Chemical structure of tacrine
Find information on thousands of medical conditions and prescription drugs.

Tacrine

Tacrine is a parasympathomimetic and a centrally acting cholinesterase inhibitor (anticholinesterase). It was the first centrally-acting cholinesterase inhibitor approved for the treatment of Alzheimer's disease, and was first synthesised at the Department of Pharmacology at the University of Sydney. more...

Home
Diseases
Medicines
A
B
C
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
Oxytetracycline
Phentermine
Tacrine
Tacrolimus
Tagamet
Talbutal
Talohexal
Talwin
Tambocor
Tamiflu
Tamoxifen
Tamsulosin
Tao
Tarka
Taurine
Taxol
Taxotere
Tazarotene
Tazobactam
Tazorac
Tegretol
Teicoplanin
Telmisartan
Temazepam
Temocillin
Temodar
Temodar
Temozolomide
Tenex
Teniposide
Tenoretic
Tenormin
Tenuate
Terazosin
Terbinafine
Terbutaline
Terconazole
Terfenadine
Teriparatide
Terlipressin
Tessalon
Testosterone
Tetrabenazine
Tetracaine
Tetracycline
Tetramethrin
Thalidomide
Theo-24
Theobid
Theochron
Theoclear
Theolair
Theophyl
Theophyl
Theostat 80
Theovent
Thiamine
Thiomersal
Thiopental sodium
Thioridazine
Thorazine
Thyroglobulin
Tiagabine
Tianeptine
Tiazac
Ticarcillin
Ticlopidine
Tikosyn
Tiletamine
Timolol
Timoptic
Tinidazole
Tioconazole
Tirapazamine
Tizanidine
TobraDex
Tobramycin
Tofranil
Tolazamide
Tolazoline
Tolbutamide
Tolcapone
Tolnaftate
Tolterodine
Tomoxetine
Topamax
Topicort
Topiramate
Tora
Toradol
Toremifene
Tracleer
Tramadol
Trandate
Tranexamic acid
Tranxene
Tranylcypromine
Trastuzumab
Trazodone
Trenbolone
Trental
Trest
Tretinoin
Triacetin
Triad
Triamcinolone
Triamcinolone hexacetonide
Triamterene
Triazolam
Triclabendazole
Triclosan
Tricor
Trifluoperazine
Trilafon
Trileptal
Trimetazidine
Trimethoprim
Trimipramine
Trimox
Triprolidine
Triptorelin
Tritec
Trizivir
Troglitazone
Tromantadine
Trovafloxacin
Tubocurarine chloride
Tussionex
Tylenol
Tyrosine
U
V
W
X
Y
Z

Clinical uses

Tacrine was the prototypical cholinesterase inhibitor for the treatment of Alzheimer's disease. Its clinical effectiveness was hindered by poor oral bioavailability and considerable adverse drug reactions (including nausea, diarrhoea, urinary incontinence, and hepatotoxicity) such that few patients could tolerate therapeutic doses. Indeed there is some doubt as to its efficacy in humans at all.

Other newer cholinesterase inhibitors, such as donepezil, are now preferred over tacrine.

Sources

  • Brenner, G. M. (2000). Pharmacology. Philadelphia, PA: W.B. Saunders Company. ISBN 0-7216-7757-6

Read more at Wikipedia.org


[List your site here Free!]


Methodology, results and quality of clinical trials of tacrine in the treatment of Alzheimer's disease: a systematic review of the literature
From Age and Ageing, 3/1/98 by Jesus Lopez Arrieta

Keywords: Alzheimer's disease, clinical trials, meta-analysis, systematic review, tacrine

Introduction

Alzheimer's disease (AD) ranks among the principal public health problems confronting developed countries [1, 2]. Cholinergic deficiency is one of the main defects found in the brains of such patients and correlates with poor performance in cognitive tests [3, 4], although other mediators of neurotransmission have also been linked to AD physiopathology [5-10]. This finding led to the hypothesis that enhancing the cholinergic function of the central nervous system could slow the mental deterioration associated with the disease [12, 13]. An initial AD treatment strategy was thus based on administration of choline, but clinical studies failed to demonstrate its efficacy [14]. This was followed by studies of muscarinic agonists, such as bethanechol, arecoline and oxotremorine, but adverse effects and low selectivity for the central nervous system limited their clinical use [15]. Lastly, cholinesterase inhibitors were used in order to augment the availability of acetylcholine in neuron receptors. Among these drugs is tacrine (tetrahydroaminoacridine). This aminoacridine acts on the central nervous system in a manner that is both complex and diverse: its known pharmacological actions include longer lasting cholinesterase inhibition than physostigmine and restoration of nicotinic receptors in AD-affected brains [16-19].

Reviews of clinical trials help obtain clear scientific evidence on the benefits, risks and costs of any given therapeutic intervention and are a tool of evidence-based medicine [20]. Narrative reviews have been published describing trials of drugs, including tacrine, for the treatment of AD [21-29]. The chief advantages of systematic as opposed to narrative reviews are the lower probability and magnitude of biases along with greater repeatability [30, 31]. Accordingly, the aim of this study is to review systematically the methodological characteristics, results and quality of trials of tacrine in the treatment of AD.

Materials and methods

Study-inclusion criteria

Trials included were those conducted on AD patients, aged 40 years or over, assigned to tacrine- or placebo-based treatment. Trial participants were selected on the basis of the degree of probability of the diagnosis of AD, as indicated by National Institute of Neurological and Communicative Disorders and Stroke-Alzheimer's Disease and Related Disorders Association (NINCDS-ADRDA) criteria [32], and the degree of severity, as indicated by Diagnostic and Statistical Manual of Mental Disorders, third edition (DSM-III-R) criteria [33]. Information on tacrine efficacy was rated by one of the following types of scales: cognitive function, behavioural, functional ability, global impression of clinical change and global scales of mental deterioration.

Study-search strategy

Studies were identified in the following electronic databases: the Cochrane Library (which includes the Cochrane Database of Systematic Reviews, the Cochrane Controlled Trials Register, the Cochrane Review Methodology Database and the York Database of Abstracts of Reviews of Effectiveness) and MEDLINE, for the period 1 January 1980-1 May 1997, using the search strategy proposed by Dickersin et al. [34].

Extraction and presentation of data

Selection of trials for systematic review and data extraction purposes was carried out separately by two reviewers (J.L.A. and F.R.A.) working independently. Where differences of opinion arose, these were resolved by discussion between the two. Study reporting quality was assessed according to guidelines laid down by the Standards of Reporting Trials Group [35]. These guidelines list the information to be provided in the publication of a random clinical trial and are useful for evaluating the quality of the reporting but not that of the study per se. Overall scores were obtained by totalling the points for 32 items, grouped within the following areas: description of the allocation of participants to each treatment; description of the blinding of the intervention and whether this proved successful; description of subject follow-up and possible losses to follow-up; and description of the statistical analysis in the Methods and Results sections of the paper. The presence of each piece of information is awarded a score of 1, and its absence 0, so the maximum possible score for any paper is 32. Total scores for the paper and subscores for each of its sections are provided as absolutes and as a percentage of the possible maximum. Due to the heterogeneity of trial methods and the low number of studies published, no statistical synthesis of study results has been drawn up.

Results

We located 49 publications on the efficacy of tacrine in the treatment of AD, published in the period 1981-1996 [36-84]. Of these, one was a summary of a communication delivered to a congress [74], three were chapters of a book [39, 40, 45] and the remainder were papers included in MEDLINE.

Of the 49 publications, 22 corresponded to non-randomized and/or open clinical trials and the remaining 27 reported 18 double-blind, randomized clinical trials. To identify the most appropriate tacrine dosage, three of the 18 random trials included titration phases with randomized, double-blind, treatment allocation. These trials--reported by Forette et al [74-75], by Davis et al. [76-77] and Foster et al. [84]--were therefore deemed to be studies in their own right, independent of the main study, thereby bringing the figure of random trials in this review to 21.

The methodological characteristics and results of the 22 non-randomized and/or open trials are summarized in Table 1. The first trials were notable for their smaller sample size and the fact that tacrine-treatment dosage and duration were variable, ranging from single doses to 9 weeks of treatment. 1990 signalled the commencement of longer studies. The follow-up phases of random trials of tacrine were found among these studies, which also included mortality or degree of institutionalization as outcome variables [57]. All trials used psychometric instruments to rate patients' mental function. Furthermore, in six trials, brain function techniques--positron emission tomography and single photon computed tomography imaging [48-50, 52, 55, 56]--were employed, in four trials the electroencephalographic register was studied [44-47] and in two, indirect methods were used to evaluate the cholinergic effect of tacrine [43, 51]. In seven studies, lecithin was associated with tacrine [37, 39, 41, 42, 46-48]. In all the open studies there was improvement in some of the variables measured; only in the studies conducted by Summers et al. [36], Kaye et al. [37], Gauthier et al. [39, 41], Alhainen et al. [43] and Knopman et al. [57] did such improvement attain statistical significance, although, on occasion, discontinuation of tacrine led to a reversal to the baseline situation [39, 41]. In seven trials, adverse events were described for almost 60% (19-80%) of subjects treated with tacrine [36, 38-42, 54].

[TABULAR DATA 1 NOT REPRODUCIBLE IN ASCII]

Table 2 summarizes the design, treatment characteristics and patient-enrolment criteria in the 21 random clinical trials of tacrine in AD. Of these, eight were parallel and 13 cross-over in design. Tacrine dosage ranged from 25 to 200 mg/day. Duration of treatment ranged from 3 to 36 weeks, with a mean [+ or -] standard deviation of 9.3 [+ or -] 8.6 weeks (6.53 [+ or -] 3.8 weeks in the cross-over and 13.9 [+ or -] 12.3 weeks in the parallel random trials). Only two studies [78, 82] used a pre-established tacrine dosage; the remaining trials included a preliminary titration phase. Mean age of participants was 68.9 years: 67.6 years in the cross-over studies and 70.7 years in the parallel-group random trials. In 17 random trials, NINCDS-ADRDA criteria were used for diagnosis of AD among participants and in seven random trials DSM-III-R criteria were used to assess severity of mental deterioration. Subject enrolment was based on diagnosis of probable AD of mild to moderate severity.

[TABULAR DATA 2 NOT REPRODUCIBLE IN ASCII]

In all, 3555 patients were enrolled. Parallel-group random trials had a larger sample size, with 2244 patients enrolled versus 1424 in the cross-over random trials (Table 2). A total of 1149 subjects failed to complete the trial, yielding a mean of 33.4% patients per trial. Of these, 1116 (97%) patients received treatment with tacrine. Adverse events, generally in the form of cholinergic manifestations (digestive symptoms, sweating) or serum transaminase elevations exceeding three times the upper limit of normal, appeared in 59.3% of individuals; 30.2% suffered some type of cholinergic symptom and 28.6% showed hepatotoxicity (Table 3). Adverse events were the chief reason for patient withdrawal.

[TABULAR DATA 3 NOT REPRODUCIBLE IN ASCII]

The 21 random trials employed 157 outcome variables, with 3-31 used per trial, except for the Forette et al. [74-75] and Davis et al. [74, 75] titration phases where only the Mini-Mental State Examination (MMSE) [85] was used. Among the instruments used for studying intellectual functions, the MMSE proved to be the most frequent (Table 4). Of the behavioural scales, the most common was the Alzheimer's Disease Assessment Scale (ADAS) non cognitive subscale [86]. The most widely used functional scales were Lawton's [87, 88], particularly the instrumental activities of daily living scale and physical self-maintenance scale. General impression tests were used to grade assessor-based subjective aspect of improvement, with the clinician's global impression of change [89] being most in evidence. The global scale of intellectual deterioration most widely favoured was the total score yielded by Al)AS [86] rating (Table 4). Only seven studies [69-73, 74-75, 78, 80, 82-84] specified the primary and secondary assessment variables at the outset of the trial.

ADAS, Alzheimer's disease assessment scale; MMSE, mini-mental state examination; WMS, Wechsler memory scale.

The results on tacrine efficacy in the 21 random trials are summarized in Table 5. In 12 trials, tacrine produced statistically significant improvement (P [is less than] 0.05) in one or more outcome variable, in five trials the drug led to non-significant improvement and in the remainder scarcely any difference was found between tacrine and placebo. The improvement was in cognitive variables in 12 instances and it was in overall ratings in seven. Instruments of functional ability and global clinical impression ratings showed improvements in three trials each and in only one case did tacrine show efficacy in a behavioural scale. Bearing in mind that of the subjects exposed to tacrine, 24.4% discontinued the treatment and 50.2% manifested adverse effects which generally forced them to reduce the dosage, only a little over 20% of patients initially treated with tacrine achieved some clinical benefit as against the placebo.

[TABULAR DATA 5 NOT REPRODUCIBLE IN ASCII]

In line with Schneider's classification [20], random trials were divided according to sample size into small [58-61, 64-68, 79] (having under 50 participants) and large [62, 63, 69-78, 80-84] (having over 50 participants). The seven small trials were carried out earlier and all but one had a cross-over design. Of the seven, tour showed some significant difference (P [is less than] 0.05) in favour of tacrine and three yielded negative results. Of the large-scale trials, half were parallel and all were carried out more recently. The results in 12 of these 14 showed clinical improvements attributable to tacrine.

The reporting quality of the 21 random trials is shown in Table 6. No study gave a description of the concealment of the randomization sequence or the success of the double-blind procedure. Over 50% of the relevant information was reported only for the following areas: coverage of follow-up and statistical analyses in the Results section of the paper (Table 6). Cross-over random trials supplied 30.7% and parallel-group random trials 40.2% of all relevant information. The study with the best reporting quality was that by Knapp et al. [82], which supplied 50% of the relevant information.

Table 6. Reporting quality of randomized clinical trials of tacrine in Alzheimer's disease

(a) Percentage of the maximum possible score: assessment as per Guidelines of the Standards of Reporting Trials Group (1994) [35].

Discussion

This systematic review has pinpointed three stages in clinical research on tacrine in AD. The first stage comprises trials conducted in the 1980s and early 1990s, with small sample sizes, non-randomized and/or open designs and relatively small doses of tacrine over short periods of time [36-45] (Table 1). These studies generated excessively optimistic expectations as to the benefits of tacrine. Their chief contribution was to demonstrate the hepatotoxic effects of tacrine, even at low doses, and to stimulate research in this field.

The second stage was of trials that were more methodologically sound, characterized by randomized allocation of treatment and conducted in the second half of the 1980s and in the 1990s (Tables 2-6) [58-84]. The considerable number of scales used in the random trials in order to rate response to treatment (Table 4) suggests that none proved altogether satisfactory [90]. Moreover, there are serious shortcomings in the reporting of the results of these studies. Particularly noteworthy is the lack of information on the concealment of the randomization sequence and the success of the double-blind procedure, since there is empirical evidence to show that this can spuriously magnify the effect estimators [91, 92].

Among these `second-stage' random trials, the most informative are those by Farlow et al. [78] and Knapp et al. [82], together with the comparative double-blind phases of the studies by Forette et al. [74, 75] and Davis et al. [76, 77]. These studies have a large sample size, thereby enabling more precise measurement of the effect of tacrine, and a parallel design, which makes it possible to control for time-dependent confounders. This is important as the clinical course of Al) varies widely, with periods of cognitive stabilization of up to 2 years [93]. Inclusion criteria are clear (DSM-III-R and NINCDS-ADRDA), affording relative assurance on the diagnosis and homogeneity of patients studied. In most cases the ADAS scale was used as the principal variable of assessment, this scale being one of the most reliable having been drawn up to evaluate the therapeutic effects of drugs in AD [94]. Furthermore, these studies report the relationship between tacrine dosage and the drug's efficacy and toxicity. Finally, they earned some of the highest ratings for reporting quality (Table 6).

The third stage takes in studies that provided continued follow-up for some `second-stage' random trials and were carried out in the period 1993-96 [52-57] (Table 1). They supply information on the long-term effects (safety) of tacrine and, in certain cases, on its effect t)n the quality of life, institutionalization and mortality of AD patients [57]. The chief limitation on these studies lies in their lack of a control group (ideally composed of untreated AD patients), with the result that they have a limited capacity for drawing inferences on clinical usefulness.

`Second-stage' random trials have shown that tacrine has a modest dose-dependent efficacy in cases of mild to moderate AD. The greatest benefits are obtained at doses of 120-160 mg/day [82], although it is possible to obtain certain benefits at a dosage of 80 mg/day, thereby ameliorating the adverse effects of treatment [78]. The benefit yielded by tacrine is particularly evident in cognitive function, functional ability and global scales of mental deterioration. The clinical relevance of benefit may be inferred by comparison with the usual evolution of cognitive function in AD. The MMSE falls by 3 points every 6-12 months [95] and the cognitive component of the ADAS rises by 7 points per year [96] in mild to moderate AD in the absence of treatment. It is therefore estimated that tacrine, at a dosage of 80-160 mg/day over a period of 6-30 weeks, slows the course of AD by approximately 6 months [71, 74-76, 78, 82]. This benefit is considered clinically relevant by the US Food and Drug Administration [97], even when it represents no more than 10% of the clinical phase of the disease [98, 99].

However, the adverse effects of tacrine--both cholinergic and hepatic--mean that only one-third of all patients receive the more effective dosage levels of 160 mg/day [82]. Furthermore, at least 50% of patients on therapeutic dosages suffer adverse effects [100] and only 20% of patients treated with high doses experience the benefits of the medication, with such benefits tending to disappear a few weeks after medication is halted [39, 41]. It is, therefore, unlikely that more than 20% of patients initially treated with tacrine will benefit from the treatment.

It is not known which AD patient subgroup could benefit from the treatment. Information on the long-term effects of tacrine (periods exceeding 7 months) and on its effects on quality of life, patient institutionalization and mortality and patient burden on caregivers, is inadequate. Tile cost-effectiveness of tacrine in AD is also unknown, although it would have to delay institutionalization by a minimum of 9 months for the treatment to be cost-effective [101].

In conclusion, tacrine shows a modest degree of efficacy among a small proportion of patients with mild to moderate AD and has adverse effects which limit its clinical usefulness. However tacrine should be used as a benchmark for the evaluation of other aminoacridines such as donepezil, galantamine and metrifonate as anti-dementia drugs. The ideal drug would have greater clinical efficacy than tacrine, a lower dosage frequency, greater safety and lower cost.

Acknowledgements

We thank Carlos Rodriguez Pascual and Teresa Olcoz Chiva for their helpful comments on an earlier draft of the paper and Michael D. Benedict for preparing the English-language version. This study was funded in part by grant no. 96/0477 from the Fondo de Investigacion Sanitaria (Health Research Fund).

Key points

* This systematic review of 49 clinical trials of tacrine in Alzheimer's disease showed that one-third of patients were withdrawn from treatment.

* Most withdrawals were because of cholinergic side effects or elevations in serum transaminase concentrations.

* Some patients with mild to moderate Alzheimer's disease show clinical benefit on tacrine but these patients cannot be identified before starting treatment.

References

[1.] Hofman A, Rocca W, Brayne C et al. The prevalence of dementia in Europe: a collaborative study of 1980-1990 findings. Int J Epidemiol 1991; 20: 734-48.

[2.] Jorm AF, Korten AE, Jacomb PA. Projected increases in the number of dementia cases for 29 developed countries: application of a new method for making projections. Acta Psychiat Scand 1988; 78: 493-500.

[3.] Whitehouse PT, Price DL, Struble RG, Clark AW, Coyle JT, DeLong MR. Alzheimer's disease and senile dementia: loss of neurons in the basal forebrain. Science 1982; 215: 1237-9.

[4.] Perry E, Tomlinson BE, Blessed G, Bergmann K, Gibson P, Perry R. Correlation of cholinergic abnormalities with senile plaques and mental test scores in senile dementia. Br Med J 1978; 2: 1457-9.

[5.] Davies P. The neurochemistry of Alzheimer's disease and senile dementia. Med Res Rev 1983; 3:221-36.

[6.] Greenmayre JT, Young AB. Excitatory aminoacids and Alzheimer's disease. Neurobiol Aging 1993; 3: 408-27.

[7.] Leake A, Ferrier IN. Alterations in neuropeptides in aging and disease. Pathophysiology and potential for clinical intervention. Drug Aging 1993; 3: 408-27.

[8.] Langlais PJ, Thal L, Hansen L, Galasko D, Alford M, Masliah E. Neurotransmitters in basal ganglia and cortex of Alzheimer's disease with and without Lewy bodies. Neurology 1993; 43: 1927-34.

[9.] Gottfries CG. Brain monoamines and their metabolites in dementia. Acta Neurol Scand 1990; 129: 8-11.

[10.] Bowen D. The cholinergic system and excitatory amino acids in Alzheimer's disease. Acta Neurol Scand 1990; 129: 15-20.

[11.] Bartus RT, Dean RL, Pontecorvo MJ, Flicker C. The cholinergic hypothesis: a historical overview, current perspectives and future directions. Ann NY Acad Sci 1985; 444: 332-58.

[12.] Bartus RT, Dean RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982; 217: 408-17.

[13.] Coyle JT, Price CL, DeLong MR. Alzheimer's disease: a disorder of cholinergic innervation. Science 1983; 219: 1184-90.

[14.] Jorm AF. Effects of cholinergic enhancement therapies oil memory dysfunction in Alzheimer's disease: a meta-analysis of the literature. Aust NZ J Psychiatry 1986; 20: 237-40.

[15.] Giacobini E. Cholinergic therapy of Alzheimer's disease. Ann Psychiatry 1994; 4:281-91.

[16.] Nordberg A, Lilja A, Lundquist H, Hartvig P, Amberla K, Viitanen M. Tacrine restores cholinergic nicotinic receptors and glucose metabolism in Alzheimer patients as visualized by emission positron tomography. Neurobiol Aging 1992; 13: 747-58.

[17.] Mesulam MM, Geula C, Moran MA. Anatomy of cholinesterase inhibition in Alzheimer's disease: effect of physostigmine and tetrahydroaminoacridine on plaques and tangles. Ann Neurol 1987; 22: 683-91.

[18.] Thornton J, Gershon S. The history of THA. In: Giacobini E, Becker R eds. Current Research in Alzheimer's Therapy. New York: Taylor & Francis, 1988: 267-78.

[19.] Rosenberg DR, Wright BA, Gershon S. Cognitive enhancing agents for the treatment of senile dementia of the Alzheimer type. Drugs Today 1989; 26: 449-71.

[20.] Lopez Arrieta JM, Qizilbash N. La medicina basada en pruebas: revisiones sistematicas. La Colaboracion Cochrane. Med Clin (Barc) 1996; 107: 581-5.

[21.] Schneider LS. Clinical pharmacology of aminoacridines in Alzheimer's disease. Neurology 1993; 43 (suppl. 4): S64-79.

[22.] Schneider LS. Tacrine development experience: early clinical trials and enrichment and parallel designs. Alzheimer Dis Assoc Disord 1994; 8 (suppl. 2): S12-21.

[23.] Roberts E Lazareno S. Cholinergic treatments for Alzheimer's disease. Biochem Soc Trans 1989; 17: 76-9.

[24.] Crismon ML. Tacrine: first drug approved for Alzheimer's disease. Ann Pharmacol 1994; 28: 744-51.

[25.] Cacabelos R. Tacrina: primera generacion de farmacos antidemencia. Med Clin (Barc) 1995; 105: 105-15.

[26.] Soares JC, Gershon S. THA--historical aspects, review of pharmacological properties and therapeutic effects. Dementia 1995; 6: 225-34.

[27.] Woo JK, Lantz MS. Alzheimer's disease: how to give and monitor tacrine therapy. Geriatrics 1995; 50: 50-3.

[28.] Boller F, Orgogozo JM. Tacrine. Alzheimer's disease and the cholinergic theory. A critical review and results of a new therapy. Neurologia 1995; 10: 194-9.

[29.] Davis KL, Powchik P. Tacrine. Lancet 1995; 345: 625-30.

[30.] Antman EM, Lau J, Kupelnick B, Mosteller F, Chalmers TC. A comparison of results of meta-analyses of randomized control trials and recommendations of clinical experts. Treatment for myocardial infarction, JAMA 1992; 268: 240-8.

[31.] Cooper H, Hedges LV. Potentials and limitations of research synthesis. In: Cooper H, Hedges LV eds. The Handbook of Research Synthesis. New York: Russell Sage Foundation, 1994:521-9.

[32.] McKahn G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease. Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's disease. Neurology 1984; 34: 939-44.

[33.] American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 3rd ed, revised. Washington: American Psychiatric Association, 1987.

[34.] Dickersin K, Scherer R, Lefebvre C. Identifying relevant studies for systematic reviews of the effects of health care. Br Med J 1994; 309: 862-5.

[35.] The Standards of Reporting Trials Group. A proposal for structured reporting of randomized controlled trials. JAMA 1994; 272: 1926-31.

[36.] Summers WK, Viesselman JO, Marsh GM, Candelora K. Use of THA in treatment of Alzheimer-like dementia: pilot study in twelve patients. Biol Psychiatry 1981; 16: 145-53.

[37.] Kaye WH, Sitaram N, Weingartner H, Ebert MH, Smallberg S, Gillin J. Modest facilitation of memory in dementia with combined lecithin and anticholinerestase treatment. Biol Psychiatry 1982; 17: 275-80.

[38.] Dal-Bianco P, Maly J, Deecke L. THA-therapie bei patienten mit seniler demenz vom Alzheimertyp. Wien Klin Wochenschr 1988; 100: 415-6.

[39.] Gauthier S, Masson H, Gauthier L et al. Tetrahydroaminoacridine and lecithin in Alzheimer's disease. In: Giacobini E, Becker Reds. Current Research in Alzheimer Therapy. London: Taylor & Francis, 1988: 237-45.

[40.] Nyback H, Nyman H, Ohman G, Nordgren I, Lindstrom B. Preliminary experiences and results with THA for the amelioration of symptoms of Alzheimer's disease. In: Giacobini E, Becker Reds. Current Research in Alzheimer Therapy. London: Taylor & Francis, 1988:231-6.

[41.] Gauthier S, Bouchard R, Bacher Y et al. Progress report on the Canadian trial of tetrahydroaminoacridine with lecithin in Alzheimer's disease. Can J Neurol Sci 1989; 16: 543-6.

[42.] Forsyth DR, Surmon DJ, Morgan RA, Wilcock GK. Clinical experience with and side-effects of tacrine hydrochloride in Alzheimer's disease: a pilot study. Age Ageing 1989; 18: 223-9.

[43.] Alhainen K, Sirvio J, Helkala EL, Reinikainen K, Riekkinen P. Somatostatin and cognitive functions in Alzheimer's disease--the relationship of cerebrospinal fluid somatostatin increase with clinical response to tetrahydroaminoacridine. Neuro Sci Lett 1991; 130: 46-8.

[44.] Alhainen K, Partanen J, Reinikainen K et al. Discrimination of tetrahydroaminoacridine responders by a single dose pharmaco-EEG in patients with Alzheimer's disease. Neuro Sci 1991; 127: 113-6.

[45.] Alhainen K, Riekkinen P, Helkala EL et al. The effect of THA on cognitive functions and spectral power EEG in Alzheimer's disease: preliminary results of an open study. In: Iqbal K, McLachlan DRC, Winblad B, Wisniewski HM eds. Alzheimer's Disease: basic mechanisms, diagnosis and therapeutic strategies. Chichester: Wiley, 1991.

[46.] Perryman KM, Fitten LJ. Quantitative EEG during a double-blind trial of THA and lecithin in patients with Alzheimer's disease. J Geriatr Psychiatry Neurol 1991; 4: 127-33.

[47.] van Gool WA, Waardenburg J, Meyjes FEP, Weinstein HC, de Wilde A. The effect of tetrahydroaminoacridine (THA) on P300 in Alzheimer's disease. Biol Psychiatry 1991; 30:953-7.

[48.] Cohen MB, Fitten LJ, Lake RR, Peryman KM, Graham LS, Sevrin R. SPECT brain imaging in Alzheimer's disease during treatment with oral tetrahydroaminoacridine and lecithin. Clin Nucl Med 1992; 17: 312-5.

[49.] O'Brien JT, Syed GMS, Levy R. The effects of tacrine on regional cerebral blood flow in Alzheimer's disease. Int J Geriatr Psychiatry 1992; 7: 835-8.

[50.] Nordberg A, Lilja A, Lundqvist H et al. Tacrine restores cholinergic nicotinic receptors and glucose metabolism in Alzheimer's patients as visualized by positron emission tomography. Neurobiol Aging 1992; 13: 747-58.

[51.] Alhainen K, Helkala EL, Reinikainen K, Riekkinen P. The relationship of cerebrospinal fluid monoamine metabolites with clinical response to tetrahydroaminoacridine in patients with Alzheimer's disease. J Neurol Transm Park Dis Dement Sect 1993; 5: 185-92.

[52.] Nordberg A. Effect of long-term treatment with tacrine (THA) in Alzheimer's disease as visualized by PET. Acta Neurol Scand 1993; 149 (suppl.): 62-5.

[53.] Amberla K, Nordberg A, Viitanen M, Winblad B. Long-term treatment with tacrine (THA) in Alzheimer's disease-evaluation neuropsychogical data. Acta Neurol Scand 1993: 149 (suppl.): 55-7.

[54.] Eagger SA, Richards M, Levy R. Long-term effects of tacrine in Alzheimer's disease: an open study: Int J Geriatr Psychiatry 1994; 9: 643-7.

[55.] Minthon L, Nilsson K, Edvinsson L, Wendt PE, Gustafson LTI. Long-term effects of tacrine on regional cerebral blood flow changes in Alzheimer's disease. Dementia 1995; 6: 245-51.

[56.] Riekkinen P Jr, Kuikka J, Soininen H, Helkala EL, Hallikainen M, Rielddnen P. Tetrahydroaminoacridine modulates technetium-99m labelled ethylene dicysteinate retention in Alzheimer's disease measured with single photon emission computed tomography imaging. Neurosci Lett 1995; 195: 53-6.

[57.] Knopman D, Schneider L, Davis K et al. Long-term tacrine (Cognex) treatment: effects on nursing home placement and mortality. Neuroloy 1996; 47: 166-77.

[58.] Summers WK, Majovski LV, Marsh GM, Tachiki K, Kling A. Oral tetrahydroaminoactidine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med 1986; 315: 1241-5.

[59.] Davies B, Andrews D, Stargatt R, Ames D, Tuckwell V, Davis S. Tetrahydroaminoactidine in Alzheimer's disease. Int J Geriatr Psychiatry 1990; 5: 317-21.

[60.] Davies B, Andrews D, Stargatt R, Ames D, Tuckwell V, Davis S. Tacrine in Alzheimer's disease. Lancet 1989; 2: 1634.

[61.] Fitten LJ, Perryman KM, Gross PL, Fine H, Cummins J, Marshall C. Treatment of Alzheimer's disease with short- and long-term oral THA and lecithin: a double-blind study. Am J Psychiatry 1990; 147: 239-42.

[62.] Gauthier S, Bouchard R, Lamontagne A et al. Tetrahydroaminoacridine-lecithin combination treatment in patients with intermediate-stage Alzheimer's disease. N Engl J Med 1990; 322: 1272-6.

[63.] Chatellier G, Lacomblez L, on behalf of Groupe d'Etude de la Tetrahydroaminoactidine. Tacrine (tetrahydroaminoacridine; THA) and lecithin in senile dementia of the Alzheimer type. Br Med J 1990; 300: 495-9.

[64.] Ahlin A, Nyback H, Junthe T, Ohman G, Nordgren I. Tetrahydroaminoacridine in Alzheimer's dementia: clinical and biochemical results of a double-blind crossover trial. Hum Psychopharmacol 1991; 6:109-18.

[65.] Ahlin A, Nyback H, Junthe T, Ohman G, Nordgren I. Tetrahydroaminoacridine (THA) in Alzheimer's dementia: clinical and biochemical results of a double-blind crossover trial. Neurobiol Aging 1990; 11: 344.

[66.] Ahlin A, Adem A, Junthe T, Ohman G, Nyback H. Pharmacokinetics of tetrahydroaminoacridine: relations to clinical and biochemical effects of Alzheimer's patients. Int Clin Psycopharmacol 1992; 7: 29-36.

[67.] Molloy W, Guyatt GH, Wilson DB, Duke R, Rees L, Singer J. Effect of tetrahydroaminoacridine on cognition, function and behaviour in Alzheimer's disease. Can Med Assoc J 1991; 144: 29-34.

[68.] Weinstein HC, Teunisse S, van Gool WA. Tetrahydroaminoacridine and lecithin in the treatment of Alzheimer's disease. J Neurol 1991; 238: 34-8.

[69.] Eagger SA, Levy R, Sahakian BJ. Tacrine in Alzheimer's disease. Lancet 1991; 337: 989-92.

[70.] Eagger SA, Sahakian BJ. Tacrine in Alzheimer's disease. Acta Neurol Scand 1992; 139 (suppl.): 75-80.

[71.] Eagger SA, Morant N. Levy R, Sahakian B. Tacrine in Alzheimer's disease. Time course of changes in cognitive function and practice effects. J Psychiatr 1992; 160: 36-40.

[72.] O'Brien JT, Eagger S, Levy R. Effects of tetrahydroaminoacridine on liver function in patients with Alzheimer's disease. Age Ageing 1991; 20: 129-31.

[73.] Sahakian B, Coull JT. Nicotine and tetrahydroaminoacridine: evidence for improved attention in patients with dementia of the Alzheimer type. Drug Dev Res 1994; 31: 80-8.

[74.] Forette F. The French THA multicenter trial: purpose, design and discussion of the metodology used (abstract). 5th Congress of the International Psychogeriatric Association, Rome, 1991.

[75.] Forette F, Hoover T, Gracon S et al. A double-blind, placebo-controlled, enriched population study of tacrine in patients with Alzheimer's disease. Eur J Neurol 1995; 2: 229-38.

[76.] Davis KL, Thal LJ, Gamzu ER et al. A double-blind, placebo-controlled multicenter study of tacrine for Alzheimer's disease. N Engl J Med 1992; 327: 1253-9.

[77.] Gamzu ER, Thal LJ, Davis KL. Therapeutic trials using tacrine and other cholinesterase inhibitors. Adv Neurol 1990; 51: 241-5.

[78.] Farlow M, Gracon S, Hershey LA, Lewis KW, Sadowsky CH, Dolan-Urero J, for the Tacrine Study Group. A controlled trial of tacrine in Alzheimer's disease. JAMA 1992; 268: 2523-9.

[79.] Minthon L, Gustafson L, Dalfet G et al. Oral tetrahydroaminoacridine treatment of Alzheimer's disease evaluated clinically and by regional cerebral blood flow and EEG. Dementia 1993; 4: 32-42.

[80.] Wilcock GK, Surmon DJ, Scott M et al. An evaluation of the efficacy and safety of tetrahydroaminoacridine (THA) without lecithin in the treatment of Alzheimer's disease. Age Ageing 1993; 22: 316-24.

[81.] Maltby N, Broe GA, Creasy H, Jorm AE Christensen H, Brooks WS. Efficacy of tacrine and lecithin in mild to moderate Alzheimer's disease: double blind trial. Br Med J 1994; 308: 879-83.

[82.] Knapp MJ, Knopman DS, Solomon PR, Pendlebury WW, Davis CS, Gracon SI, for the Tacrine Study Group. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer's disease. JAMA 1994; 271: 985-91.

[83.] Wood PC, Castleden CM. A double-blind, placebo controlled, multicentre study of tacrine lot Alzheimer's disease. Int J Geriatr Psychiatry 1994; 9: 649-54.

[84.] Foster NL, Petersen RC, Gracon SI, Lewis K, The Tacrine 970-6 Study Group. An enriched-population, double-blind, placebo controlled, crossover study of tacrine and lecithin in Alzheimer's disease. Dementia 1996; 7: 260-66.

[85.] Folstein ME Folstein SE, McHugh PR. Mini-Mental State. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975; 12: 189-98.

[86.] Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer's disease. Am J Psychiatr 1984; 141: 1356-64.

[87.] Lawton MP, Brody EM. Assessment of older people: self-maintaining and instrumental activities of daily living. Gerontologist 1969; 9: 179-86.

[88.] Lawton ME Assessment of the competence of older people. In: Kent D, Kastenbaum R, Sherwood Seds. Research, Planning and Action for the Elderly. New York: Behavioral Publications, 1972:122-43.

[89.] Clinical global assessment. In: Guy W ed. ECDEU Assessment Manual for Psychopharmacology. Rockville, MD: US Dept of Health Education and Welfare, National Institute of Mental Health, 1976: 218-22.

[90.] Frank A, Barreiro P, Diez-Tejedor E. Clinical trials in Alzheimer's disease: evaluation of the therapeutic response. Rev Neurol 1995: 23:111-7.

[91.] Schulz KE Chalmers I, Hayes RJ, Altman DG. Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA 1995; 273: 408-12.

[92.] Schulz KF, Grimes DA, Altman DG, Hayes RJ. Blinding and exclusions after allocation in randomised controlled trials: survey of published parallel group trials in obstetrics and gynaecology. Br Med J 1996; 312: 742-4.

[93.] Piccini C, Bracco L, Falcini M, Pracucci G, Amaducci LTI. Natural history of Alzheimer's disease: prognostic value of plateaux. J Neurol Sci 1995; 131: 177-82.

[94.] Mots RC, Rosen WG, Davis KL. The Alzheimer's disease assessment scale. An instrument for assessing treatment efficacy. Psychopharmacol Bull 1983; 19: 448-50.

[95.] Salmon DP, Thai LJ, Butters N, Heindel WC. Longitudinal evaluation of dementia of the Alzheimer type: A comparison of three standardized mental status examinations. Neurology 1990; 40:1225-30.

[96.] Kramer-Ginsberg E, Mohs RC, Aryan M. Clinical predictors of course for Alzheimer patients in a longitudinal study: a preliminary report. Psychopharmacol Bull 1988; 24: 458-62.

[97.] Department of Health and Human Services Public Health Service. Peripheral and Central Nervous System Drugs Advisory Committee meeting, July 7. Rockville, MD: Food and Drug Administration, 1989; 227.

[98.] Kokmen E, Chandra V, Schoenberg BS. Trends in incidence of dementing illness in Rochester, Minnesota, in three quinquennial periods, 1960-1974. Neurology 1988; 38: 975-80.

[99.] Rorsman B, Hagnell O, Lanke J. Prevalence of age psychosis and mortality among age psychotics in the Lundby study. Neuropsychobiology 1985; 13: 167-72.

[100.] Watkins PB, Zimmerman HJ, Knapp MJ, Gracon SI, Lewis KW. Hepatotoxic effects of tacrine administration in patients with Alzheimer's disease. JAMA 1994; 271: 992-8.

[101.] Knopman D. Tacrine for Alzheimer's disease. Cost and benefits. PharmacoEconomics 1995; 7: 275-9.

Received 5 August 1997

COPYRIGHT 1998 Oxford University Press
COPYRIGHT 2000 Gale Group

Return to Tacrine
Home Contact Resources Exchange Links ebay