Sie sind auf Seite 1von 21

DRUG DISPOSITION Clin Pharmacokinet 2002; 41 (10): 719-739

0312-5963/02/0010-0719/$25.00/0

© Adis International Limited. All rights reserved.

Clinical Pharmacokinetics and


Pharmacodynamics of
Cholinesterase Inhibitors
Michael W. Jann,1 Kara L. Shirley1,2 and Gary W. Small 3
1 Department of Clinical and Administrative Sciences, Southern School of Pharmacy,
Mercer University, Atlanta, Georgia, USA
2 Department of Pharmacy Practice, Albany College of Pharmacy, Albany, New York, USA
3 Department of Psychiatry and Biobehavioral Sciences, Center on Aging and Alzheimer’s Disease
Research Center, University of California, Los Angeles, California, USA

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 720
1. Pharmacokinetic Aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
1.1 Tacrine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
1.1.1 Absorption, Bioavailability and Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . 721
1.1.2 Metabolism and Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
1.1.3 Effects of Age, Hepatic and Renal Impairment . . . . . . . . . . . . . . . . . . . . . . . 723
1.2 Donepezil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
1.2.1 Absorption, Bioavailability and Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . 723
1.2.2 Metabolism and Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 724
1.2.3 Effects of Age, Hepatic and Renal Impairment . . . . . . . . . . . . . . . . . . . . . . . 724
1.3 Rivastigmine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 724
1.3.1 Absorption, Bioavailability and Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . 724
1.3.2 Metabolism and Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725
1.3.3 Effects of Age, Hepatic and Renal Impairment . . . . . . . . . . . . . . . . . . . . . . . 725
1.4 Galantamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725
1.4.1 Absorption, Bioavailability and Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . 725
1.4.2 Metabolism and Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725
1.4.3 Effects of Age and Hepatic or Renal Impairment . . . . . . . . . . . . . . . . . . . . . . 726
2. Drug Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726
2.1 Tacrine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726
2.1.1 Influence of Cytochrome P450 1A2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726
2.1.2 Cimetidine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726
2.1.3 Hormone Replacement Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.1.4 Fluvoxamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.1.5 Theophylline . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.2 Donepezil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.2.1 Cimetidine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.2.2 Digoxin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.2.3 Ketoconazole . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
2.2.4 Psychotropic Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728
2.2.5 Theophylline and Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728
2.3 Rivastigmine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728
720 Jann et al.

2.4 Galantamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728


3. Pharmacological Aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 729
3.1 Reversible versus Irreversible Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 729
3.2 Acetylcholinesterase versus Butyrylcholinesterase Inhibition . . . . . . . . . . . . . . . . . . . 729
3.3 Dose versus Cholinesterase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 730
4. Pharmacodynamic Aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 731
4.1 Clinical Efficacy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 731
4.1.1 Clinical Rating Scales . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 731
4.1.2 Correlation of Clinical Efficacy, Dose and Enzyme Inhibition . . . . . . . . . . . . . . . . 732
4.1.3 Effects on Behavioural Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 733
4.1.4 Imaging Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 734
4.2 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 735
4.2.1 Dose-Dependent Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 735
4.2.2 Dose Titration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 735
4.2.3 Serum Concentration Relationships . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 736
4.2.4 Hepatotoxicity with Tacrine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 736
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 737

Abstract Cholinesterase inhibitors are the ‘first-line’ agents in the treatment of Alz-
heimer’s disease. This article presents the latest information on their pharmaco-
kinetic properties and pharmacodynamic activity.
Tacrine was the first cholinesterase inhibitor approved by regulatory agencies,
followed by donepezil, rivastigmine and recently galantamine. With the excep-
tion of low doses of tacrine, the cholinesterase inhibitors exhibit a linear relation-
ship between dose and area under the plasma concentration-time curve.
Cholinesterase inhibitors are rapidly absorbed through the gastrointestinal tract,
with time to peak concentration usually less than 2 hours; donepezil has the
longest absorption time of 3 to 5 hours. Donepezil and tacrine are highly protein
bound, whereas protein binding of rivastigmine and galantamine is less than 40%.
Tacrine is metabolised by hepatic cytochrome P450 (CYP) 1A2, and donepezil
and galantamine are metabolised by CYP3A4 and CYP2D6. Rivastigmine is
metabolised by sulfate conjugation.
Two cholinesterase enzymes are present in the body, acetylcholinesterase
(AChE) and butyrylcholinesterase (BChE). Tacrine and rivastigmine inhibit both
enzymes, whereas donepezil and galantamine specifically inhibit AChE.
Galantamine also modulates nicotine receptors, thereby enhancing acetyl-
cholinergic activity at the synapse. These different pharmacological profiles pro-
vide distinctions between these agents. Cholinesterase inhibitors show a nonlinear
relationship between dose and cholinesterase inhibition, where a plateau effect
occurs. Cholinesterase inhibitors display a different profile as each agent achieves
its plateau at different doses.
In clinical trials, cholinesterase inhibitors demonstrate a dose-dependent effect
on cognition and functional activities. Improvement in behavioural symptoms
also occurs, but without a dose-response relationship. Gastrointestinal adverse
events are dose-related. Clinical improvement occurs with between 40 and 70%
inhibition of cholinesterase. A conceptual model for cholinesterase inhibitors has
been proposed, linking enzyme inhibition, clinical efficacy and adverse effects.
Currently, measurement of enzyme inhibition is used as the biomarker for cho-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 721

linesterase inhibitors. New approaches to determining the efficacy of cholines-


terase inhibitors in the brain could involve the use of various imaging techniques.
The knowledge base for the pharmacokinetics and pharmacodynamics of cho-
linesterase inhibitors continues to expand. The increased information available
to clinicians can optimise the use of these agents in the management of patients
with Alzheimer’s disease.

Alzheimer’s disease (AD) is a chronic neuro- hibitors in detail with respect to their pharmaco-
degenerative disorder that causes decline in cogni- kinetic, pharmacological and pharmacodynamic
tive function, behavioural disturbances and wors- properties. Eptastigmine will be discussed only
ening of the ability to perform daily activities. briefly in supporting the concept of cholinesterase
Over the past decade, a tremendous amount of inhibitors. Other cholinesterase inhibitors, such as
progress has been made in identifying the molecu- metrifonate, physostigmine and velnacrine, have
lar origins of this disease and its genetic linkage.[1] been investigated clinically; however, these agents
However, despite these advances, a cure for this were not approved by regulatory agencies because
disease remains to be found and, as for many of safety issues and will not be extensively dis-
chronic diseases, medications currently available cussed in this paper.
treat the symptoms of AD with an overall goal of
maintaining functional and cognitive ability and 1. Pharmacokinetic Aspects
improving behavioural disturbances. A summary of the pharmacokinetic parameters
Many different therapeutic approaches to AD of the cholinesterase inhibitors is presented in table
have been investigated, including antioxidants, I. With the exception of donepezil, most cholines-
anti-inflammatory agents, calcium channel antag- terase inhibitors are rapidly absorbed and possess
onists, and muscarinic and nicotinic agonists.[2-7] elimination half-lives of ≤12 hours.[10] The phar-
The use of these agents stems from the heteroge- macokinetics and pharmacodynamics vary be-
neous aetiologies associated with the disease. The tween these agents which results in different dos-
cholinergic hypothesis of AD was proposed over ing regimens or the rationale for switching drugs
20 years ago, and deterioration of cholinergic neu- during treatment. Cholinesterase inhibitors usually
rons over time for various biological reasons re- display dose-dependent linear pharmacokinetics;
mains a consistent finding.[8] however, specific details are provided on each
Only one class of medications has been exten- agent in subsequent sections.
sively evaluated in placebo-controlled trials with
AD patients and is approved by the US Food and 1.1 Tacrine
Drug Administration (FDA) and other governmen-
tal agencies for the treatment of AD. These medi- Tacrine was the first medication approved by
cations are the cholinesterase inhibitors (figure 1). the FDA for the treatment of Alzheimer’s disease.
Basically, cholinesterase inhibitors prevent the Although tacrine improved psychomotor test scores
breakdown of acetylcholine by inhibiting the ac- in mild to moderately impaired patients, adverse
tivity of cholinesterase enzymes that metabolise effects were frequent and significantly elevated he-
acetylcholine. These enzymes are known as acetyl- patic transaminase levels occurred in approxi-
cholinesterase (AChE) and butyrylcholinesterase mately 25% of patients.[12]
(BChE).[9] 1.1.1 Absorption, Bioavailability and Distribution
Tacrine was the first cholinesterase inhibitor The pharmacokinetic properties of tacrine have
approved in the US and Europe, followed by been evaluated following short- and long-term ad-
donepezil, rivastigmine and galantamine. The aim ministration.[13-16] Large interindividual variations
of this paper is to compare the cholinesterase in- exist in the pharmacokinetic profile of tacrine, which

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
722 Jann et al.

NH2
CH3
N O
C

O
N N N

Tacrine Eptastigmine C C

CH3 O CH3
O
N O
OH CH3

N CH3
CH3 N
CH3 CH3

Galantamine Rivastigmine

OCH3

N OCH3
O

Donepezil

Fig. 1. Structure of the current cholinesterase inhibitors used in the treatment of Alzheimer’s disease.

affect both efficacy and occurrence of adverse tients with AD.[21] Tacrine dosages ranged be-
events. The bioavailability of tacrine ranges from tween 40 and 140 mg/day. Mean plasma and CSF
17 to 37%.[17,18] concentrations were 8.01 ± 7.07 and 5.21 ± 6.00
The rate and extent [area under the concentra- μg/L, respectively. The mean ratio of CSF to plas-
tion-time curve (AUC)] of absorption of tacrine is ma concentrations was 0.50 ± 0.45, with no rela-
decreased by about 25% when it is given with food tionship to dose.
or within 2 hours after food intake.[12,18] When Nonlinear disposition at low doses may also
taken 1 hour before meals, absorption is not af- contribute towards the wide interpatient variability
fected. In patients with AD, time to reach peak of tacrine. In comparing single doses of 25 and
plasma concentration (tmax) was 0.5 to 3.0 hours 50mg, their respective mean AUCs were 29.0 ± 6.0
following oral administration. Tacrine appears to and 83.2 ± 26.7 μg • h/L.[14] With repeated 50mg
have a wide tissue distribution, which is reflected doses given three times a day for 1 month, the mean
by its large volume of distribution of 3.7 to 5.0 AUC after the last morning dose was 143.6 ± 68.4
L/kg.[19] Tacrine has also been found to be about μg • h/L. In another study comparing tacrine dos-
75% bound to plasma proteins (42% for albumin age and AUC under steady-state conditions, it was
and 23% for α1-acid glycoprotein).[20] found that the mean (SD not provided) AUC in-
Tacrine concentrations were compared between creased from 19.7 to 82.9 μg • h/L for dosages of
plasma and cerebrospinal fluid (CSF) in seven pa- 40 and 80 mg/day, respectively. A smaller increase

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 723

in AUC, up to 139.0 μg • h/L, was noted for the 1.1.3 Effects of Age, Hepatic and
120 mg/day dosage.[15] Although the mechanism Renal Impairment
of the nonlinear metabolism is uncertain, it was Tacrine pharmacokinetics were reported to be
unaffected in elderly patients with renal insuffi-
suggested that the lower bioavailability at lower
ciency.[12] Because of the hepatotoxicity of tacr-
doses may reflect saturable hepatic first-pass in-
ine, most clinicians agree that patients with hepatic
take of tacrine.[15,17]
impairment would not be suitable candidates for
1.1.2 Metabolism and Elimination treatment.
Tacrine is rapidly and extensively metabolised
by cytochrome P450 (CYP) isoenzymes 1A2 and 1.2 Donepezil
2D6. Five major metabolites have been found in
Donepezil was the next cholinesterase inhibitor
serum and urine.[19] Tacrine reactive metabolites
to be available. Donepezil is a selective reversible
then undergo glutathione conjugation.[22] The pre- inhibitor for AChE rather than BChE, as shown by
sence of genetic deficiencies in the glutathione S- comparison with tacrine and physostigmine in in
transferase system was found to increase suscepti- vitro and ex vivo rat studies.[25]
bility to tacrine hepatotoxicity in patients with
mild to moderate AD.[22] The elimination half-life 1.2.1 Absorption, Bioavailability and Distribution
(t1⁄2β) of tacrine has been observed to range from After oral administration, peak plasma done-
1.3 to 7 hours in patients with AD,[14,15,17,18] and pezil concentrations are achieved after 3 to 5 hours
most investigators agree on a value of about 4 (table I), which is longer than for other cholines-
hours.[13,23] terase inhibitors. Food does not affect its absorp-
tion rate, Cmax, tmax or AUC. The relationship be-
The major tacrine metabolites include phenol
tween donepezil dose and AUC is linear over the
tacrines and 1-, 2-, 4- and 7-hydroxy-tacrine.[17]
dosage range 1 to 10 mg/day.[26,27] Under steady-
The major metabolite, 1-hydroxy-tacrine, is pres-
state conditions, the mean AUCs for the 1, 3 and
ent in plasma and is found in the CSF at a concen-
5mg doses were 118.5 ± 52.3, 357.7 ± 64.0 and
tration 10 times higher than that of tacrine.[18] The 546.0 ± 52.6 μg • h/L, respectively.[28] Under clini-
t1⁄2β of the 1-hydroxy-tacrine metabolite appears to cal trial conditions, mean trough (Cmin) plasma
be similar to that of the parent compound.[17] Its drug concentrations were 25.9 ± 0.7 and 50.6 ± 1.9
formation and elimination are stereospecific, μg/L with therapeutic doses of 5 and 10mg, respec-
favouring the dextrorotary isomer. The metabo- tively.[29]
lism of 1-hydroxy-tacrine to dihydroxy-tacrines Donepezil clearance is independent of dose, as
appears to be nonstereospecific.[17] The 1-hydroxy- observed in short- or long-term administration in
tacrine metabolite (which was under development volunteers.[27,28] Information concerning the dis-
as velnacrine at the time) can produce clinical ef- tribution of donepezil, particularly in the CNS as
fects through additional inhibition of cholinester- measured by CSF concentrations, is not available.
ase.[24] Donepezil is highly protein bound (96%; 75% to

Table I. Summary of the pharmacokinetic parameters of the cholinesterase inhibitors[11]


Drug Bioavailabilty (%) tmax (h) Protein binding Elimination Hepatic metabolism Linear/nonlinear
(%) half-life (h)
Tacrine 17-37 0.5-3.0 75 1.3-7.0 CYP1A2, CYP 2D6 Nonlinear
Donepezil 100 3-5 96 60-90 CYP2D6, CYP3A4 Linear
Rivastigmine 40 0.8-1.7 40 2 Nonhepatic Linear
Galantamine 85-100 0.5-1.5 18 5-7 CYP2D6, CYP3A4 Linear
CYP = cytochrome P450; tmax = time to maximum serum concentration.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
724 Jann et al.

albumin and 21% to α1-acid glycoprotein).[26] 1.3 Rivastigmine


Steady-state pharmacokinetics of donepezil are
reached within 14 to 22 days after repeated admin- Rivastigmine is considered a ‘pseudoirreversi-
istration of 5 or 10mg for 21 or 28 days.[27,28] ble’ cholinesterase inhibitor that forms a carba-
moylated complex with the enzymes, and has been
1.2.2 Metabolism and Elimination
reported to inhibit both AChE and BChE with
Donepezil is primarily metabolised by CYP2D6
equal potency.[18,35] After single-dose administra-
and 3A4 and undergoes extensive first-pass meta-
tion, enzyme inhibition was reported to persist for
bolism.[25,26,30] The major metabolites of donepezil
10 to 12 hours.[36] This longer duration of action is
consist of a hydrolysis product (6-O-desmethyl-
unique among cholinesterase inhibitors.
donepezil) and an oxidation product (donepezil-
cis-N-oxide).[31] However, donepezil has only 1.3.1 Absorption, Bioavailability and Distribution
one active metabolite (6-O-desmethyl-donepezil), Rivastigmine exhibits rapid and nearly com-
which has equal pharmacological activity to the plete oral absorption, with tmax of 0.8 (0.5 in some
parent drug. 6-O-Desmethyl-donepezil is present reports[18]) to 1.7 hours (table I). Food slows ab-
at about 20% of the plasma concentration of sorption and results in a decrease of Cmax of about
donepezil.[30] The t1⁄2β of donepezil was reported to 30% with an increase in the extent of absorption,
be between 70 and 80 hours.[25] also by 30%.[37] Rivastigmine should be taken with
1.2.3 Effects of Age, Hepatic and meals (breakfast and dinner), which decreases gas-
Renal Impairment trointestinal problems. ZNS 114-666 (NAP-226-
Comparison of the pharmacokinetics of done- 90), its decarbamylated metabolite, is also rapidly
pezil 2mg in elderly patients (68 to 82 years) versus detected within 2 hours of administration.[35]
young adults (20 to 27 years) found that mean tmax About 96% of a radiolabelled dose is absorbed, but
(5.2 vs 3.2 hours), t1⁄2β (103.8 vs 59.7 hours) and the presystemic biotransformation is high, indicat-
mean residence time (MRT; 144 vs 85 hours) were ing a significant first-pass effect with an AUC ratio
significantly (p < 0.05) greater in the elderly pa- of oral to intravenous drug of 0.355 (35% bioavail-
tients.[32] However, AUC and clearance did not dif- ability).[35,37] Protein binding of rivastigmine is
fer significantly between populations. The slower quite low at 40%, with about 40 to 50% of rivas-
absorption rate in the elderly accounts for the tigmine bound with red blood cells. The volume of
slightly longer tmax. The greater t1⁄2β and MRT were distribution of rivastigmine and its metabolite are
due to the significantly greater mean volume of 1.8 to 2.7 and 4.3 to 5.9 L/kg, respectively.[38]
distribution found in the elderly (1217.2 vs Rivastigmine displays a linear relationship of
852.5L). These changes were not considered clin- dose to AUC.[39]. When doses of 2, 8 and 12mg
ically significant, and dosage adjustments were not were administered, the corresponding mean AUCs
suggested. were 6.2 ± 3.2, 41.7 ± 13.0 and 55.9 ± 16.5 μg •
Donepezil disposition was reported to be unaf- h/L, respectively. Under multiple dose conditions
fected by hepatic or renal impairment. With the maintained for at least 3 days, the tmax of rivastig-
exception of Cmax in patients with impaired liver mine in the CSF was 1.4 to 3.8 hours, which was
function (37.5% higher, p < 0.02), pharmaco- slightly longer than the plasma tmax due to com-
kinetic parameters did not differ significantly be- partmentalisation from drug in the plasma entering
tween healthy volunteers and patients with chronic into the CSF.[39] Peak CSF concentrations were
compensated liver cirrhosis and patients with mod- about 40% of those detected in plasma, with rapid
erate to severe renal impairment (creatinine clear- clearance characterised by a t1⁄2β that ranged from
ance <30 ml/min).[33,34] Although these were single- 0.3 to 3.0 hours. A significant correlation was
dose studies, the situation for steady-state conditions found between dose and AUC for rivastigmine and
should not differ. its NAP-226-90 metabolite (r = 0.84, p < 0.0001; r

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 725

= 0.92, p < 0.0001, respectively). Similar results 1.4 Galantamine


were found comparing rivastigmine plasma and
CSF AUCs (r = 0.93, p < 0.0001). In another phar- Galantamine is a phenanthrene alkaloid similar
macokinetic study with a 3mg single dose, CSF to codeine, and was isolated from the snowdrop
rivastigmine concentrations were below the lower plant Galanthus nivalis.[18] In addition to its ac-
limit of quantification (<0.65 μg/L); however, tions upon AChE, in vitro evidence suggests that
NAP-226-90 was detectable for over 15 hours.[38] it also modulates nicotinic receptors, thereby en-
Mean tmax for NAP-226-90 was 2.93 hours and hancing acetylcholine activity at the synapse.[41]
mean Cmax was 3.14 ± 0.57 μg/L. These two actions form a different type of ‘dual’
pharmacological mechanism to enhance acetyl-
1.3.2 Metabolism and Elimination choline function.
Rivastigmine is not significantly metabolised 1.4.1 Absorption, Bioavailability and Distribution
by hepatic oxidative CYP isoenzymes.[35,37] Rivas- Galantamine has been administered by a variety
tigmine is rapidly and extensively metabolised, of routes, including intravenous, oral, rectal and
primarily by cholinesterases, to the NAP-226-90 subcutaneous.[41-44] Only the oral formulation will
metabolite.[37] NAP-226-90 may then undergo N- be described in detail regarding its pharmaco-
demethylation and/or sulphate conjugation. After kinetic parameters and compared with other for-
metabolism, it undergoes rapid renal elimination. mulations.
Accumulation of rivastigmine and NAP-226-90 Galantamine is readily absorbed after oral ad-
does not occur after repeated administration of 1 ministration (tablet and solution) with an 85 to
to 6 mg/day.[37] The t1⁄2β of rivastigmine from the 100% bioavailability.[43,44] After oral administra-
CSF was reported to range from 0.31 to 2.95 hours tion, tmax ranged from 0.5 to 2.0 hours.[18,41,44]
with multiple dose administration.[39] Food did not affect AUC, but Cmax was decreased
by 25% and tmax was delayed by 1.5 hours. The
1.3.3 Effects of Age, Hepatic and mean volume of distribution in the central com-
Renal Impairment
partment was 0.93 L/kg, indicating an initial rapid
The t1⁄2β of rivastigmine was slightly prolonged distribution, and the mean volume of distribution
in the elderly compared with adult volunteers at steady state was 2.64 L/kg, reflecting tissue ac-
(ranges 0.88 to 1.25 vs 0.80 to 0.99 hours), which cumulation.[44] Plasma protein binding was rela-
does not appear to be clinically significant.[39] In tively low at 18%,[41] but in patients with AD who
patients with renal and moderate hepatic impair- received 30 or 40mg, plasma protein binding at 2
ment, the AUC of rivastigmine was 1.4-fold and to 3 hours post-dose was reported to be 28 to 34%,
2.3-fold higher, respectively, than in adult healthy without correlation with amounts of total protein
controls.[40] The AUC of NAP-226-90 was also or albumin.[41] Although galantamine concentra-
0.8-fold lower in renally impaired patients, but tions were not measured, mean CSF AChE inhibi-
1.5-fold greater in hepatically impaired patients. tion (reported as units/ml) in patients was found to
Significant differences in the t1⁄2β of parent drug increase from 76.5 ± 10.4 to 221.5 ± 17.7 units/ml
were not found in patients with moderate or severe (p = 0.028), indicating significant pharmaco-
renal impairment. The t1⁄2β of NAP-226-90 was dynamic activity from the drug.[45]
longer only in patients with severe renal impair-
ment (6.0 vs 3.4 hours). Although the AUC of 1.4.2 Metabolism and Elimination
rivastigmine was higher in patients with hepatic The major (75%) route of metabolism of
impairment, specific dosage recommendations galantamine occurs via hepatic CYP isoenzymes.
may not be needed in these special populations. In vitro studies indicate that CYP2D6 and 3A4 are
Rivastigmine has not been studied in patients with involved with galantamine metabolism.[41] San-
severe hepatic impairment. guinine (O-demethyl-galantamine) is the major

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
726 Jann et al.

metabolite of galantamine. It is formed by CYP- (Child Pugh score >9) and/or renal (creatinine
2D6, can account for up to 20% of orally adminis- clearance <9 ml/min) impairment.
tered galantamine, and is reported to be three times
more potent than galantamine as an AChE inhibi- 2. Drug Interactions
tor.[46] Other less significant metabolites include
galantaminone and N-demethyl-galantamine.
About 97% of a single radiolabelled 4mg dose of 2.1 Tacrine
galantamine to healthy volunteers was recovered
in the urine and the remaining portion in the fae- 2.1.1 Influence of Cytochrome P450 1A2
ces.[41] Significant differences in the urinary and CYP1A2 activity was investigated in patients
faecal excretion amounts between poor and exten- with AD (n = 19) who received a 40mg single dose
sive metabolisers of CYP2D6 were not found.[41] of tacrine together with a caffeine probe (2 mg/kg)
The t1⁄2β of galantamine ranges between 5 and 7 followed by breath and urine tests.[47] Tacrine oral
hours (table I). Mean total clearance was reported clearance showed a 15-fold interpatient variability.
to be 0.34 L/h/kg with a 25% lower mean renal Oral clearance of tacrine significantly correlated
clearance of 0.084 L/h/kg, which represents about with the 2-hour production of 13CO2 (r = 0.56, p =
25% of the total drug clearance.[44] Population 0.01), with the p-xanthine/caffeine metabolic uri-
pharmacokinetic analysis indicated a 20% lower nary ratio (r = 0.76, p = 0.0002) and the caffeine
clearance in females, which was not considered to metabolic urinary ratio (r = 0.76, p = 0.0001).
be of clinical relevance. Clearance was reduced by These observations support a central role for
25% in poor metabolisers of CYP2D6, and again CYP1A2 in the in vivo disposition of tacrine and
this finding may not be clinically significant be- demonstrate a potential for drug-drug interactions,
cause of the wide interpatient variability and in- but did not support the routine use of caffeine to
volvement of CYP3A4.[41,46] individualise tacrine therapy.
Significantly higher plasma concentrations of
1.4.3 Effects of Age and Hepatic or tacrine have been reported in women compared
Renal Impairment with men.[48] Although mean values were not pro-
Patients with AD have Cmax values for galan- vided, the tacrine plasma concentrations ranged
tamine 30 to 40% higher than those found in from 11 to 30 μg/L in women compared with 8 to
healthy adult volunteers.[41] However, differences 15 μg/L in men. It was proposed that this difference
were not observed in AUC and t1⁄2β.[41] Galantamine may be due to lower CYP1A2 isoenzyme activity
disposition did not significantly differ between in women.[18,48] Smoking is a well-known inducer
healthy volunteers and patients with mild hepatic of CYP1A2, and smokers have a significantly
impairment (Child Pugh score 5 to 6), but in pa- lower tacrine t1⁄2β than nonsmokers (2.1 vs 3.2
tients with moderate hepatic impairment (Child hours), with consistently lower concentrations of
Pugh score 7 to 9), AUC and t1⁄2β were increased by tacrine and metabolites.[49]
about 30%.[41] In patients with creatinine clearan-
ces ≥9 ml/min, peak and trough galantamine con- 2.1.2 Cimetidine
centrations were similar to those found in patients Coadministration of cimetidine with tacrine in
with AD.[41] an elderly population (n = 12) resulted in a 39%
A maximum dosage of 16 mg/day is recom- increase in tacrine AUC (mean 56 vs 75 μg • h/L)
mended for patients with moderate hepatic impair- with a corresponding decrease in clearance by 30%
ment. No dosage adjustments are suggested for pa- (mean 23.4 vs 16.3 L/min).[50] As a result of in-
tients with mild hepatic or renal impairment creased parent drug, the AUCs of the 1-, 2- and
(creatinine clearance ≥9 ml/min). Galantamine is 4-hydroxy metabolites of tacrine also showed in-
contraindicated in patients with severe hepatic creases that ranged from 17 to 60%.[51]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 727

2.1.3 Hormone Replacement Therapy 2.2 Donepezil


Ten healthy female volunteers received hor-
mone replacement therapy (HRT) [estradiol valer-
ate 2mg plus levonorgestrel 0.25mg] or placebo for 2.2.1 Cimetidine
10 days in a randomised crossover study. At 1 hour The coadministration of cimetidine 800 mg/day
after the last dose of HRT or placebo, a single with donepezil 5 mg/day for 7 days in 19 healthy
male volunteers aged 18 to 55 years resulted in
40mg dose of tacrine was given and its pharmaco-
significantly higher donepezil mean Cmax (26.6 ±
kinetic profile evaluated.[52] Coadministration of
1.5 vs 29.9 ± 1.3 μg/L, p = 0.0002) and AUC (472.3
HRT with tacrine resulted in an increase of tacrine ± 26.9 vs 526.9 ± 24.3 μg • h/L, p = 0.0001).[55]
AUC by 60% (p = 0.009) with a reduction of mean However, coadministration did not significantly ef-
clearance by 31% (p = 0.14). Significant changes fect tmax, t1⁄2β and drug accumulation profiles. Al-
in t1⁄2β were not found. The metabolic ratio of 1- though these changes are considered clinically not
hydroxy-tacrine to tacrine was significantly re- significant, clinicians should carefully monitor pa-
duced in all subjects (mean 26%, p < 0.001). This tients when cimetidine is used.
interaction between HRT and tacrine, resulting in
reduced metabolism of the parent drug, indicates 2.2.2 Digoxin
inhibition of CYP1A2 during the first-pass phase. The pharmacokinetic parameters of digoxin
0.25 mg/day for 6 days were not affected by co-
2.1.4 Fluvoxamine adminstration of donepezil (5 mg/day for 6 to 10
In vitro and in vivo studies have been conducted days or 10 mg/day for 12 days) in 12 healthy male
to investigate the interaction between tacrine and volunteers.[50] Changes in ECG or cardiac conduc-
fluvoxamine.[53,54] Fluvoxamine is a potent tion did not occur. The pharmacokinetic parame-
CYP1A2 inhibitor. Healthy volunteers were ran- ters of donepezil (tmax, Cmax, AUC and t1⁄2β) were
domised to receive fluvoxamine 100 mg/day or not altered by digoxin.
placebo for 6 days and then crossed over to the
other treatment arm. On the last day, tacrine was 2.2.3 Ketoconazole
coadministered with fluvoxamine or placebo.[54] Ketoconazole is a potent CYP3A4 inhibitor,
Mean oral tacrine clearance significantly de- and the possibly of a significant interaction with
creased from 1683 ± 802 to 200 ± 106 L/h (p < donepezil was evaluated. Coadministration of
donepezil 5 mg/day and ketoconazole 200 mg/day
0.05). The AUCs of all three tacrine hydroxy me-
for 7 days significantly altered the pharmacokinet-
tabolites significantly increased with fluvoxamine
ics of donepezil.[56] Significant increases in Cmax
coadministration, and five of the thirteen volun-
(27.6 ± 1.7 vs 37.7 ± 2.2 μg/L, p < 0.0001), AUC
teers experienced gastrointestinal adverse events. (501.1 ± 38.3 vs 680.9 ± 48.4 μg • h/L, p < 0.0001)
In vitro modelling using human liver micro- and drug accumulation ratio (4.2 ± 0.2 vs 5.0 ± 0.2,
somes and yeast-expressed human CYP1A2 re- p < 0.0001) were observed, but tmax and t1⁄2β were
ported changes in kinetic parameters during unaltered. The clinical significance of this interac-
coincubation of tacrine and fluvoxamine that cor- tion remains to be determined, but clinicians
related with the in vivo study.[53] should always carefully monitor patients. Keto-
conazole pharmacokinetics were unaffected by
2.1.5 Theophylline donepezil. Extrapolation of donepezil data was
Concomitant administration of tacrine and conducted to estimate the effects of ketoconazole
theophylline resulted in a reduction in theophyl- on steady-state concentrations of donepezil, and a
line clearance by about 50% (from a mean of 41.8 23 to 30% increase was predicted by using the
to 21.6 L/min).[12] maximum effect (Emax) model.[56]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
728 Jann et al.

2.2.4 Psychotropic Drugs was not significantly affected.[62] Similarly, the phar-
Two case reports have documented a possible macokinetic parameters of (R)- and (S)-warfarin
interaction between paroxetine (a potent CYP2D6 (given as a single 25mg dose of racemic warfarin)
inhibitor) 20 mg/day and donepezil 5 mg/day.[57] were not significantly affected when given with
Both patients experienced gastrointestinal adverse donepezil (5 mg/day for 6 to 10 days or 10 mg/-
effects (severe diarrhoea and flatulence), insom-
day for 12 days) in healthy volunteers.[63] Subse-
nia, agitation, confusion and aggression. Done-
quently, no significant changes in prothrombin
pezil plasma concentrations were not measured
(and are not routinely conducted in the clinical set- times occurred. Therefore, it was concluded that
ting). Drug-drug interactions with serotonin reup- donepezil can be safely given with theophylline or
take inhibitors could be very complex, and could warfarin without dosage adjustments to either
involve other CYP isoenzymes and drug transport agent.
systems.[58]
Another case report described fulminant hepa- 2.3 Rivastigmine
titis when sertraline and donepezil were com-
bined.[59] Donepezil plasma concentrations were Drug-drug interactions of clinical importance
not obtained, but a possible mechanism suggested are not expected with rivastigmine because of its
was inhibition of CYP3A4 and 2D6. Although short pharmacokinetic t1⁄2β, low protein binding and
these cases do not prove an actual pharmacokinetic minimal metabolism by CYP.[35,37] In vitro and phar-
drug interaction with donepezil, the possibility of macokinetic studies conducted in humans who
this type of interaction cannot be fully dismissed received digoxin, warfarin, diazepam or fluoxetine
and careful patient monitoring is recommended.
reported no significant interactions.[37] However
Drug interaction studies with risperidone and
pharmacodynamic interactions may occur with
thioridazine have been reported. Both agents in-
medications that affect the cholinergic system.[37]
hibit CYP2D6. In healthy young subjects (n = 12),
A retrospective analysis of the four major clinical
a 50mg single dose of thioridazine was given prior
to and after 14 days of administration of donepezil trials (2459 patients) found that no increase in ad-
5mg.[60] Donepezil produced no significant changes verse events occurred among those patients who
in thioridazine pharmacokinetics (AUC and tmax). received 22 different classes of medication com-
All ECGs remained normal during coadministra- monly used in elderly populations, including anti-
tion, indicating a lack of significant pharmaco- anginal, antihypertensive, antiemetic, antihista-
dynamic interaction between thioridazine and mine, anxiolytic, anti-inflammatory or estrogenic
donepezil. In the second study, risperidone 1 to 4 agents.[64]
mg/day was maintained in schizophrenic patients
(n = 16) and donepezil 5mg was coadministered for
2.4 Galantamine
7 days.[61] Risperidone pharmacokinetic parame-
ters were evaluated prior to and during donepezil Since this agent is metabolised by CYP3A4 and
coadministration, and no significant changes in
2D6, potent inhibitors of these isoenzymes could
risperidone disposition were reported.
result in significant drug-drug interactions. The
2.2.5 Theophylline and Warfarin
bioavailability of galantamine was reported to be
When donepezil (5 mg/day for 6 to 10 days or increased by 40% when coadministered with
10 mg/day for 12 days) was coadministered with paroxetine, and by 30 and 12% when given with
theophylline (100mg twice daily titrated up to op- ketoconazole and erythromycin, respectively.[41]
timal plasma concentrations) in healthy volun- Galantamine had no significant effect upon the
teers, the pharmacokinetic profile of theophylline pharmacokinetics of warfarin or digoxin.[41]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 729

3. Pharmacological Aspects agent is metrifonate, which is no longer in clinical


development.[35,65]
The main pharmacological action of cholines-
terase inhibitors is to inhibit the enzymes AChE 3.2 Acetylcholinesterase versus
and BChE responsible for the breakdown of ace- Butyrylcholinesterase Inhibition
tylcholine at the synapse. This action produces
acetylcholine accumulation or prolongs its effects Both AChE and BChE are present in the human
at the synapse, which can be equivalent to exces- brain.[18,67] The three-dimensional structures of
sive stimulation of cholinergic receptors (both these enzymes have been published, and only a
muscarinic and nicotinic) throughout the central simple schematic diagram is presented in figure 2.
and peripheral nervous system.[9] This section de- AChE plays an important role in the development
scribes how cholinesterase inhibitors accomplish of neurons and neurite outgrowths.[10] It is found
this function. in erythrocytes, platelets and lymphocytes. The ac-
tivity of AChE has been reported to be decreased
in the brains of patients with AD.
3.1 Reversible versus Irreversible Inhibition The structure of BChE is similar to that of
AChE, but it has a slight difference in its amino
The structure of AChE and BChE is well acid sequence. BChE is synthesised in the liver and
known. The active centre consists of two sites, an secreted into the plasma. It is found in the gastro-
anionic site and an esteratic site. Acetylcholine and intestinal tract and heart with AChE, and in the
related compounds bind to the anionic site by cou- neurons and glial cells of the brain. BChE activity
lombic and hydrophobic forces, whereas binding was reported to be increased in the brains of pa-
occurs at the esteratic site via nucleophilic binding tients with AD, in association with increased β-
on the acyl carbon (figure 2).[65] The terms ‘revers- amyloid density.[68]
ible’ and ‘irreversible’ have a long history.[9] Cur- Cholinesterase inhibitors may exert additional
rently, cholinesterase inhibitors are classified pharmacological actions beyond inhibition of cho-
pharmacologically into three groups on the ba-
sis of their duration of inhibition – short-acting,
intermediate-acting and long-acting.
Reversible or short-acting agents such as tacr-
Cholinesterase enzyme
ine and donepezil bind reversibly to cholinesterase
and their durations of action are very short (min- E A
+
utes). Rivastigmine is classified as an intermediate- O N
acting or ‘pseudo-irreversible’ agent, as its alcohol O Gorge area
Ach
moiety is cleaved to form a carbamoyl enzyme and A/B
the inhibition lasts for 10 to 12 hours after a single Hypothetical ChEI
dose.[23,35,65] However, since the body does not A
need to synthesise new enzyme protein, rivastig- B
mine can be considered a ‘reversible’ carbamate E Peripheral area
inhibitor[9] and indeed is so described in the pack-
age insert.[66]
Fig. 2. Model of cholinesterase activity on acetylcholine (ACh).
Long-acting agents or irreversible inhibitors Two types of binding sites have been identified, known as es-
bind to cholinesterase and prevent any further en- teratic (E) and anionic (A) sites, located in the gorge and pe-
zyme activity. The body must synthesise new en- ripheral areas of the enzyme. The hypothetical cholinesterase
inhibitor (ChEI) A/B must bind to at least one site in the gorge.
zymes for replacement and continued pharmaco- The drug can also bind to either of the peripheral sites (section
logical function. An example of an irreversible B of the hypothetical drug).

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
730 Jann et al.

linesterases. In vitro models have shown that cho- concentrations (IC50) of 0.12 and 24.0 μmol/L, re-
linesterase inhibitors can increase the secretion of spectively.[46] Galantamine binds at the esteratic
the soluble amyloid precursor protein (APP).[69] site of the gorge, and its tertiary amine group ap-
The long-term efficacy of cholinesterase inhibitors pears to be associated with Asp-72, binding it to
may be related to their dual inhibitory action on the peripheral esteratic site.[10]
acetylcholine and on AChE and/or BChE.[35,69] Cholinesterase inhibitors can also bind to an al-
The different molecular structures of the cholin- losteric site on the nicotinic acetylcholine receptor
esterase inhibitors (figure 1) account for their se- and may further enhance cholinergic transmission.
lective binding to AChE and/or BChE. The three- This independent function is separate from cho-
dimensional structure of AChE and BChE forms a linesterase inhibition. Galantamine and physos-
‘gorge’ around the esteratic and anionic sites (fig- tigmine were shown to possess this additional
ure 2).[67] Acetylcholine binds at both sites in the nicotinic binding action in vivo.[41] However,
gorge. Each drug binds to the enzyme in a different galantamine demonstrated the most potent ability
three-dimensional manner, either alone or in com- to modulate the nicotinic receptor.[71] The modula-
bination at the esteratic and anionic sites. Esteratic tion results in increased ion channel opening in-
and anionic sites are located in the gorge and pe- duced by acetylcholine and nicotinic agonists and
ripheral areas. The inhibitor (hypothetical drug slows down receptor desensitisation. Cell culture
A/B) must bind to one site in the gorge area to models have shown potentiation by galantamine of
produce inhibition. However, cholinesterase inhibi- the action of acetylcholine in HEK-293 cells trans-
tors can additionally bind to either site in the peri- fected with human α4β2 nicotinic acetylcholine
pheral areas, as shown by section B of a hypothet- receptors.[71] The 50% effective concentration
(EC50) for acetylcholine was improved from 2.3
ical drug.
μmol/L without galantamine to 0.9 μmol/L with
Tacrine forms ionic bonds with the amino acids
galantamine. The Hill coefficient (nH) also im-
on the anionic site and displays slight specificity
proved from 1.2 without galantamine to 1.7 with
for BChE inhibition versus AChE inhibition.[10]
galantamine.
Rivastigmine interacts with the esteratic and an-
ionic sites at the foot of the gorge.[10] Based upon
3.3 Dose versus Cholinesterase Inhibition
measurements with CSF in healthy volunteers
given rivastigmine 1 to 6mg twice a day, rivastig- A comparison of the relationship between the
mine shows an equal potency to inhibit both AChE doses of the different cholinesterase inhibitors and
and BChE.[39] Donepezil binds to the anionic site their effects on cholinesterase inhibition is pre-
at the bottom of the gorge and at the peripheral sented in table II. With the exception of eptastig-
anionic site.[10] Donepezil and galantamine display mine and galantamine, these doses reflect those
higher potency for AChE than for BChE.[25,41] used in clinical practice.[72] Galantamine has been
Based upon in vitro data, donepezil has much evaluated up to 32 mg/day, but recommended dos-
greater potency and specificity for AChE inhibi- ages are from 16 to 24 mg/day.[41]
tion than for BChE inhibition, with an AChE/- Data regarding tacrine dose and enzyme inhibi-
BChE ratio of 1265 : 1.[70] The selectivity of done- tion have not been published. Data with more than
pezil for AChE is based upon its binding with two doses studied are only available for rivastig-
amino acids Trp-279 and Phe-330, which are ab- mine and eptastigmine, but demonstrate the fol-
sent from BChE.[10] Galantamine displays inhibi- lowing biochemical features common to cholines-
tion for both AChE and BChE, but its AChE/BChE terase inhibitors. Rivastigmine displays equal
ratio is much lower at about 53 : 1.[70] The O- inhibition of AChE and BChE, although there was
demethyl metabolite of galantamine also displays some disparity reported at the 6mg dose which is
AChE and BChE inhibition, with 50% inhibitory most probably due to assay variability or interpati-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 731

Table II. Relationship between dose and cholinesterase inhibition for cholinesterase inhibitors
Reference Drug Dose (mg) Enzyme inhibition
Tacrine 40-160 40% (BChE)
73 Donepezil 5 63.7% (AChE)
10 77.3% (AChE)
39 Rivastigmine 2 20% (AChE); 24% (BChE)
4 36% (AChE); 28% (BChE)
6 46% (AChE); 76% (BChE)
10 55% (AChE); 51% (BChE)
12 62% (AChE); 62% (BChE)
44 Galantamine 5a 34% (AChE)
10a 53% (AChE)
10 42% (AChE)
72 Eptastigmine 8 20% (AChE)
20 29% (AChE)
32 45% (AChE)
40 54% (AChE)
a Intravenous administration.
AChE = acetylcholinesterase; BChE = butyrylcholinesterase.

ent variation. Enzyme inhibition data was evalu- cations is needed. Tacrine was the first cholines-
ated for only the 5 and 10mg doses of donepezil. terase inhibitor to be approved for AD, and many
A general picture for these agents emerges of inhi- issues were discovered and eventually resolved
bition reaching a plateau, with different drugs during and after its clinical trials.[74]
reaching nonlinearity at different doses. The cho- Assessment of AD consists of three domains:
linesterase inhibitors exhibit linear disposition, cognitive impairment, behavioural symptoms, and
and increasing the dosage can lead to a correspond- functional changes in daily activities. The inter-
ing increase in AUC, but the increase in enzyme relationship between these three domains forms
inhibition may or may not be reflective of the dos- the basis of evaluating antidementia compounds,
age change. and cholinesterase inhibitors have led the way in
this field. This section will briefly describe the
4. Pharmacodynamic Aspects most widely accepted standardised clinical assess-
ment tools to evaluate cholinesterase inhibitors in
Cholinesterase inhibitors can be evaluated by a
AD. The usual duration for a clinical trial in AD is
variety of methods in the areas of clinical efficacy
6 months, and changes are compared (the delta
and adverse effects. These two areas can be further
score) from baseline to the study endpoint.
divided into dose-related and dose-unrelated ac-
The assessment tool that has become recog-
tions.
nised by many clinicians to reflect changes in cog-
nition is the Alzheimer’s Disease Assessment
4.1 Clinical Efficacy
Scale – cognitive section (ADAS-cog).[75] ADAS-
4.1.1 Clinical Rating Scales cog scores range from 0 to 70 points, with high
The nature of AD presents several unique scores indicating impairment; however, its short-
challenges in the design and analysis of clinical comings are that changes in the delta score from
trials. The majority of the assessment tools used baseline to study endpoint may not reflect mean-
have limited value in the clinical setting and are ingful changes in the patient’s status. Yet, the
unfamiliar to many clinicians. However, a basic ADAS-cog has become the standardised tool to as-
understanding of the many scores and their impli- sess cognition in AD trials. Regulatory agencies

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
732 Jann et al.

have also specified that cognition cannot be the samples.[77,81] Cholinesterase inhibitors typically
only indicator of response, and that a global assess- display a dose-dependent activity where the thera-
ment by the clinician is equally if not more import- peutic index measured by changes in cognitive
ant in determining drug response. The clinical as- scores closely approaches the maximum tolerated
sessment tool utilised to evaluate global changes dose. Many factors complicate these findings, in-
is the Clinical Global Impression of Change cluding wide interpatient variability in metabo-
(CGIC).[76] The patient and their caregiver is inter- lism, tolerance, disease stage and efficacy. Fur-
viewed by the clinician. Based upon the clinician’s thermore, in dose-ranging studies, cholinesterase
total experience with this population, the rater inhibitors appear to be better tolerated in AD pa-
makes an assessment of the patient’s cognition, tients compared with healthy volunteers.[81]
behaviour and functional ability scored as one A conceptual model for cholinesterase inhibi-
evaluation score. Both ADAS-cog and CGIC tors, relating enzyme inhibition to clinical efficacy
scores have been used to develop pharmaco- and adverse effects, is presented in figure 3. As
dynamic models of clinical response to cholines- some cholinesterase inhibitors bind to both en-
terase inhibitors.[77] Perhaps the best known instru-
zymes, plasma and erythrocyte cholinesterase
ment used to assess cognition is the Folstein
have been used as ‘surrogate’ biomarkers for clini-
Mini-Mental Status Exam (MMSE).[78] Although
cal efficacy. Direct comparisons between cholin-
well known, its use has been restricted in clinical
esterase inhibitors cannot be conducted, as their
trials for inclusion/exclusion of patients and sec-
dose and plasma concentrations differ when com-
ondary outcome measures.
paring these parameters to cholinesterase inhibi-
Although cognition declines progressively with
tion. The approximate range for cholinesterase in-
time, behavioural symptoms can increase or de-
crease in frequency and/or severity. The Neuropsy- hibition to achieve clinical efficacy varies between
chiatric Inventory (NPI) scale is commonly used to 40 and 70% (60% for donepezil, tacrine and
evaluate behavioural symptoms in AD patients by rivastigmine, 74% for galantamine and 35% for
the clinician, accounting for both frequency and eptastigmine).[77,81] Tables II and III also illustrate
severity.[79] Scores are tabulated and range from 10 this concept of enzyme inhibition in relationship to
to 120 points, but again the delta score is used to dose and clinical efficacy. The maximal level of
evaluate the efficacy of cholinesterase inhibitors. inhibition without adverse effects differs among
A variety of assessment scales are available to
evaluate functional daily activities of the patient.
A widely used validated scale is the Progressive Clinical efficacy
Adverse effects
Deterioration Scale (PDS), in which the patient is
evaluated by their caregiver.[80] PDS scores range
Adverse effects

from 0 to 100 points, and average scores for mild,


Efficacy

moderately and severely ill patients are 48, 34 and


15 points, respectively. Like the other rating
scales, the delta PDS score is used to determine
changes in a patient’s function over time.

4.1.2 Correlation of Clinical Efficacy, Dose and


Enzyme Inhibition
A common method to evaluate the pharmaco- 0 10 40 70 100
dynamic actions of cholinesterase inhibitors in- Cholinesterase inhibition (%)
volves the measurement of plasma and/or erythro- Fig. 3. Conceptual model relating cholinesterase inhibition to
cyte cholinesterase inhibition in venous blood clinical efficacy and adverse events.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 733

the cholinesterase inhibitors. The higher the level model proposed for cholinesterase inhibitors in
of inhibition that can be achieved without or with figure 3.
only minimal adverse effects, the greater proba- A linear relationship between ADAS-cog, PDS,
bility of meaningful therapeutic benefit for the pa- CGIC and MMSE was suggested for rivastigmine
tient.[82] for the pooled data from four large multicentre
These factors are expressed in the conceptual clinical trials.[86] The maximum daily rivastigmine
model for cholinesterase inhibitors in figure 3. The dosage reported in the linear-regression model was
range of 40 to 70% probably represents the optimal 12mg, which corresponded to the maximum dos-
therapeutic range for enzyme inhibition. Adverse age used in the study. It remains unknown whether
effects are also dose-dependent, as their incidence or not higher dosages could further improve cog-
and severity increase with increasing dosage. At nition or functional activities in patients with AD.
lower dosages, the likelihood of adverse effects is Galantamine dosages of 16 and 24 mg/day were
reported to be the optimal dosages for therapeutic
minimal but can rapidly increase as enzyme inhi-
response. When higher dosages of 32 mg/day were
bition gradually increases. The point where the
used, the mean ADAS-cog score (versus placebo)
lines of clinical efficacy and adverse effects cross
was 3.8 points, whereas the 24 mg/day dosage had
becomes the maximal tolerated dose (MTD). As
a mean ADAS-cog score of 3.9 points. The inci-
enzyme inhibition increases (a function of dose), dence of adverse effects was much higher in the 32
clinical efficacy is maximised but adverse effects mg/day group versus the 24 mg/day dose group,
predominate. This relationship cannot be de- which provides a ‘ceiling’ dosage for maximum
scribed as an inverted U-shaped curve, since clini- benefit.[87] The improvement did not significantly
cal efficacy can be maximised with increasing en- decrease, which further supports the proposed con-
zyme inhibition. Theoretically, one can have 100% ceptual model.
enzyme inhibition with maximal benefit from the
drug; however, its adverse effects would become 4.1.3 Effects on Behavioural Symptoms
intolerable and the risk of continued drug exposure A small nonblinded study with tacrine assessed
would outweigh the benefits of continued therapy. behavioural symptoms in 28 patients with AD.[88]
The relationship between dose of cholinester- Tacrine dosages started at 40 mg/day and were in-
ase inhibitors and clinical response (delta ADAS- creased to 160 mg/day at 6-week intervals; how-
cog scores) is presented in table III.[73,83-85] All ever, only 10 patients were able to tolerate the
maximum daily dosage. NPI evaluations were
cholinesterase inhibitors appear to display a mini-
completed at 6-week intervals, and mean scores
mum dose ‘threshold’ where a statistically signifi-
were consistently lower (better) as the tacrine dos-
cant difference from placebo can be found. For
ages were increased. The mean changes in NPI
example, although a slight decrease (or improve-
scores were –4.53, –6.92 and –8.0 points with tac-
ment) in the mean ADAS-cog score was observed rine dosages of 80, 120 and 160 mg/day, respec-
for tacrine 80mg, the difference was not signifi- tively. Statistical significance was achieved with
cant. Similar findings occurred with rivastigmine the 120 (p = 0.04) and 160 (p = 0.01) mg/day dos-
1 to 4mg and galantamine 8mg. Correspondingly, ages.
changes in functional daily activities paralleled the Another nonblinded study with donepezil in 25
improvement in cognitive scores shown in table patients with AD showed similar findings, where
III. The daily dosages of cholinesterase inhibitors both 5 and 10 mg/day produced improvement in
needed to achieve a consistent cholinesterase inhi- emotional and behavioural symptoms.[89] A dose-
bition of 40 to 70% correspond to those causing dependent effect could not be found with done-
improvements in ADAS-cog and functional activ- pezil, as all patients started with 5mg and the dose
ity scores. These findings fit into the conceptual increased to 10mg after 4 months.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
734 Jann et al.

Table III. Summary of the pharmacodynamic properties of cholinesterase inhibitors


Agent Enzyme inhibition Clinical effects
selectivity dose (mg) inhibition (%) dosage (mg/day) Δ ADAS-cog Δ daily activity score
Tacrine BChE > AChE 160 40 (BChE) P NR NR
80 –1.4 3.1
120 –2.0* 4.5*
160 –2.1* 4.6*
Donepezil AChE >>> BChE 10 77 (AChE) P 1.82 NR
5 –0.67* NR
10 –1.06* NR
Rivastigmine AChE = BChE 12 62 (BChE) P 4.15 –4.90a
62 (AChE) 1-4 2.27 –5.19
6-12 –0.79* –1.52*
Galantamine AChE >> BChE NR NR P 1.8 –4.0a
8 0.1 –3.1
16 –1.5* –0.5*
24 –1.8* –1.6*
a Decline from baseline.
AChE = acetylcholinesterase; ADAS-cog = Alzheimer’s Disease Assessment Scale – cognitive section; BChE = butyrylcholinesterase; NR
= not reported; P = placebo; Δ = change in score from baseline to study endpoint; * indicates significantly different from placebo, p < 0.05.

In the multicentre galantamine trial, the mean studies have only been completed in very small
NPI scores improved in patients in the 16 mg/day patient numbers and in nonblinded studies. Two
(–0.1 points) and 24 mg/day (–0.1 points) groups small separate studies with a total of 13 patients
compared with placebo recipients (+2.3 points) with AD have used single photon emission com-
and the 8 mg/day group (+2.3 points).[84] puted tomography (SPECT).[90,91] In each study,
Psychotropic benefits of cholinesterase inhibi- patients were treated with physostigmine and tacr-
tors have been proposed.[11] Various clinical stud- ine plus lecithin. Scans were conducted prior to and
ies with different cholinesterase inhibitors, both after several weeks of treatment. One study re-
nonblinded and controlled multicentre trials, have ported increased cortical perfusion in the left fron-
noted improvement in behavioural symptoms. Re- tal and high frontal regions,[90] whereas the other
duced psychosis was reported with rivastigmine. reported no significant changes.[91]
Delusions, agitation, pacing, aberrant motor activ- Recently, functional magnetic resonance imag-
ity, hallucinations, apathy and depression were ing (fMRI) coupled with apolipoprotein E (apoE)
found to improve with tacrine, physostigmine and genotyping was evaluated in 30 neurologically
metrifonate. Improvements in total NPI scores normal subjects.[92] apoE has been established as a
were noted with donepezil, rivastigmine and
potential risk factor for AD, but does not appear to
metrifonate. These findings support the sugges-
be a predictor of response to cholinesterase inhib-
tions of a psychotropic benefit with cholinesterase
itors.[86] Patterns of brain activation in the hippo-
inhibitors; however, a relationship between beha-
campal, parietal and prefrontal areas detected by
vioural symptoms and drug dose or enzyme inhi-
fMRI, and carriage of the apoE-4 genotype, were
bition remains to be established.
found to be associated with memory impairment.
4.1.4 Imaging Studies These combined tests may eventually be used to
The concept and application of using noninvas- examine effects of cholinesterase inhibitors.[92]
ive imaging techniques for diagnostic and drug Positron emission tomography (PET) can be a
therapeutic monitoring in AD appeals to many cli- valuable tool for the early detection of AD, identi-
nicians and scientists. However, to date, imaging fication of patients with mild cognitive impair-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 735

ment, and development of surrogate markers for 80 to 160 mg/day were evaluated, safety data was
diagnosis and pharmacological interventions.[93] A presented for the entire patient population but a
PET method for detecting β-amyloid plaques and dose-dependent pattern could not be determined.
neurofibrillary tangles in the brains of AD patients The overall incidence of nausea and vomiting was
has been described with the development of a 35%, and of diarrhoea 18%.[83] Since these rates of
hydrophobic radiofluorinated derivative of 1,1- adverse effects were higher than in the lower dose
dicyano-[2- (6-dimethylamino)naphthalen- 2-yl]- tacrine study, it is a reasonable assumption that a
propene (FDDNP).[93] When injected intrave- dose-dependent effect can be found with tacrine.
nously into seven AD patients, FDDNP showed The incidences of gastrointestinal adverse ef-
greater accumulation and slower clearance in fects with the other cholinesterase inhibitors are
brain regions with high concentrations of amyloid shown in table IV. Like tacrine, the other cholin-
plaques and neurofibrillary tangles, particularly esterase inhibitors display dose-dependent adverse
the hippocampus, amygdala and entorhinal cortex. effects. At maximum doses, the incidence appears
The FDDNP residence time in these areas had sig- to increase by at least 2-fold. However, methods to
nificant correlations with immediate and delayed improve gastrointestinal adverse effects have been
memory performance. recommended (see section 4.2.2).
Other PET applications include combining ge- Dose-dependent CNS adverse effects may be
netic risk factors (apoE-4), longitudinal studies present with cholinesterase inhibitors. The main
with glucose metabolism in persons at risk for de- CNS adverse effect reported is dizziness, but not
mentia and clinical trials with surrogate markers. all clinical trials with cholinesterase inhibitors se-
Two placebo-controlled trials are underway with lectively reported this particular event. The fre-
celecoxib (a cyclo-oxygenase-2 inhibitor) and done- quency of dizziness with donepezil 5 and 10mg
pezil that combine PET imaging with apoE-4 ge- was reported to be 5 and 9%, respectively.[95] This
netic analysis in patients with mild cognitive im- pattern was not found with the earlier donepezil
pairment to determine if these medications can trial, where dizziness was reported to be 10 and
affect progression to AD. 8% with the 5 and 10mg doses, respectively.[73]
Rivastigmine had a reported incidence of dizziness
4.2 Adverse Effects of 8 and 14% with the 1 to 4mg and 6 to 12mg dose
Adverse effects from cholinesterase inhibitors groups, respectively.[86] Trials with galantamine
are linked to their pharmacological cholinergic ac- reported rates of dizziness of 14 and 19% for the
tions upon various organ systems. The majority of 24 and 32mg dose groups, respectively.[87]
adverse effects presented in this article were ob-
4.2.2 Dose Titration
served in the multicentre clinical trials with vari-
The incidence of gastrointestinal adverse ef-
ous cholinesterase inhibitors.
fects decreases substantially when cholinesterase
4.2.1 Dose-Dependent Effects inhibitors are titrated every 4 to 6 weeks rather than
The most often reported adverse effects with over shorter time periods.[73,83-85] The design of the
cholinesterase inhibitors occur in the gastrointes- clinical trials with rivastigmine with a forced
tinal tract. A summary of these effects is presented weekly dose titration scheme is typical. [84,96] In
in table IV. Increasing dosages resulted in in- these studies, patients had to reach their assigned
creased adverse effects. Of the gastrointestinal ad- dosage range by week 7 or be discontinued from
verse effects, the most frequently reported were the study. A slower dose titration can greatly re-
nausea, vomiting and diarrhoea. The incidence of duce the incidence of adverse effects with
nausea and vomiting was reported to increase with rivastigmine. When the maintenance dosages were
tacrine doses from 20 to 80mg.[94] In the tacrine achieved, the frequency of adverse effects in the
high dose study, where maximum daily dosages of high dosage group decreased to nausea 20%, vomi-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
736 Jann et al.

Table IV. Summary of the main gastrointestinal adverse effects of cholinesterase inhibitors
References Drug Dosage (mg/day) Patients with adverse effect (%)a
nausea vomiting diarrhoea
94 Tacrine P 3.2 3.2 3.2
20 4.7 4.7 3.4
40 5.9 5.9 3.2
80 11.7 11.7 10.0
73,95 Donepezil P 7, 7 2, 4 7, 4
5 4, 7 3, 4 9, 10
10 17, 24 10, 16 17, 16
84,94 Rivastigmineb P 11, 10 3, 6 NR, 9
1-4 14, 17 7, 8 NR, 10
6-12 48, 50 27, 34 NR, 17
85,87 Galantamine P 4.5, 3.1 1.4, 7.5 5.9, 9.9
8 5.7, NA 3.6, NA 5.0, NA
16 13.3, NA 6.1, NA 12.2, NA
24 16.5, 37.3 9.9, 20.8 5.5, 12.3
32 NA, 43.6 NA, 25.6 NA, 19.4
a Where two values are shown, these represent data from separate trials.
b Forced dose titration data.
NA = not applicable; NR = not reported; P = placebo.

ting 16% and diarrhoea 17%. These findings were fects, tremors, restlessness, sweating, delusions,
similar to rates of adverse effects reported with faintness and skin rash. Patients with adverse ef-
donepezil.[35] Similar results occurred with done- fects had significantly higher mean serum concen-
pezil and galantamine titrated every 4 to 6 weeks. trations than patients without adverse effects (16
Gradual dose titration methods are practical and vs 7 μg/L, p < 0.001). Women with a concentration
often used in the clinical setting. This time period of 17 μg/L had a higher frequency of adverse ef-
of 4 to 6 weeks is consistent among these agents, fects than women who had a concentration of 7
and may represent a feature of cholinesterase in- μg/L (borderline significant, p value not provided).
hibitors whereby the body needs time to adjust to Men with a concentration of 13 μg/L had a signifi-
the pharmacodynamic effects of enzyme inhibi- cantly higher incidence of adverse effects than men
tion. Another factor is drug absorption rate, since with a concentration of 8 μg/L (p < 0.01).
cholinesterase inhibitors that are rapidly absorbed
4.2.4 Hepatotoxicity with Tacrine
may cause a rapid stimulation of the cholinergic
The hepatotoxicity associated with tacrine was
system with adverse effects closely following.[10]
well documented in clinical trials.[83,94] Women
Administering the drug with food can lower the
were reported to be more susceptible than men to
amount of gastrointestinal effects by delaying tmax.
developing elevated (three times greater than the
Donepezil has a longer tmax of 3 to 5 hours, and may
normal range) serum alanine aminotransferase
not have this problem.
(ALT).[97] A dose-dependent relationship between
4.2.3 Serum Concentration Relationships hepatotoxicity and tacrine has not been estab-
A therapeutic window for tacrine serum con- lished, since its peak occurrence is between 4 and
centration was suggested to be between 7.5 and 20 10 weeks of initial treatment with patients on lower
μg/L, and concentrations were reported to predict dosages of 40 to 80 mg/day.[97] Correlations be-
adverse effects.[48] Symptomatic adverse effects tween serum tacrine concentrations and hepatotoxi-
were defined as the presence of gastrointestinal ef- city were not found.[48]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 737

A link between tacrine-induced ALT elevations need to design appropriate outcome measures for
and the genotype of glutathione S-transferase has the long-term treatment of these patients.[100]
been proposed, where patients with hepatoxicity
had combined deficient alleles of M1 (mu) and T1 Acknowledgements
(tau) isoenzymes.[22] Analysis of only one geno-
type could not provide a significant link with sus- Kara L. Shirley, Pharm.D., at the time of this paper was
the Novartis Fellow in Clinical Neuropsychopharmacology,
ceptibility to hepatotoxicity.[98] Therefore, geno- and received only salary support from Novartis for her
typing for these two isoenzymes may be needed for training. Michael W. Jann, Pharm.D., has received research
patients who are likely candidates for tacrine ther- grant support from Novartis and Janssen, Otsuka, Bristol
apy. However, with the use of tacrine use dimini- Myers Squibb, Solvay, Glaxo Smith Kline, Lipocine, Lilly
shing, the likelihood of genotyping may not be and Pfizer Pharmaceuticals.
economically justified.
References
1. Martin JB. Molecular basis of the neurodegenerative disorders.
N Engl J Med 1999; 340: 1970-80
5. Conclusions 2. Parnetti L, Senin U, Mecocci P. Cognitive enhancement ther-
apy for Alzheimer’s disease. Drugs 1997; 53: 752-68
3. Sramek JJ, Cutler NR. Recent developments in the drug treat-
Cholinesterase inhibitors have become the ment of Alzheimer’s disease. Drugs Aging 1999; 14: 359-73
‘first-line’ agents in the treatment of AD.[99] A 4. VanDenBerg CM, Kazmi Y, Jann MW. Cholinesterase inhibi-
tors for the treatment of Alzheimer’s disease in the elderly.
thorough understanding of their pharmacokinetic Drugs Aging 2000; 16: 123-36
and pharmacodynamic profiles is necessary in 5. Court JA, Perry EK. CNS nicotinic receptors. CNS Drugs 1994;
2: 216-33
order to maximise therapeutic benefits while 6. Practico D, Delanty N. Oxidative injury in diseases of the cen-
minimising adverse events. With the exception of tral nervous system: focus on Alzheimer’s disease. Am J Med
2000; 109: 577-85
low doses of tacrine, cholinesterase inhibitors have 7. Jick H, Zornberg GL, Jick SS, et al. Statins and the risk of
linear pharmacokinetics, but have nonlinear phar- dementia. Lancet 2000; 356: 1627-31
8. Small GW, Rabins PV, Barry PP, et al. Diagnosis and treatment
macodynamic actions in the inhibition of cholin- of Alzheimer’s disease and related disorders. JAMA 1997;
esterase enzymes. Measurement of enzyme inhibi- 278: 1363-71
9. Taylor P. Anticholinesterase agents. In: Hardman JG, Limbird
tion is the only method currently widely used as a LE, editors. Goodman’s and Gilman’s the pharmacological
biomarker for cholinesterase inhibitors. A sigmoi- basis of therapeutics. 9th ed. New York: McGraw-Hill, 1996:
161-75
dal curve for the relationship between cholinester- 10. Imbimbo BP. Pharmacodynamic tolerability relationships of
ase inhibition and efficacy has been proposed. A cholinesterase inhibitors for Alzheimer’s disease. CNS Drugs
2001; 15: 375-90
similar, but slightly different, sigmoidal curve has 11. Cummings JL. Cholinesterase inhibitors: a new class of psy-
been proposed for the adverse event profile. Un- chotropic compounds. Am J Psychiatry 2000; 157: 4-15
12. Crismon ML. Tacrine: first drug approved for Alzheimer’s dis-
derstanding these relationships can identify the ease. Ann Pharmacother 1994; 28: 744-51
maximum tolerated dose for cholinesterase inhibi- 13. Parnetti L. Clinical pharmacokinetic of drugs for Alzheimer’s
disease. Clin Pharmacokinet 1995; 29: 110-29
tors. 14. Forsyth DR, Wilcock GK, Morgan RA, et al. Pharmacokinetics
Future pharmacodynamic methods to evaluate of tacrine hydrochloride in Alzheimer’s disease. Clin Phar-
macol Ther 1989; 46: 634-41
cholinesterase inhibitors may include the use of 15. Cutler NR, Sedman AJ, Prior P, et al. Steady-state pharmaco-
various imaging techniques. Coupled with clinical kinetics of tacrine in patients with Alzheimer’s disease. Psy-
chopharmacol Bull 1990; 26: 231-4
observations and/or standardised assessments of 16. Hartvig P, Askmark H, Aquilonius SM, et al. Clinical pharmaco-
neurochemical biomarkers such as β-amyloid or kinetics of intravenous and oral 9-amino-1,2,3,4-tetra-
hydroacridine, tacrine. Eur J Clin Pharmacol 1990; 38:
tau protein measurements in CSF or plasma, these 259-63
may represent future tools for monitoring the ef- 17. Madden S, Spaldin V, Park BK. Clinical pharmacokinetics of
tacrine. Clin Pharmacokinet 1995; 28: 449-57
fects of cholinesterase inhibitors. Long-term trials 18. Nordberg A, Svensson AL. Cholinesterase inhibitors in the
have demonstrated the continued benefits of cho- treatment of Alzheimer’s disease. Drug Saf 1998; 19: 465-80
19. Wagstaff A, McTavish D. Tacrine: a review of its pharmaco-
linesterase inhibitors in patients with AD.[76] Cli- logical and pharmacokinetic properties and therapeutic po-
nicians, caregivers and healthcare organisations tential in Alzheimer’s disease. Drugs Aging 1994; 4: 1-31

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
738 Jann et al.

20. Makela PM, Truman CA, Ford JM, et al. Plasma protein binding 43. Johansson IM, Nordberg A. Pharmacokinetic studies of cholin-
characteristics of tacrine hydrochloride (THA) [abstract]. Br esterase inhibitors. Acta Neurol Scand 1993; 149: 22-5
J Clin Pharmacol 1994; 38: 489P 44. Bickel U, Thomsen T, Weber W, et al. Pharmacokinetics of
21. Grothe DR, Piscitelli SC, Dukoff R, et al. Penetration of tacrine galantamine in humans and corresponding cholinesterase in-
into cerebrospinal fluid in patients with Alzheimer’s disease. hibition. Clin Pharmacol Ther 1991; 50: 420-8
J Clin Psychopharmacol 1998; 18: 78-81 45. Davidson P, Blennow K, Andreasen N, et al. Differential in-
22. Simon T, Becquemont L, Mary-Krause M, et al. Combined glu- crease in CSF acetylcholinesterase after treatment with ace-
tathione-S-transferase M1 and T1 genetic polymorphism and tylcholinesterase inhibitors in patients with Alzheimer’s
tacrine hepatotoxicity. Clin Pharmacol Ther 2000; 67: 432-7 disease. Neurosci Lett 2001; 30: 157-60
23. McGleenon BM, Dyman KB, Passmore AP. Acetylcholinester- 46. Bachus R, Bickel U, Thomsen T, et al. The O-demethylation of
ase inhibitors in Alzheimer’s disease. Br J Clin Pharmacol the antidementia drug galantamine is catalysed by cyto-
1999; 48: 471-80 chrome P450 2D6. Pharmacogenetics 1999; 9: 661-8
24. Murphy MF, Hardiman ST, Nash RJ, et al. Evaluation of HP- 47. Fontana RJ, deVries TM, Woolf TF, et al. Caffeine based meas-
029 (velnacrine maleate) in Alzheimer’s disease. Ann N Y ures of CYP 1A2 correlate with oral clearance of tacrine in
Acad Sci 1991; 640: 253-62 patients with Alzheimer’s disease. Br J Clin Pharmacol 1998;
25. Dooley M, Lamb MH. Donepezil. Drugs Aging 2000; 16: 199-226 46: 221-8
26. Barner EL, Gray SL. Donepezil use in Alzheimer’s disease. 48. Ford JM, Truman CA, Wilcock GK, et al. Serum concentrations
Ann Pharmacother 1998; 32: 70-7 of tacrine hydrochloride predict its adverse effects in Alz-
27. Rogers SL, Friedhoff LT. Pharmacokinetics and pharmacody- heimer’s disease. Clin Pharmacol Ther 1993; 53: 691-5
namic profile of donepezil HCL following single oral dose. 49. Welty D, Pool W, Woolf T, et al. The effect of smoking on the
Br J Clin Pharmacol 1998; 46 Suppl. 1: 1-6 pharmacokinetics and metabolism of Cognex in healthy vol-
28. Rogers SL, Cooper NM, Sukovarty R, et al. Pharmacokinetics unteers [abstract]. Pharm Res 1993; 10: S344
and pharmacodynamic profile of donepezil HCl following 50. Tiseo PJ, Perdomo CA, Friedhoff LT. Concurrent administra-
multiple oral dose. Br J Clin Pharmacol 1998; 46 Suppl. 1: tion of donepezil HCl and digoxin: assessment of pharmaco-
7-12 kinetic changes. Br J Clin Pharmacol 1998; 46 Suppl. 1: 40-4
29. Rogers SL, Doody RS, Mohs RC, et al. Donepezil improves 51. Forgue ST, Reece PA, Sedman AJ, et al. Inhibition of tacrine
cognition and global function in Alzheimer’s disease. Arch oral clearance by cimetidine. Clin Pharmacol Ther 1998; 59:
Intern Med 1998; 158: 1021-31 444-9
30. Shintani EY, Uchida KM. Donepezil: an anticholinesterase in- 52. Laine K, Palovaara S, Tapanainen P, et al. Plasma tacrine con-
hibitor for Alzheimer’s disease. Am J Health Syst Pharm centrations are significantly increased by concomitant hor-
1997; 54: 2805-10 mone replacement therapy. Clin Pharmacol Ther 1999; 66:
602-8
31. Tiseo PJ, Perdomo CA, Friedhoff LT. Metabolism and elimina-
53. Becquemont L, LeBot MA, Riche C, et al. Use of heterolo-
tion of 14C donepezil in healthy volunteers: a single dose
gously expressed human cytochrome P450 1A2 to predict
study. Br J Clin Pharmacol 1998; 46 Suppl. 1: 19-24
tacrine-fluvoxamine drug interaction in man. Pharmacogenet-
32. Ohnishi A, Mihara M, Kamakura H, et al. Comparison of the ics 1998; 8: 101-8
pharmacokinetics of E2020, a new compound for Alz-
54. Becquemont L, Ragueneau I, LeBot MA, et al. Influence of the
heimer’s disease, in healthy young and elderly subjects. J Clin CYP 1A2 inhibitor fluvoxamine on tacrine pharmacokinetics
Pharmacol 1993; 33: 1086-91 in humans. Clin Pharmacol Ther 1997; 61: 619-27
33. Tiseo PJ, Vargas R, Perdono CA, et al. An evaluation of the 55. Tiseo PJ, Perdomo CA, Friedhoff LT. Concurrent administra-
pharmacokinetics of donepezil hydrochloride in patients with tion of donepezil HCl and cimetidine: assessment of pharma-
impaired hepatic function. Br J Clin Pharmacol 1998; 46 cokinetic changes following single and multiple doses. Br J
Suppl. 1: 51-5 Clin Pharmacol 1998; 46 Suppl. 1: 25-9
34. Tiseo PJ, Foley K, Friedhoff LT, et al. An evaluation of the 56. Tiseo PJ, Perdomo CA, Friedhoff LT. Concurrent administra-
pharmacokinetics of donepezil hydrochloride in patients with tion of donepezil HCl and ketoconazole: assessment of phar-
impaired moderate to severe renal impairment. Br J Clin Phar- macokinetic changes following single and multiple doses. Br
macol 1998; 46 Suppl. 1: 56-60 J Clin Pharmacol 1998; 46 Suppl. 1: 30-4
35. Jann MW. Rivastigmine, a new generation cholinesterase inhib- 57. Carrier L. Donepezil and paroxetine: possible drug interaction
itor for the treatment of Alzheimer’s disease. Pharmacothe- [letter]. J Am Geriatr Soc 1999; 47: 1037
rapy 2000; 20: 1-12 58. Rojas-Fernandez C, Fisher C. Drug interactions with donepezil.
36. Sramek JJ, Anand R, Wardie TS, et al. Safety/tolerability trial J Am Geriatr Soc 2000; 48: 597-8
of SDZ 713 in patients with probable Alzheimer’s disease. 59. Verrico MM, Nace DA, Towers AL. Fulminant chemical hep-
Life Sci 1996; 58: 1201-7 atitis possibly associated with donepezil and sertraline ther-
37. Spencer CM, Noble S. Rivastigmine. Drugs Aging 1998; 13: apy. J Am Geriatr Soc 2000; 48: 1659-63
391-411 60. Ravic M, Warrington S, Boyce M. The safety and pharmacoki-
38. Kennedy JS, Polinsky RJ, Johnson B, et al. Preferential cere- netics of thioridazine during co-administration with donepezil
brospinal fluid acetylcholinesterase inhibition by rivastigm- [abstract]. Clin Pharmacol Ther 2001; 69: P44
ine in humans. J Clin Psychopharmacol 1999; 19: 513-21 61. Reyes JF, Preskorn S, Khan A, et al. Pharmacokinetics of
39. Cutler NR, Polinsky RJ, Sramek JJ, et al. Dose-dependent CSF donepezil and risperidone in males with schizophrenia [ab-
acetylcholinesterase inhibition by SDZ ENA 713 in Alz- stract]. Clin Pharmacol Ther 2001; 69: P44
heimer’s disease. Acta Neurol Scand 1998; 97: 244-50 62. Tiseo PJ, Foley K, Friedhoff LT. Concurrent administration of
40. Schran HF, Habucky K, Mancione LC, et al. The effects of renal donepezil HCl and theophylline: assessment of pharmacoki-
and hepatic impairment on the disposition of acetylcholines- netic changes following multiple doses in healthy volunteers.
terase SDZ ENA 713 [abstract]. Pharm Res 1996; 13: S-428 Br J Clin Pharmacol 1998; 46 Suppl. 1: 35-9
41. Scott LJ, Goa KL. Galantamine. Drugs 2000; 60: 1095-122 63. Tiseo PJ, Foley K, Friedhoff LT. The effect of multiple doses
42. Mihailova D, Yamboliev I, Zhivkova Z, et al. Pharmacokinetic of donepezil HCl on the pharmacokinetics and pharmacody-
of galantamine hydrobromide after single subcutaneous and namics of warfarin. Br J Clin Pharmacol 1998; 46 Suppl. 1:
oral dosage in humans. Pharmacology 1989; 39: 50-8 45-50

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)
Cholinesterase Inhibitors 739

64. Grosssberg GT, Stamelin HB, Messina JC, et al. Lack of ad- 85. Tariot PN, Solomon PR, Morris JC, et al. A 5-month random-
verse pharmacodynamic drug interactions with rivastigmine ized placebo controlled trial of galantamine in AD. Neurol-
and twenty two classes of medications. Int J Geriatr Psychiatr ogy 2000; 54: 2269-76
2000; 15: 242-7 86. Anand R, Messina J, Hartman R. Dose-response effect of
65. Jann MW. Preclinical pharmacology of metrifonate. Pharma- rivastigmine in the treatment of Alzheimer’s disease. Int J
cotherapy 1998; 18 (2 pt 2): 55S-67S Geriatr Psychopharmacol 2000; 2: 68-72
66. Rivastigmine (Exelon®) package insert. Physician’s desk refer- 87. Raskind MA, Peskind ER, Wessel T, et al. Galantamine in AD:
ence. Montvale (NJ): Medical Economic Company, Inc., 2000 a 6-month randomized placebo controlled trial with a 6-
67. Taylor P, Radic Z. The cholinesterases: from genes to proteins. month extension. Neurology 2000; 54: 2261-8
Annu Rev Pharmacol Toxic 1994; 34: 281-320 88. Kaufer DI, Cummings JL, Christine D. Effect of tacrine on
68. Guillozet AL, Smiley JF, Mash DC, et al. Butyrylcholinesterase behavioral symptoms in Alzheimer’s disease: an open label
in the life cycle of amyloid plaques. Ann Neurol 1997; 42: study. J Geriatr Psychiatry Neurol 1996; 9: 1-6
909-18 89. Weiner MF, Martin-Cook K, Foster BM, et al. Effects of
69. Giacobini E. Do cholinesterase inhibitors have disease-modify- donepezil on emotional/behavioral symptoms in Alzheimer’s
ing effects in Alzheimer’s disease? CNS Drugs 2001; 15: disease patients. J Clin Psychiatry 2000; 61: 487-92
85-91
90. Hunter R, Wyper DJ, Patterson J, et al. Cerebral pharmacody-
70. Cummings JL. Metrifonate: overview of safety and efficacy.
namics of physostigmine in Alzheimer’s disease investigated
Pharmacotherapy 1998; 18 (2 pt 2): 43S-6S
using SPECT. Br J Psychiatry 1991; 158: 351-7
71. Maelicke A, Samochocki M, Jostock R, et al. Allosteric sensi-
tization of nicotinic receptors by galantamine, a new treat- 91. Cohen MB, Fitten LJ, Lake RR, et al. SPECT brain imaging in
ment for Alzheimer’s disease. Biol Psychiatry 2001; 49: Alzheimer’s disease during treatment with oral THA and lec-
279-88 ithin. Clin Nucl Med 1992; 17: 312-5
72. Mant T, Troetel WM, Imbimbo BP. Maximum tolerated dose 92. Bookheimer SY, Strojwas H, Cohen MS, et al. Patterns of brain
and pharmacodynamics of eptastigmine in elderly healthy activation in people at risk for Alzheimer’s disease. New
volunteers. J Clin Pharmacol 1998; 38: 610-7 Engl J Med 2000; 343: 450-6
73. Rogers SL, Farlow MR, Doody RS, et al. A 24-week double 93. Small GW. Structural and functional imaging of Alzheimer’s dis-
blind placebo controlled trial of donepezil in patients with ease. In: Charney DS, Coyle J, Davis K, et al., editors. Psycho-
Alzheimer’s disease. Neurology 1998; 50: 136-45 pharmacology: the fifth generation of progress. Philadelphia:
74. Davis CS. Statistical issues in the design and analysis of clinical Lippincott, Williams and Wilkens, 2002: 1231-42
trials in Alzheimer’s disease. Drug Inf J 1996; 30: 339-50 94. Farlow M, Gracon SI, Hershey LA, et al. A controlled trial of
75. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alz- tacrine in Alzheimer’s disease. JAMA 1992; 268: 2523-9
heimer’s disease. Am J Psychiatry 1984; 141: 1356-64 95. Burns A, Rosser M, Hecker J, et al. The effect of donepezil in
76. Schneider LS, Olin JT, Doody RS, et al. Validity and reliability Alzheimer’s disease: results from a multinational trial. De-
of the Alzheimer’s disease cooperative study: clinical global ment Geriatr Cogn Disord 1999; 10: 237-44
impression of change. Alzheimer Dis Assoc Disord 1997; 11 96. Rosler M, Anand R, Cicin-Sain A, et al. Efficacy and safety of
(S2): S22-32 rivastigmine in patients with Alzheimer’s disease: interna-
77. Giacobini E. Cholinesterase inhibitors for Alzheimer’s disease tional randomized controlled trial. BMJ 1999; 318: 633-40
therapy: from tacrine to future applications. Neurochem Lett 97. Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic
1998; 32: 413-9 effects of tacrine administration in patients with Alzheimer’s
78. Folstein MF, Folstein SE, McHugh PR. Mini-mental state disease. JAMA 1994; 271: 992-8
exam. J Psychiatr Res 1975; 12: 189-98 98. Green VJ, Pirmohamed M, Kitteringham NR, et al. Glutathione
79. Cummings JL, Mega M, Gray K, et al. The neuropsychiatric S-transferase μ genotype (GSTM 1*0) in Alzheimer’s pa-
inventory. Neurology 1994; 44: 2308-14 tients with tacrine transaminitis. Br J Clin Pharmacol 1995;
80. DeJong R, Osterlund OW, Roy GW. Measurement of quality 39: 411-5
of life changes in patients with Alzheimer’s disease. Clin 99. American Psychiatric Association. Practice guideline for the
Ther 1989; 11: 545-54 treatment of patients with Alzheimer’s disease and other de-
81. Krall WJ, Sramek JJ, Cutler NR. Cholinesterase inhibitors: a mentias of late life. Am J Psychiatry 1997; 154 (5): 1-39
therapeutic strategy for Alzheimer’s disease. Ann Phar- 100. Steele CD, Steinberg M, Miles G, et al. Recognition and man-
macother 1999; 33: 441-50 agement of early Alzheimer’s disease. J Clin Outcome Man-
82. Farlow MR, Evans RM. Pharmacologic treatment of cognition in agement 1999; 6: 52-68
Alzheimer’s dementia. Neurology 1998; 51 Suppl. 1: S36-44
83. Knapp MJ, Knopman DS, Solomon PR, et al. A 30-week ran-
domized controlled trial of high-dose tacrine in patients with
Alzheimer’s disease. JAMA 1994; 271: 985-91
84. Corey-Bloom J, Anand R, Veach J, et al. A randomized trial Correspondence and offprints: Dr Michael W. Jann, South-
evaluating the efficacy and safety of ENA 713 (rivastigmine
ern School of Pharmacy, Mercer University, 3001 Mercer
tartrate), a new acetylcholinesterase inhibitor, in patients with
mild to moderately severe Alzheimer’s disease. Int J Geriatr University Drive, Atlanta, GA 30341, USA.
Psychopharmacol 1999; 1: 55-65 E-mail: jann_mw@mercer.edu

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (10)

Das könnte Ihnen auch gefallen