhibition. Clin Pharmacol Ther 1991; 50: 420–8PubMedCrossRef
64.
go back to reference Tiseo PJ, Rogers SL, Friedhoff LT. Pharmacokinetic and pharmacodynamic profile of donepezil HC1 following evening administration. Br J Clin Pharmacol 1998; 46Suppl. 1: 13–8PubMed Tiseo PJ, Rogers SL, Friedhoff LT. Pharmacokinetic and pharmacodynamic profile of donepezil HC1 following evening administration. Br J Clin Pharmacol 1998; 46Suppl. 1: 13–8PubMed
65.
go back to reference Imbimbo BP, Licini M, Schettino M, et al. Relationship between pharmacokinetics and pharmacodynamics of eptastigmine in young healthy volunteers. J Clin Pharmacol 1995; 35: 285–90PubMed Imbimbo BP, Licini M, Schettino M, et al. Relationship between pharmacokinetics and pharmacodynamics of eptastigmine in young healthy volunteers. J Clin Pharmacol 1995; 35: 285–90PubMed
66.
go back to reference Kennedy JS, Polinsky RJ, Johnson B, et al. Preferential cerebrospinal fluid acetylcholinesterase inhibition by rivastigmine in humans. J Clin Psychopharmacol 1999; 19: 513–21PubMedCrossRef Kennedy JS, Polinsky RJ, Johnson B, et al. Preferential cerebrospinal fluid acetylcholinesterase inhibition by rivastigmine in humans. J Clin Psychopharmacol 1999; 19: 513–21PubMedCrossRef
67.
go back to reference Ray DE. Chronic effects of low level exposure to anticholines-terases — a mechanistic review. Toxicol Lett 1998; 102-103: 527–33PubMedCrossRef Ray DE. Chronic effects of low level exposure to anticholines-terases — a mechanistic review. Toxicol Lett 1998; 102-103: 527–33PubMedCrossRef
68.
go back to reference Madden S, Spaldin V, Park BK. Clinical pharmacokinetics of tacrine. Clin Pharmacokinet 1995; 28: 449–57PubMedCrossRef Madden S, Spaldin V, Park BK. Clinical pharmacokinetics of tacrine. Clin Pharmacokinet 1995; 28: 449–57PubMedCrossRef
69.
go back to reference Johansson M, Hellstrom-Lindahl E, Nordberg A. Steady-state pharmacokinetics of tacrine in long-term treatment of Alzheimer patients. Dementia 1996; 7:111–7PubMed Johansson M, Hellstrom-Lindahl E, Nordberg A. Steady-state pharmacokinetics of tacrine in long-term treatment of Alzheimer patients. Dementia 1996; 7:111–7PubMed
70.
go back to reference Rogers SL, Cooper NM, Sukovaty R, et al. Pharmacokinetic and pharmacodynamic profile of donepezil HC1 following multiple oral doses. Br J Clin Pharmacol 1998; 46 Suppl. 1: 7–12 Rogers SL, Cooper NM, Sukovaty R, et al. Pharmacokinetic and pharmacodynamic profile of donepezil HC1 following multiple oral doses. Br J Clin Pharmacol 1998; 46 Suppl. 1: 7–12
71.
go back to reference Polinsky RJ. Clinical pharmacology of rivastigmine: a new-generation acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Clin Ther 1998; 20: 634–47PubMedCrossRef Polinsky RJ. Clinical pharmacology of rivastigmine: a new-generation acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Clin Ther 1998; 20: 634–47PubMedCrossRef
72.
go back to reference Pettigrew LC, Bieber F, Lettieri J, et al. Pharmacokinetics, pharmacodynamics, and safety of metrifonate in patients with Alzheimer’s disease. J Clin Pharmacol 1998; 38: 236–45PubMed Pettigrew LC, Bieber F, Lettieri J, et al. Pharmacokinetics, pharmacodynamics, and safety of metrifonate in patients with Alzheimer’s disease. J Clin Pharmacol 1998; 38: 236–45PubMed
73.
go back to reference Asthana S, Greig NH, Hegedus L, et al. Clinical pharmacokinetics of physostigmine in patients with Alzheimer’s disease. Clin Pharmacol Ther 1995; 58: 299–309PubMedCrossRef Asthana S, Greig NH, Hegedus L, et al. Clinical pharmacokinetics of physostigmine in patients with Alzheimer’s disease. Clin Pharmacol Ther 1995; 58: 299–309PubMedCrossRef
74.
go back to reference Cutler NR, Polinsky RJ, Sramek JJ, et al. Dose-dependent CSF acetylcholinesterase inhibition by SDZ ENA 713 in Alzheimer’s disease. Acta Neurol Scand 1998; 97: 244–50PubMedCrossRef Cutler NR, Polinsky RJ, Sramek JJ, et al. Dose-dependent CSF acetylcholinesterase inhibition by SDZ ENA 713 in Alzheimer’s disease. Acta Neurol Scand 1998; 97: 244–50PubMedCrossRef
75.
go back to reference Unni L, Vicari S, Moriearty P, et al. The recovery of cerebrospinal fluid acetylcholinesterase activity in Alzheimer’s disease patients after treatment with metrifonate. Methods Find Exp Clin Pharmacol 2000; 22: 57–61PubMedCrossRef Unni L, Vicari S, Moriearty P, et al. The recovery of cerebrospinal fluid acetylcholinesterase activity in Alzheimer’s disease patients after treatment with metrifonate. Methods Find Exp Clin Pharmacol 2000; 22: 57–61PubMedCrossRef
76.
go back to reference Schmidt BH, Heinig R. The pharmacological basis for metrifonate’s favourable tolerability in the treatment of Alzheimer’s disease. Dement Geriatr Cogn Disord 1998; 9 Suppl. 2: 15–9 Schmidt BH, Heinig R. The pharmacological basis for metrifonate’s favourable tolerability in the treatment of Alzheimer’s disease. Dement Geriatr Cogn Disord 1998; 9 Suppl. 2: 15–9
77.
go back to reference Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64PubMed Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64PubMed
78.
go back to reference Schneider LS, Olin JT, Doody RS, et al. Validity and reliability of the Alzheimer’s Disease Cooperative Study-Clinical Global Impression of Change. Alzheimer Dis Assoc Disord 1997; 11(Suppl. 2): S22–S32PubMedCrossRef Schneider LS, Olin JT, Doody RS, et al. Validity and reliability of the Alzheimer’s Disease Cooperative Study-Clinical Global Impression of Change. Alzheimer Dis Assoc Disord 1997; 11(Suppl. 2): S22–S32PubMedCrossRef
79.
go back to reference Little R, Yau L. Intent-to-treat analysis for longitudinal studies with drop-outs. Biometrics 1996; 52: 1324–33PubMedCrossRef Little R, Yau L. Intent-to-treat analysis for longitudinal studies with drop-outs. Biometrics 1996; 52: 1324–33PubMedCrossRef
80.
go back to reference Flood JF, Landry DW, Jarvik ME. Cholinergic receptor interactions and their effects on long-term memory processing. Brain Res 1981; 215: 177–85PubMedCrossRef Flood JF, Landry DW, Jarvik ME. Cholinergic receptor interactions and their effects on long-term memory processing. Brain Res 1981; 215: 177–85PubMedCrossRef
81.
go back to reference Haroutunian V, Barnes E, Davis KL. Cholinergic modulation of memory in rats. Psychopharmacology 1985; 87: 266–71PubMedCrossRef Haroutunian V, Barnes E, Davis KL. Cholinergic modulation of memory in rats. Psychopharmacology 1985; 87: 266–71PubMedCrossRef
82.
go back to reference Nabeshima T, Yoshida S, Kameyama T. Effects of the novel compound NIK-247 on impairment of passive avoidance response in mice. Eur J Pharmacol 1988; 154: 263–9PubMedCrossRef Nabeshima T, Yoshida S, Kameyama T. Effects of the novel compound NIK-247 on impairment of passive avoidance response in mice. Eur J Pharmacol 1988; 154: 263–9PubMedCrossRef
83.
go back to reference Yamazaki N, Kato K, Kurihara E, et al. Cholinergic drugs reverse AF64A-induced impairment of passive avoidance learning in rats. Psychopharmacology 1991; 103: 215–22PubMedCrossRef Yamazaki N, Kato K, Kurihara E, et al. Cholinergic drugs reverse AF64A-induced impairment of passive avoidance learning in rats. Psychopharmacology 1991; 103: 215–22PubMedCrossRef
84.
go back to reference Wanibuchi F, Nishida T, Yamashita H, et al. Characterization of novel muscarinic receptor agonist, YM796: comparison with cholinesterase inhibitors in in vivo pharmacological studies. Eur J Pharmacol 1994; 265: 151–8PubMedCrossRef Wanibuchi F, Nishida T, Yamashita H, et al. Characterization of novel muscarinic receptor agonist, YM796: comparison with cholinesterase inhibitors in in vivo pharmacological studies. Eur J Pharmacol 1994; 265: 151–8PubMedCrossRef
85.
go back to reference Braida D, Paladini E, Griffini P, et al. An inverted U-shaped curve for heptylphysostigmine on radial maze performance in rats: comparison with other cholinesterase inhibitors. Eur J Pharmacol 1996; 302: 13–20PubMedCrossRef Braida D, Paladini E, Griffini P, et al. An inverted U-shaped curve for heptylphysostigmine on radial maze performance in rats: comparison with other cholinesterase inhibitors. Eur J Pharmacol 1996; 302: 13–20PubMedCrossRef
86.
go back to reference Marchi M, Paudice P, Raiteri M. Autoregulation of acetylcholine release in isolated hippocampal nerve endings. Eur J Pharmacol 1981; 73: 75–9PubMedCrossRef Marchi M, Paudice P, Raiteri M. Autoregulation of acetylcholine release in isolated hippocampal nerve endings. Eur J Pharmacol 1981; 73: 75–9PubMedCrossRef
87.
go back to reference Consolo S, Ladinsky H, Vinci R, et al. An in vivo pharmacological study on muscarinic receptor subtypes regulating cholinergic neurotransmission in rat striatum. Biochem Pharmacol 1987; 36: 3075–7PubMedCrossRef Consolo S, Ladinsky H, Vinci R, et al. An in vivo pharmacological study on muscarinic receptor subtypes regulating cholinergic neurotransmission in rat striatum. Biochem Pharmacol 1987; 36: 3075–7PubMedCrossRef
88.
go back to reference Messamore E, Warpman U, Williams E, et al. Muscarinic receptors mediate attenuation of extracellular acetylcholine levels in rat cerebral cortex after cholinesterase inhibition. Neurosci Lett 1993; 158: 205–8PubMedCrossRef Messamore E, Warpman U, Williams E, et al. Muscarinic receptors mediate attenuation of extracellular acetylcholine levels in rat cerebral cortex after cholinesterase inhibition. Neurosci Lett 1993; 158: 205–8PubMedCrossRef
Metadata
Title
Pharmacodynamic-Tolerability Relationships of Cholinesterase Inhibitors for Alzheimer’s Disease
Author
Dr Bruno Pietro Imbimbo
Publication date
01-05-2001
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 5/2001
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.2165/00023210-200115050-00004

Other articles of this Issue 5/2001

CNS Drugs 5/2001 Go to the issue

Therapy in Practice

Psychogenic Excoriation