Skip to main content
Erschienen in: BMC Geriatrics 1/2015

Open Access 01.12.2015 | Research article

Anticholinergic burden quantified by anticholinergic risk scales and adverse outcomes in older people: a systematic review

verfasst von: Mohammed Saji Salahudeen, Stephen B Duffull, Prasad S Nishtala

Erschienen in: BMC Geriatrics | Ausgabe 1/2015

Abstract

Background

The cumulative effect of taking multiple medicines with anticholinergic properties termed as anticholinergic burden can adversely impact cognition, physical function and increase the risk of mortality. Expert opinion derived risk scales are routinely used in research and clinical practice to quantify anticholinergic burden. These scales rank the anticholinergic activity of medicines into four categories, ranging from no anticholinergic activity (= 0) to definite/high anticholinergic activity (= 3). The aim of this systematic review was to compare anticholinergic burden quantified by the anticholinergic risk scales and evaluate associations with adverse outcomes in older people.

Methods

We conducted a literature search in Ovid MEDLINE, EMBASE and PsycINFO from 1984-2014 to identify expert opinion derived anticholinergic risk scales. In addition to this, a citation analysis was performed in Web of Science and Google Scholar to track prospective citing of references of selected articles for assessment of individual scales for adverse anticholinergic outcomes. The primary outcomes of interest were functional and cognitive outcomes associated with anticholinergic burden in older people. The critical appraisals of the included studies were performed by two independent reviewers and the data were extracted onto standardised forms.

Results

The primary electronic literature search identified a total of 1250 records in the 3 different databases. On the basis of full-text analysis, we identified 7 expert-based anticholinergic rating scales that met the inclusion criteria. The rating of anticholinergic activity for medicines among these rating scales was inconsistent. For example, quetiapine was rated as having high anticholinergic activity in one scale (n = 1), moderate in another scale (n = 1) and low in two other scales (n = 2). Citation analysis of the individual scales showed that the Anticholinergic Cognitive Burden (ACB) scale was the most frequently validated expert based anticholinergic scale for adverse outcomes (N = 13).

Conclusions

In conclusion, there is not one standardised tool for measuring anticholinergic burden. Cohort studies have shown that higher anticholinergic burden is associated with negative brain effects, poorer cognitive and functional outcomes.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12877-015-0029-9) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MSS and PSN designed the search strategy, extracted data, analysed data and drafted the manuscript. SBD assisted with analyses and drafting the manuscript. All authors contributed to data analyses and interpretation, critically commented on, and approved the final manuscript.
Abkürzungen
ADS
Anticholinergic drug scale
ABC
Anticholinergic burden classification
CrAS
Clinician-rated anticholinergic score
ARS
Anticholinergic risk scale
ACB
Anticholinergic cognitive burden scale
AAS
Anticholinergic activity scale
ACL
Anticholinergic loading scale
SAA
Serum anticholinergic activity

Background

Medicines with anticholinergic properties are frequently prescribed in the older population for various medical conditions [1]. The cumulative effect of taking one or more medicines with anticholinergic properties is referred to as anticholinergic burden [2]. The majority of medicines commonly prescribed to older people are not routinely recognised as having anticholinergic activity and empirically physicians prescribe these medicines based on their anticipated therapeutic benefits overlooking the risk of cumulative anticholinergic burden [3].
A number of studies have reported on the adverse effects associated with higher anticholinergic burden. Studies have found that anticholinergic medicines may adversely affect cognitive and physical function [4-13] and anticholinergic burden is a strong predictor of cognitive and physical impairments in older people living in both community and residential care [4-7,12,14]. A retrospective study conducted in Finland found that the use of medicines with anticholinergic properties is a strong independent predictor of mortality in older people [15,16]. More recently, several studies in the older population have also reported an association between anticholinergic exposure and mortality with an increased risk of hospitalisations [1,6,17,18].
Expert rating scales are routinely used in research and clinical practice to quantify anticholinergic burden. Expert opinion derived rating scales generally rank the anticholinergic activity of drugs into four categories, ranging from no known anticholinergic activity (= 0) to definite/high anticholinergic activity (= 3) [3,5,9,19,20]. The aim of this systematic review was to compare anticholinergic burden quantified by the anticholinergic risk scales and evaluate associations with adverse outcomes in older people.

Methods

Data sources and search strategy

A literature search in Ovid MEDLINE, EMBASE and PsycINFO covering the period 1984 - September 2014 was completed to identify anticholinergic risk scales using the keywords; (anticholinergic*.mp), AND (cogniti#.mp). The search was then limited to English language AND humans AND ("all aged (65 and over)" OR "aged (80 and over)"). The MEDLINE search strategy is presented in Additional file 1.
Following the primary literature search to identify the relevant studies, we carried out a citation analysis of individual rating scales to identify potential studies validating the association between anticholinergic burden quantified by the anticholinergic risk scales and adverse outcomes. The citation analysis was performed with the aid of Web of Science and Google Scholar to track prospective citing of references of selected articles.
Potentially relevant articles were selected according to the pre-defined inclusion and exclusion criteria. A flowchart of search strategy and citation search is depicted in Figure 1.

Study screening and selection

Selecting the title and abstract of the publication, studies retrieved were screened by two independent reviewers for its eligibility for inclusion in the review process (M.S.S. and P.S.N.). The eligible studies were subject to a thorough full text analysis for relevance and pre-defined inclusion criteria. Studies that met the following criteria were included in the final review.
a)
The quantification tool was based on expert opinion.
 
b)
Studies that reported the use of expert opinion quantification scale/tool to measure anticholinergic burden.
 
c)
Studies that include participants of either sex, of mean age 65 years or older and living in primary care or nursing homes or hospitals.
 
We excluded articles in languages other than English, as well as case reports, commentaries, letters and editorials from the primary search and citation analysis. Anticholinergic rating scales based predominantly on serum anticholinergic activity (SAA) were also excluded from the review.
The primary aim of this review was to compare anticholinergic burden quantified by the anticholinergic risk scales and evaluate associations with physical, cognitive outcomes in older people.
Ethical approval was noted for all published papers included in the review.

Data extraction and synthesis

Two reviewers (M.S.S. and P.S.N.) extracted data onto standardised format based on study population, study design, use of appropriate rating scales to quantify anticholinergic burden and outcome measures. The primary outcomes of interest were functional and cognitive outcomes associated with anticholinergic burden quantified by the expert opinion derived anticholinergic rating scales.
A citation analysis was performed to identify and compare the clinical utility of individual anticholinergic rating scales to quantify anticholinergic burden and to evaluate its association with adverse outcomes (cognitive, functional, mortality) in older people. Studies that used the rating scales for assessing the adverse outcomes in older people are reported in this review.
The quality of the included studies were critically appraised by two authors (M.S.S. and P.S.N.) using the United States Preventive Services Task Force (USPSTF) criteria [21]. The criteria to assess the internal validity of studies included: initial assembly and maintenance of comparable groups, measurements, clear definition of interventions, outcomes assessed and analysis. Critical appraisal scores derived from the USPSTF criteria were rated as poor, fair or good. Any differences between review authors concerning eligibility were reviewed by the third author (S.B.D.) and decisions were made by consensus.

Results

The primary search using three databases identified a total of 7 scales as being relevant to this systematic review. A qualitative description of the included studies is shown in Table 1.
Table 1
Overview of included anticholinergic rating scales
Expert opinion based rating scales
Description
Number of anticholinergic activity medicines listed (N)
Carnahan USA, 2006 [9]
ADS is a four-point (0-3) scale that ranks anticholinergic drugs based on expert opinion
117
Ancelin France, 2006 [25]
ABC is a four-point scale (0-3) based on SAA and expert opinion
27
Han USA, 2008 [22]
CrAS is a four-point scale (0-3) based on pre-existing published anticholinergic scales and expert opinion
60
Rudolph USA, 2008 [19]
ARS is a four-point scale (0-3) based on extensive literature review and expert opinion
49
Boustani USA, 2008 [24]
ACB is a four-point (0-3) scale developed based on published data and expert opinion
88
Ehrt Norway, 2010 [26]
AAS is a five-point scale (0-4) based on existing evidence (Chew 2008 [38]) and expert opinion
99
Sittironnarit Australia, 2011 [23]
ACL is a four-point (0-3) scale based on pre-existing published anticholinergic scales and expert opinion
49
ADS = Anticholinergic Drug Scale; ABC = Anticholinergic Burden Classification; CrAS = Clinician-rated Anticholinergic Score; ARS = Anticholinergic Risk Scale; ACB = Anticholinergic Cognitive Burden Scale; AAS = Anticholinergic Activity Scale; ACL = Anticholinergic Loading Scale; SAA = Serum Anticholinergic Activity.
Points in rating scale represents, 0 = no anticholinergic activity, 1 = mild anticholinergic activity, 2 = moderate anticholinergic activity, and 3 = severe anticholinergic activity.
The primary electronic literature search identified a total of 1250 articles from 3 different databases such as Ovid MEDLINE, EMBASE, and PsycINFO. EndNote was used to eliminate duplicates and we considered 932 articles for screening. Out of 932 screened articles based on title and abstract, only 21 were eligible for full-text analysis. From the eligible 21 studies, 14 were excluded on full text analysis according to the set inclusion and exclusion criteria. Hence, in total, 7 studies were included in this review that considered expert opinion/s in the development of the anticholinergic rating scales [9,19,22-26]. Figure 1 depicts a flow-diagram of the identification, screening, eligibility and exclusion process.
The 7 scales ranked anticholinergic activity of medicines into four categories, ranging from no anticholinergic activity (= 0) to definite/high anticholinergic activity (= 3). The anticholinergic medicines described in the 7 rating scales were collated into a composite reference rating scale. The composite reference scale shows a total of 195 medicines derived from the 7 published scales that ranked anticholinergic activity from high to low as shown in Table 2.
Table 2
A composite rating scale to categorise anticholinergic activity medicines (N = 195)
High
Moderate
Low
Aceprometazine [25] (n = 1)
  
Acepromazine [25] (n = 1)
  
 
Alimemazine (trimeprazine) [25] (n = 1)
Alimemazine [24] (n = 1)
Alprazolam [25] (n = 1)
 
Alprazolam [9,22-24] (n = 4)
 
Alverine [25] (n = 1)
Alverine [24] (n = 1)
 
Amantadine [19,24] (n = 2)
Amantadine [9,22] (n = 2)
Amitriptyline [9,19,22-26] (n = 7)
  
Amoxapine [24,25] (n = 2)
  
  
Ampicillin [9] (n = 1)
  
Aripiprazole [24] (n = 1)
  
Asenapine [24] (n = 1)
  
Atenolol [22,24] (n = 2)
Atropine [9,19,22-24] (n = 5)
  
  
Azathioprine [9] (n = 1)
 
Baclofen [19,22] (n = 2)
 
Belladonna [22,25] (n = 2)
Belladonna [24] (n = 1)
 
  
Benazepril [22] (n = 1)
Benzatropine/benztropine [9,19,24,26] (n = 4)
  
  
Betaxolol [22] (n = 1)
  
Bisacodyl [23] (n = 1)
  
Bromocriptine [9] (n = 1)
Brompheniramine [9,24] (n = 2)
  
  
Bupropion [22,24] (n = 2)
  
Captopril [9,24] (n = 2)
 
Carbamazepine [9,24] (n = 2)
Carbamazepine [22] (n = 1)
  
carbidopa [19,22,23] (n = 3)
Carbinoxamine [9,24] (n = 2)
  
Carisoprodol [19] (n = 1)
  
  
Cefamandole [9] (n = 1)
  
Cefoxitin [9] (n = 1)
  
Celecoxib [23] (n = 1)
  
Cephalothin [9] (n = 1)
 
Cetirizine [19,22,23] (n = 3)
Cetirizine [24] (n = 1)
  
Chlordiazepoxide [9,22] (n = 2)
Chlorphenamine/chlorpheniramine [9,19,22-25] (n = 6)
  
Chlorpromazine [9,19,22,24] (n = 4)
  
  
Chlorthalidone/chlortalidone [9,24] (n = 2)
 
Cimetidine [9,19] (n = 2)
Cimetidine [24] (n = 1)
  
Citalopram [23,26] (n = 2)
Clemastine [9,24] (n = 2)
  
  
Clidinium [24] (n = 1)
  
Clindamycin [9] (n = 1)
Clomipramine [9,24,25] (n = 3)
  
  
Clonazepam [9,23] (n = 2)
Clorazepate [25] (n = 1)
 
Clorazepate [9,24] (n = 2)
Clozapine [9,24,26] (n = 3)
Clozapine [19] (n = 1)
 
 
Codeine [25] (n = 1)
Codeine [9,22-24] (n = 4)
Colchicine [25] (n = 1)
 
Colchicine [24] (n = 1)
  
Cortisone [9] (n = 1)
 
Cyclobenzaprine [9,19,24] (n = 3)
Cyclobenzaprine [22] (n = 1)
  
Cycloserine [9] (n = 1)
  
Cyclosporine [9] (n = 1)
Cyproheptadine [19,23] (n = 2)
Cyproheptadine [9,24] (n = 2)
 
Darifenacin [9,24] (n = 2)
  
Desipramine [9,24] (n = 2)
Desipramine [19,22] (n = 2)
 
  
Desloratadine [24] (n = 1)
  
Dexamethasone [9] (n = 1)
Dexchlorpheniramine [23,25] (n = 2)
  
  
Dextromethorphan [22] (n = 1)
  
Diazepam [9,22-24,26] (n = 5)
Dicyclomine [9,19,24] (n = 3)
  
  
Digitoxin [9] (n = 1)
Digoxin [25] (n = 1)
 
Digoxin [9,23,24,26] (n = 4)
  
Diltiazem [9] (n = 1)
Dimenhydrinate [9,24] (n = 2)
  
Diphenhydramine [9,19,22,24] (n = 4)
  
  
Dipyridamole [9,24] (n = 2)
 
Disopyramide [9] (n = 1)
Disopyramide [24] (n = 1)
  
Divalproex sodium [9] (n = 1)
  
Domperidone [23] (n = 1)
 
Dothiepin/dosulepin [23] (n = 1)
 
Doxepin [9,22-24,26] (n = 5)
  
Doxylamine [24] (n = 1)
  
Emepronium [26] (n = 1)
  
  
Entacapone [19] (n = 1)
  
Escitalopram [23] (n = 1)
  
Estazolam [9] (n = 1)
  
Famotidine [9] (n = 1)
  
Fentanyl [9,24] (n = 2)
Fesoterodine [24] (n = 1)
  
 
Fexofenadine [22,23] (n = 2)
 
Flavoxate [9,24] (n = 2)
  
  
Fluoxetine [9,22,23,26] (n = 4)
Fluphenazine [19,23] (n = 2)
 
Fluphenazine [9] (n = 1)
  
Flurazepam [9] (n = 1)
  
Fluticasone-salmeterol [9] (n = 1)
  
Fluvoxamine [9,23,24,26] (n = 4)
Furosemide [25] (n = 1)
 
Furosemide [9,24] (n = 2)
  
Gentamicin [9] (n = 1)
  
Guaifenesin [22] (n = 1)
 
Haloperidol [23] (n = 1)
Haloperidol [19,24] (n = 2)
Homatropine [22] (n = 1)
  
  
Hydralazine [9,24] (n = 2)
 
Hydrocodone [22] (n = 1)
 
  
Hydrocortisone [9,24] (n = 2)
Hydroxyzine [9,19,24,25] (n = 4)
  
Hyoscyamine [9,19,24] (n = 3)
  
  
Iloperidone [24] (n = 1)
Imipramine [9,19,22-25] (n = 6)
  
Ipratropium [26] (n = 1)
  
  
Isosorbide [9,24] (n = 2)
  
Ketotifen [9] (n = 1)
  
Ketorolac [22] (n = 1)
  
Ketotifen [9] (n = 1)
  
Levocetirizine [24] (n = 1)
Levomepromazine [9,24,25] (n = 3)
  
  
Lithium [23] (n = 1)
 
Loperamide [19] (n = 1)
Loperamide [9,22-24] (n = 4)
 
Loratadine [19] (n = 1)
Loratadine [22-24] (n = 3)
  
Lorazepam [9] (n = 1)
 
Loxapine [9,24] (n = 2)
 
  
Lumiracoxib [23] (n = 1)
Maprotiline [25] (n = 1)
  
Meclizine/meclizine [9,19,24] (n = 3)
  
 
Meperidine [9,24] (n = 2)
 
  
Metformin [23] (n = 1)
 
Methadone [22] (n = 1)
 
  
Methocarbamol [19,22] (n = 2)
  
Methotrexate [23] (n = 1)
 
Methotrimeprazine [9,24] (n = 2)
 
  
Methylprednisolone [9] (n = 1)
  
Metoclopramide [19,23] (n = 2)
  
Metoprolol [22,24] (n = 2)
  
Midazolam [9] (n = 1)
  
Mirtazapine [19] (n = 1)
 
Molindone [9,24] (n = 2)
 
  
Morphine [9,22,24] (n = 3)
  
Naratriptan [23] (n = 1)
  
Nefazodone [22] (n = 1)
 
Nefopam [24] (n = 1)
 
  
Nifedipine [9,24] (n = 2)
  
Nizatidine [9] (n = 1)
Nortriptyline [9,22,24] (n = 3)
Nortriptyline [19,26] (n = 2)
 
Olanzapine [24] (n = 1)
Olanzapine [19,26] (n = 2)
Olanzapine [9,22] (n = 2)
Opipramol [25] (n = 1)
  
Orphenadrine [9,24-26] (n = 4)
  
  
Oxazepam [9,23] (n = 2)
 
Oxcarbazepine [9,24] (n = 2)
 
Oxybutynin [9,19,24-26] (n = 5)
Oxybutynin [23] (n = 1)
 
  
Oxycodone [9,22,23] (n = 3)
  
Paliperidone [24] (n = 1)
  
Pancuronium [9] (n = 1)
Paroxetine [24] (n = 1)
Paroxetine [22,23,26] (n = 3)
Paroxetine [9,19] (n = 2)
Perphenazine [19,24] (n = 2)
Perphenazine [22] (n = 1)
Perphenazine [9] (n = 1)
  
Phenelzine [9] (n = 1)
  
Phenobarbital [22] (n = 1)
 
Pimozide [9,24] (n = 2)
 
  
Piperacillin [9] (n = 1)
  
Pramipexole [19] (n = 1)
  
Prednisolone [9] (n = 1)
  
Prednisone [9,24] (n = 2)
 
Prochlorperazine [19,22,23] (n = 3)
Prochlorperazine [9] (n = 1)
Procyclidine [9] (n = 1)
  
 
Promazine [26] (n = 1)
 
Promethazine [9,19,24] (n = 3)
  
Propantheline [9,24] (n = 2)
Propantheline [22] (n = 1)
 
Propiverine [24] (n = 1)
  
 
Propoxyphene [22] (n= 1)
 
Protriptyline [9,23] (n = 2)
  
 
Pseudoephedrine [19,23] (n = 2)
 
Pyrilamine [9] (n = 1)
  
Quetiapine [24] (n = 1)
Quetiapine [22] (n = 1)
Quetiapine [19,26] (n = 2)
  
Quinidine [24] (n = 1)
 
Ranitidine [9,22] (n = 2)
Ranitidine [19,23,24,26] (n = 4)
Reglan [22] (n = 1)
  
  
Risperidone [19,22-24] (n = 4)
  
Robitussin [22] (n = 1)
Scopolamine(hyoscine) [9,22,24] (n = 3)
  
  
Selegiline [19] (n = 1)
  
Sertraline [9,22] (n = 2)
Solifenacin [24] (n = 1)
  
  
Sumatriptan [23] (n = 1)
  
Temazepam [9,19,23] (n = 3)
 
Theophylline [23,25] (n = 2)
Theophylline [9,24,26] (n = 3)
Thioridazine [9,19,22,24,26] (n = 5)
  
Thiothixene [19] (n = 1)
 
Thiothixene [9] (n = 1)
Tizanidine [19] (n = 1)
  
Tolterodine [9,22-25] (n = 5)
Tolterodine [19] (n = 1)
 
 
Tramadol [22,23] (n = 2)
Tramadol [9] (n = 1)
  
Trandolapril [22] (n = 1)
  
Trazodone [19,22,24] (n = 3)
  
Triamcinolone [9]
(n = 1)
  
Triamterene [9,24] (n = 2)
  
Triazolam [9,22] (n = 2)
Trifluoperazine [19,24] (n = 2)
 
Trifluoperazine [9] (n = 1)
Trihexyphenidyl [9,22,24-26] (n = 5)
  
Trimipramine [9,24-26] (n = 4)
  
Tropatepine [25] (n = 1)
  
Trospium [24] (n = 1)
  
  
Valproic acid [9] (n = 1)
  
Vancomycin [9] (n = 1)
  
Venlafaxine [22-24] (n = 3)
  
Warfarin [9] (n = 1)
  
Ziprasidone [19] (n = 1)
  
Zolmitriptan [23] (n = 1)
Medicines in italics denote inconsistent validation.
The Anticholinergic Drug Scale (ADS) developed by Carnahan et al. [9] based on expert consensus ranks medicines with anticholinergic properties in an ordinal fashion from 0 to 3, with 0 indicating no known anticholinergic activity and 3 indicating definite/high anticholinergic activity. This scale was initially referred to as the Clinician-rated Anticholinergic Scale (CrAS) modified version. An expert panel of geriatric psychiatrists identified and reviewed 340 medicines with known anticholinergic activity and assigned a score from 0 to 3 according to their clinical experience and the pharmacologic mechanism of each medicine. The ADS scale contains 117 medicines with known anticholinergic activity. The ADS scale has shown to be of utility in various clinical settings such as community, nursing homes, outpatient clinics, and hospitals. The adverse outcomes studied in these settings were mainly, cognitive, functional, risk of hospitalisation, and mortality.
The Anticholinergic Risk Scale (ARS) score was developed based on a ranking system developed by Rudolph et al. [19]. A literature review of 500 medicines known to possess anticholinergic activity was conducted by a group of geriatricians and pharmacists within the Veterans Affairs Boston Healthcare System. The authors considered the affinity for the muscarinic receptor, experimental reporting of anticholinergic activity, and literature review on anticholinergic adverse effects. This information was used to rank medicines for anticholinergic activity on a scale of 0 to 3, with 0 indicating no known anticholinergic activity and 3 indicating definite/high anticholinergic activity. A total of 49 medicines with known anticholinergic activity were reported in the ARS scale. The clinical outcomes validated using the ARS scale were cognitive, functional, quality of life, length of hospital stay, and mortality. The ARS was validated in a veteran’s population derived from a single medical centre limiting its external validity. Higher ARS scores in veteran and primary care patients were shown to be associated with anticholinergic adverse events [19].
Anticholinergic Cognitive Burden Scale (ACB) developed by Boustani et al. [24] is based on a systematic literature review of medicines with known anticholinergic activity. The ACB scale included medicines that were likely to have a negative impact on cognition [27,28]. A multi-disciplinary panel assessed individual drugs to have none, possible, or definite anticholinergic properties with a score ranging from 0 to 3. ACB scale reported 88 medicines with known anticholinergic activity. Studies that employed the ACB scale have shown that higher anticholinergic burden predicts cognitive impairment in older people. In addition, the study conducted by Pasina L et al. showed that anticholinergic burden quantified by the ACB scale predicted impairment in physical functioning [27].
Using similar methodologies other anticholinergic risk scales have been developed in Australia [23], Norway [26], France [25] and U.S.A. [22]. The CrAS scale by Han et al. was validated in palliative care and veteran home settings for cognitive and functional outcomes. The Anticholinergic Activity Scale (AAS) by Ehrt et al., and Anticholinergic Loading Scale (ACL) by Sittironnarit et al. were validated for only cognitive outcomes.
Citation analysis of individual anticholinergic rating scales show anticholinergic burden scores associated with adverse outcomes in older people in various clinical settings. An overview of the studies is presented in Table 3. The 38 studies retrieved comprised of 2 RCTs, 12 cross-sectional studies and 24 cohort studies that validated the 6 anticholinergic rating scales.
Table 3
Summary of study characteristics and validation of anticholinergic rating scales and its association with adverse outcomes in older people
Rating scales
Validation
Study design
Study population/setting
Study duration
Adverse outcome(s) studied
Significant association
Critical appraisal
Carnahan USA, 2006 (ADS)
Cross-sectional [9]
Long-term care residents (mean age 86), N = 279
1 month
SAA
+
Good
 
RCT [39]
Nursing home residents (mean age 85), N = 64
11 months
Cognitive function
Good
 
Cross-sectional [40]
Nursing home residents (mean age 73), N = 87
1 year
Cognitive function (MMSE)
Good
Functional outcome (ADL)
 
Cross-sectional [41]
Community-dwelling (aged ≥75), N = 621
3 years
Adverse events
+
Fair
Cognitive function (MMSE, GDP)
+
Functional outcome (ADL, IADL)
+
 
Longitudinal cohort [42]
Outpatient clinics (mean age 71.9 ± 7.3), N = 102
1 year
Cognitive function
+
Fair
 
Prospective cohort [43]
Hospital inpatients with hip fracture (aged ≥65), N = 364
48 hours to 5 days
Cognitive function (delirium)
Fair
 
Cross-sectional [44]
Hospital inpatients (mean age 67.9 ± 10.5), N = 450
28-30 days
Cognitive function
Fair
 
Cross-sectional [45]
Hospitalised (mean age 84 ± 6), N = 71
1 year
Mortality
Fair
 
Retrospective cohort [46]
Australian veterans (median age 80), N = 36015
2 years
Risk of hospitalisation for confusion or dementia
+
Good
Han USA, 2008 (CrAS)
Prospective cohort [22]
Community-dwelling men (aged ≥65), N = 544
2 years
Cognitive function (Verbal recall test)
+
Good
Functional outcome (ADL)
+
RCT [47]
Palliative care (mean aged 71), N = 461
Mean survival was 8.9 weeks
Quality of life (McGill’s Quality of life index)
+
Fair
Functional outcome (Karnofsky performance scale)
+
Prospective cohort [48]
Veteran home demented residents (mean age 83.4), N = 53
12 weeks
Cognitive function (MMSE)
Fair
Functional outcome (BI)
Rudolph, USA 2008 (ARS)
Retrospective and prospective cohort (one each) [19]
Hospital and long-term care facilities (aged ≥65), N = 132 and N = 117
9 months
Central adverse effects (Confusion, dizziness, falls)
+
Good
10 months
Prospective cohort [15]
Hospital and long-term care (mean age 81.3), N = 1004
1 year
Mortality
Good
Prospective cohort [29]
Hospitalised patients (mean age 83.6 ± 6.6), N = 362
5 months
Physical function (BI)
Good
Mortality
LOS
Cohort study [49]
Hospitalised patients (mean age 83.6 ± 6.6), N = 362
5 months
Institutionalisation and comorbidities
+
Fair
Cohort study [50]
Hospital rehabilitation unit (mean age 79 ± 7), N = 117
9 months
Functional outcome (BI)
+
Fair
LOS
Cross-sectional [41]
Community-dwelling (aged ≥75), N = 621
3 years
Adverse events
+
Fair
Cognitive function (MMSE, GDP)
+
Functional outcome (ADL, IADL)
+
Cross-sectional prospective [27]
Hospital (aged ≥65), N = 1380
3 months
Cognitive function (SBT)
+
Good
Physical function (BI)
+
Longitudinal cohort [42]
Outpatient clinics (mean age 71.9 ± 7.3), N = 102
1 year
Cognitive function
+
Fair
Cross-sectional [45]
Hospitalised (mean age 84 ± 6), N = 71
1 year
Mortality
+
Good
Retrospective cohort [51]
National Health Insurance Research Database (aged ≥65), N = 54,888
1 year and 6 months
Emergency visit
+
Poor
Hospitalisation
+
Constipation
+
Delirium
+
Cardiac arrhythmia
+
Cognitive impairment
Retrospective cohort [46]
Australian veterans (median age 80), N = 36015
2 years
Risk of hospitalisation for confusion or dementia
+
Good
Boustani, USA 2008 (ACB)
Cross-sectional [52]
Nursing home patient with dementia (aged ≥66), N = 87
2 years and 2 months
Quality of life: Multiple engagement observations
Fair
Longitudinal cohort [32]
Community-dwelling (aged ≥70), N = 1652
6 years
Cognitive function
+
Good
Observational cohort [53]
Hospitalised patients with cognitive impairment, N = 147 (aged ≥65)
Duration as of hospital admission
Cognitive function (Delirium using CAM)
Fair
Part of longitudinal cohort [54]
Nursing & residential homes, day hospital and inpatients with AD (mean age 81 ± 7.4), N = 224
1 year and 6 months
Cognitive function (MMSE and SIB)
Fair
Longitudinal cohort [33]
Community-dwelling and institutionalised patients (aged ≥65), N = 1304
2 years
Cognitive function
+
Good
Mortality
+
Retrospective cohort [34]
Primary-care clinics (aged ≥65), N = 3690
1 year
Cognitive function (MCI)
+
Fair
Prospective study [55]
Community-dwelling women (aged ≥75), N = 1429
5 years
Functional outcome (IADL)
+
Good
Cognitive function (MMSE)
Longitudinal cohort [56]
Community-dwelling women (aged ≥75), N = 1484
10 years
Cognitive function (MCI)
+
Good
Dementia
+
Cross-sectional prospective [27]
Hospital (aged ≥65), N = 1380
3 months
Cognitive function (SBT)
+
Good
Physical function (BI)
+
Cohort study [57]
Community-dwelling without dementia (aged ≥65), N = 896
10 years
Cognitive function
+
Fair
Retrospective study [58]
Hospital patients (aged ≥90), N = 419
3 months
Mortality
Fair
LOS
Longitudinal cohort [42]
Outpatient clinics (mean age 71.9 ± 7.3), N = 102
1 year
Cognitive function
+
Fair
 
Cross-sectional [45]
Hospitalised (mean age 84 ± 6), N = 71
1 year
Mortality
Good
Ehrt, Norway 2010 (AAS)
Longitudinal cohort [26]
Community-based PD patients (mean age 74.7), N = 78
8 years
Cognitive function (MMSE)
+
Good
Sittironnarit Australia, 2011 (ACL)
Cross-sectional [23]
Subjects in 3 groups; healthy controls (N = 211), MCI (N = 768) and AD (N = 133) of mean age 70.0 ± 7.0, 75.7 ± 7.6, and 78.0 ± 8.6 years
1 year and 10 months
Psychomotor speed and executive function
+
Good
SAA = Serum Anticholinergic Activity; ACE = Addenbrooke's Cognitive Examination; TMT = Trail Making Test; MMSE = Mini-Mental State Examination; CAM = Confusion Assessment Method; DSST = Digit Symbol Substitution Test; ADL = Activity of Daily Living; AD = Alzheimer’s Disease; IADL = Instrumental Activities of Daily Living; RCT = Randomised Controlled Trial; SIB = Severe Impairment Battery; SBT = Short Blessed Test; BI = Barthel Index; MCI = Mild Cognitive Impairment; PD = Parkinson’s Disease; LOS = Length of Stay; GDP = Geriatric Depression Scale; ADS = Anticholinergic Drug Scale; CrAS = Clinician-rated Anticholinergic Score; ARS = Anticholinergic Risk Scale; ACB = Anticholinergic Cognitive Burden Scale; AAS = Anticholinergic Activity Scale; ACL = Anticholinergic Loading Scale.

Discussion

To our knowledge, this is the first systematic review that compare anticholinergic burden quantified by the anticholinergic risk scales and evaluated associations with adverse outcomes in older people.
The citation analysis of individual scales revealed that ACB scale by Boustani et al. [24] was the most frequently validated expert based anticholinergic scale on adverse outcomes (N = 13) followed by ARS [19] (N = 11], ADS by Carnahan et al. [9] (N = 9), CrAS scale by Han et al. [22] (N = 3) and 2 other scales [23,26]. The review found only two RCTs that showed an association with higher anticholinergic burden and adverse outcomes. The RCT that used the CrAS scale to quantify anticholinergic burden showed a positive association with functional outcome and quality of life and the RCT using the ADS scale reported a negative association with cognitive functioning. The adverse outcomes reported in the cohort studies included mainly cognitive and physical outcomes. The cognitive outcomes reported included mild-cognitive impairment, confusion, dizziness, falls, delirium, psychomotor speed and executive function. The functional outcomes reported were pertaining to activity of daily living, instrumental activity of daily living, quality of life, physical function, hospitalisation, length of hospital stay, and mortality. A detailed summary of validated studies for individual anticholinergic scales with critical appraisal is illustrated in Table 3.
Numerous studies have found an association between use of anticholinergic medicines and adverse outcomes related to physical function, cognition and falls in older people [2,4,29-31]. Pasina et al. compared anticholinergic burden derived from both ACB and ARS scales and found strong associations with impairment in cognitive and functional outcomes [27]. A study conducted by Rudolph et al. validated higher ARS scores were associated with increased risk of both peripheral and central anticholinergic adverse effects in older people [19]. Furthermore, Campbell et al. and Fox et al. conducted studies using ACB scale and found that the use of definite anticholinergics increased the risk of cognitive impairment among older people [32,33]. Overall, research has shown that use of medicines with anticholinergic activity among older people is associated with physical and cognitive decline [34,35].
The variability in quantification of anticholinergic burden between the 7 anticholinergic scales was not surprising considering that the drugs listed and anticholinergic activity ratings assigned varied considerably in the 7 scales. Expert consensus was derived from an interdisciplinary team that consisted of geriatricians, pharmacists, psychiatrists, general physicians, nurses and researchers who research aging. The subjective rating of the anticholinergic activity relied heavily on the panels knowledge of adverse effects associated with anticholinergic drugs. The 7 scales calculated the anticholinergic burden by summing the scores of each anticholinergic medicine with the assumption that the anticholinergic activity is linear and additive. The inclusion and rating of medicines with anticholinergic activity were predominantly influenced by subjective decisions. The final score was based on median values of ratings by each panel member. As a result, there are large differences between the published lists: e.g., beta‐blockers atenolol or metoprolol were assessed as anticholinergic drugs only in the studies of Han et al. [22] and Boustani et al. [24] (rating score 1) compared to the other rating scales.
Discrepancies in rating of anticholinergic medicines were noted in the scales. For example, quetiapine was reported as having high anticholinergic activity [24] in one scale (n = 1), moderate [22] in another scale (n = 1) and low [19,26] in two other scales (n = 2). A compiled reference composite scale which displays all 195 anticholinergic medicines extracted from the 7 anticholinergic rating scales is shown in Table 2 [9,19,22-26]. Similarly, a recent review collated a list of 100 medicines with definite or possible anticholinergic effects based on previously published list of anticholinergic risk scales and in conjunction with Martindale as a reference text [36].
The current anticholinergic risk scales tend to simplify the complexity of pharmacological mechanisms, which is quite challenging in geriatric risk assessment in older populations due to increased biological variation. However, there is no standardised consensus on how to quantify the anticholinergic burden and it is difficult to compare the study results from different methods and studies that have used the same method because different cut-off values for anticholinergic burden have been reported [3]. The majority of scales have not considered the multiple actions of medicines on the muscarinic receptor subtypes, the possible synergistic or antagonistic effects of medicines, or possible development of tolerance for anticholinergic medicine effects over time. Moreover, anticholinergic adverse effects are dose-dependent and the relative anticholinergic activities of various medicines are unlikely to be proportional to a 0:1:2:3 ratio. Also, there was no consensus on the definition of an anticholinergic medicine, and both the number and ranking of the anticholinergic drugs listed vary considerably between the scales [3,37]. Some scales considered the impact of different routes of administration when ranking the anticholinergic activity of medicines, while others excluded topical, ophthalmic, otologic and inhaled preparations.
This systematic review was comprehensive in that the electronic search conducted in 3 different databases endeavoured to identify all potential studies that met our eligibility criteria. The review explicitly looked into the anticholinergic scales partly or fully developed based on expert opinion. In addition to this, citation analysis provides further details about validation of the included scales. The objectives were clearly stated and the search methodology including the citation analysis was robust. A systematic approach was used to synthesise and characterise the findings of this review.

Conclusions

Medicines with anticholinergic activity are frequently prescribed in older people, and several rating scales have been developed to quantify anticholinergic burden. There is not one standardised rating scale for measuring anticholinergic burden. The reference composite scale developed would be a useful tool for clinicians to identify medicines with anticholinergic activity.

Acknowledgements

MSS was funded by Doctoral Scholarship of University of Otago, New Zealand. The funder and sponsor had no role in study design or data collection, analysis or interpretation.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MSS and PSN designed the search strategy, extracted data, analysed data and drafted the manuscript. SBD assisted with analyses and drafting the manuscript. All authors contributed to data analyses and interpretation, critically commented on, and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medications by older adults with dementia. J Am Geriatr Soc. 2002;50(5):836–42.PubMedCrossRef Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medications by older adults with dementia. J Am Geriatr Soc. 2002;50(5):836–42.PubMedCrossRef
2.
Zurück zum Zitat Tune LE. Anticholinergic effects of medication in elderly patients. J Clin Psychiatry. 2001;62 Suppl 21:11–4.PubMed Tune LE. Anticholinergic effects of medication in elderly patients. J Clin Psychiatry. 2001;62 Suppl 21:11–4.PubMed
3.
Zurück zum Zitat Kersten H, Wyller TB. Anticholinergic drug burden in older people's brain - how well is it measured? Basic Clin Pharmacol Toxicol. 2014;114(2):151–9.PubMedCrossRef Kersten H, Wyller TB. Anticholinergic drug burden in older people's brain - how well is it measured? Basic Clin Pharmacol Toxicol. 2014;114(2):151–9.PubMedCrossRef
4.
Zurück zum Zitat Cao YJ, Mager DE, Simonsick EM, Hilmer SN, Ling SM, Windham BG, et al. Physical and cognitive performance and burden of anticholinergics, sedatives, and ACE inhibitors in older women. Clin Pharmacol Therapeut. 2008;83(3):422–9.CrossRef Cao YJ, Mager DE, Simonsick EM, Hilmer SN, Ling SM, Windham BG, et al. Physical and cognitive performance and burden of anticholinergics, sedatives, and ACE inhibitors in older women. Clin Pharmacol Therapeut. 2008;83(3):422–9.CrossRef
5.
Zurück zum Zitat Hilmer SN, Mager DE, Simonsick EM, Cao Y, Ling SM, Windham BG, et al. A drug burden index to define the functional burden of medications in older people. Arch Intern Med. 2007;167(8):781–7.PubMedCrossRef Hilmer SN, Mager DE, Simonsick EM, Cao Y, Ling SM, Windham BG, et al. A drug burden index to define the functional burden of medications in older people. Arch Intern Med. 2007;167(8):781–7.PubMedCrossRef
6.
Zurück zum Zitat Lechevallier-Michel N, Molimard M, Dartigues JF, Fabrigoule C, Fourrier-Reglat A. Drugs with anticholinergic properties and cognitive performance in the elderly: results from the PAQUID Study. Br J Clin Pharmacol. 2005;59(2):143–51.PubMedPubMedCentralCrossRef Lechevallier-Michel N, Molimard M, Dartigues JF, Fabrigoule C, Fourrier-Reglat A. Drugs with anticholinergic properties and cognitive performance in the elderly: results from the PAQUID Study. Br J Clin Pharmacol. 2005;59(2):143–51.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Tune LE, Egeli S. Acetylcholine and delirium. Dement Geriatr Cognit Disord. 1999;10(5):342–4.CrossRef Tune LE, Egeli S. Acetylcholine and delirium. Dement Geriatr Cognit Disord. 1999;10(5):342–4.CrossRef
8.
Zurück zum Zitat Boustani M, Schubert C, Sennour Y. The challenge of supporting care for dementia in primary care. Clin Interv Aging. 2007;2(4):631–6.PubMedPubMedCentral Boustani M, Schubert C, Sennour Y. The challenge of supporting care for dementia in primary care. Clin Interv Aging. 2007;2(4):631–6.PubMedPubMedCentral
9.
Zurück zum Zitat Carnahan RM, Lund BC, Perry PJ, Pollock BG, Culp KR. The Anticholinergic Drug Scale as a measure of drug-related anticholinergic burden: associations with serum anticholinergic activity. J Clin Pharmacol. 2006;46(12):1481–6.PubMedCrossRef Carnahan RM, Lund BC, Perry PJ, Pollock BG, Culp KR. The Anticholinergic Drug Scale as a measure of drug-related anticholinergic burden: associations with serum anticholinergic activity. J Clin Pharmacol. 2006;46(12):1481–6.PubMedCrossRef
10.
Zurück zum Zitat Gnjidic D, Cumming RG, Le Couteur DG, Handelsman DJ, Naganathan V, Abernethy DR, et al. Drug Burden Index and physical function in older Australian men. Br J Clin Pharmacol. 2009;68(1):97–105.PubMedPubMedCentralCrossRef Gnjidic D, Cumming RG, Le Couteur DG, Handelsman DJ, Naganathan V, Abernethy DR, et al. Drug Burden Index and physical function in older Australian men. Br J Clin Pharmacol. 2009;68(1):97–105.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Hilmer SN, Mager DE, Simonsick EM, Ling SM, Windham BG, Harris TB, et al. Drug burden index score and functional decline in older people. Am J Med. 2009;122(12):1142–9. e1141-1142.PubMedPubMedCentralCrossRef Hilmer SN, Mager DE, Simonsick EM, Ling SM, Windham BG, Harris TB, et al. Drug burden index score and functional decline in older people. Am J Med. 2009;122(12):1142–9. e1141-1142.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Landi F, Russo A, Liperoti R, Cesari M, Barillaro C, Pahor M, et al. Anticholinergic drugs and physical function among frail elderly population. Clin Pharmacol Therapeut. 2007;81(2):235–41.CrossRef Landi F, Russo A, Liperoti R, Cesari M, Barillaro C, Pahor M, et al. Anticholinergic drugs and physical function among frail elderly population. Clin Pharmacol Therapeut. 2007;81(2):235–41.CrossRef
13.
Zurück zum Zitat Nishtala PS, McLachlan AJ, Bell JS, Chen TF. Determinants of antipsychotic medication use among older people living in aged care homes in Australia. Int J Geriatr Psychiatry. 2010;25(5):449–57.PubMedCrossRef Nishtala PS, McLachlan AJ, Bell JS, Chen TF. Determinants of antipsychotic medication use among older people living in aged care homes in Australia. Int J Geriatr Psychiatry. 2010;25(5):449–57.PubMedCrossRef
14.
Zurück zum Zitat Nishtala PS, Fois RA, McLachlan AJ, Bell JS, Kelly PJ, Chen TF. Anticholinergic activity of commonly prescribed medications and neuropsychiatric adverse events in older people. J Clin Pharmacol. 2009;49(10):1176–84.PubMedCrossRef Nishtala PS, Fois RA, McLachlan AJ, Bell JS, Kelly PJ, Chen TF. Anticholinergic activity of commonly prescribed medications and neuropsychiatric adverse events in older people. J Clin Pharmacol. 2009;49(10):1176–84.PubMedCrossRef
15.
Zurück zum Zitat Kumpula EK, Bell JS, Soini H, Pitkala KH. Anticholinergic drug use and mortality among residents of long-term care facilities: a prospective cohort study. J Clin Pharmacol. 2011;51(2):256–63.PubMedCrossRef Kumpula EK, Bell JS, Soini H, Pitkala KH. Anticholinergic drug use and mortality among residents of long-term care facilities: a prospective cohort study. J Clin Pharmacol. 2011;51(2):256–63.PubMedCrossRef
16.
Zurück zum Zitat Panula J, Puustinen J, Jaatinen P, Vahlberg T, Aarnio P, Kivela SL. Effects of potent anticholinergics, sedatives and antipsychotics on postoperative mortality in elderly patients with hip fracture: a retrospective, population-based study. Drugs Aging. 2009;26(11):963–71.PubMedCrossRef Panula J, Puustinen J, Jaatinen P, Vahlberg T, Aarnio P, Kivela SL. Effects of potent anticholinergics, sedatives and antipsychotics on postoperative mortality in elderly patients with hip fracture: a retrospective, population-based study. Drugs Aging. 2009;26(11):963–71.PubMedCrossRef
17.
Zurück zum Zitat Wilson NM, Hilmer SN, March LM, Cameron ID, Lord SR, Seibel MJ, et al. Associations between drug burden index and falls in older people in residential aged care. J Am Geriatr Soc. 2011;59(5):875–80.PubMedCrossRef Wilson NM, Hilmer SN, March LM, Cameron ID, Lord SR, Seibel MJ, et al. Associations between drug burden index and falls in older people in residential aged care. J Am Geriatr Soc. 2011;59(5):875–80.PubMedCrossRef
18.
Zurück zum Zitat Montamat SC, Cusack BJ, Vestal RE. Management of drug therapy in the elderly. N Engl J Med. 1989;321(5):303–9.PubMedCrossRef Montamat SC, Cusack BJ, Vestal RE. Management of drug therapy in the elderly. N Engl J Med. 1989;321(5):303–9.PubMedCrossRef
19.
Zurück zum Zitat Rudolph JL, Salow MJ, Angelini MC, McGlinchey RE. The anticholinergic risk scale and anticholinergic adverse effects in older persons. Arch Intern Med. 2008;168(5):508–13.PubMedCrossRef Rudolph JL, Salow MJ, Angelini MC, McGlinchey RE. The anticholinergic risk scale and anticholinergic adverse effects in older persons. Arch Intern Med. 2008;168(5):508–13.PubMedCrossRef
20.
Zurück zum Zitat Boustani M, Baker MS, Campbell N, Munger S, Hui SL, Castelluccio P, et al. Impact and recognition of cognitive impairment among hospitalized elders. J Hosp Med. 2010;5(2):69–75.PubMedPubMedCentralCrossRef Boustani M, Baker MS, Campbell N, Munger S, Hui SL, Castelluccio P, et al. Impact and recognition of cognitive impairment among hospitalized elders. J Hosp Med. 2010;5(2):69–75.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Han L, Agostini JV, Allore HG. Cumulative anticholinergic exposure is associated with poor memory and executive function in older men. J Am Geriatr Soc. 2008;56(12):2203–10.PubMedPubMedCentralCrossRef Han L, Agostini JV, Allore HG. Cumulative anticholinergic exposure is associated with poor memory and executive function in older men. J Am Geriatr Soc. 2008;56(12):2203–10.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Sittironnarit G, Ames D, Bush AI, Faux N, Flicker L, Foster J, et al. Effects of anticholinergic drugs on cognitive function in older Australians: results from the AIBL study. Dement Geriatr Cogn Disord. 2011;31(3):173–8.PubMedCrossRef Sittironnarit G, Ames D, Bush AI, Faux N, Flicker L, Foster J, et al. Effects of anticholinergic drugs on cognitive function in older Australians: results from the AIBL study. Dement Geriatr Cogn Disord. 2011;31(3):173–8.PubMedCrossRef
24.
Zurück zum Zitat Boustani M, Campbell N, Munger S, Maidment I, Fox C. Impact of anticholinergics on the aging brain: a review and practical application. Aging Health. 2008;4(3):311–20.CrossRef Boustani M, Campbell N, Munger S, Maidment I, Fox C. Impact of anticholinergics on the aging brain: a review and practical application. Aging Health. 2008;4(3):311–20.CrossRef
25.
Zurück zum Zitat Ancelin ML, Artero S, Portet F, Dupuy AM, Touchon J, Ritchie K. Non-degenerative mild cognitive impairment in elderly people and use of anticholinergic drugs: longitudinal cohort study. BMJ. 2006;332(7539):455–9.PubMedPubMedCentralCrossRef Ancelin ML, Artero S, Portet F, Dupuy AM, Touchon J, Ritchie K. Non-degenerative mild cognitive impairment in elderly people and use of anticholinergic drugs: longitudinal cohort study. BMJ. 2006;332(7539):455–9.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Ehrt U, Broich K, Larsen JP, Ballard C, Aarsland D. Use of drugs with anticholinergic effect and impact on cognition in Parkinson's disease: a cohort study. J Neurol Neurosurg Psychiatry. 2010;81(2):160–5.PubMedCrossRef Ehrt U, Broich K, Larsen JP, Ballard C, Aarsland D. Use of drugs with anticholinergic effect and impact on cognition in Parkinson's disease: a cohort study. J Neurol Neurosurg Psychiatry. 2010;81(2):160–5.PubMedCrossRef
27.
Zurück zum Zitat Pasina L, Djade CD, Lucca U, Nobili A, Tettamanti M, Franchi C, et al. Association of anticholinergic burden with cognitive and functional status in a cohort of hospitalized elderly: comparison of the anticholinergic cognitive burden scale and anticholinergic risk scale: results from the REPOSI study. Drugs Aging. 2013;30(2):103–12.PubMedCrossRef Pasina L, Djade CD, Lucca U, Nobili A, Tettamanti M, Franchi C, et al. Association of anticholinergic burden with cognitive and functional status in a cohort of hospitalized elderly: comparison of the anticholinergic cognitive burden scale and anticholinergic risk scale: results from the REPOSI study. Drugs Aging. 2013;30(2):103–12.PubMedCrossRef
28.
29.
Zurück zum Zitat Lowry E, Woodman RJ, Soiza RL, Mangoni AA. Associations between the anticholinergic risk scale score and physical function: potential implications for adverse outcomes in older hospitalized patients. J Am Med Dir Assoc. 2011;12(8):565–72.PubMedCrossRef Lowry E, Woodman RJ, Soiza RL, Mangoni AA. Associations between the anticholinergic risk scale score and physical function: potential implications for adverse outcomes in older hospitalized patients. J Am Med Dir Assoc. 2011;12(8):565–72.PubMedCrossRef
30.
Zurück zum Zitat Uusvaara J, Pitkala K, Kautiainen H, Tilvis R, Strandberg T. Association of Anticholinergic Drugs with Hospitalization and Mortality among Older Cardiovascular Patients. Drugs Aging. 2011;28(2):131–8.PubMedCrossRef Uusvaara J, Pitkala K, Kautiainen H, Tilvis R, Strandberg T. Association of Anticholinergic Drugs with Hospitalization and Mortality among Older Cardiovascular Patients. Drugs Aging. 2011;28(2):131–8.PubMedCrossRef
31.
Zurück zum Zitat Moore AR, O'Keeffe ST. Drug-induced cognitive impairment in the elderly. Drugs Aging. 1999;15(1):15–28.PubMedCrossRef Moore AR, O'Keeffe ST. Drug-induced cognitive impairment in the elderly. Drugs Aging. 1999;15(1):15–28.PubMedCrossRef
32.
Zurück zum Zitat Campbell NL, Boustani MA, Lane KA, Gao S, Hendrie H, Khan BA, et al. Use of anticholinergics and the risk of cognitive impairment in an African American population. Neurology. 2010;75(2):152–9.PubMedPubMedCentralCrossRef Campbell NL, Boustani MA, Lane KA, Gao S, Hendrie H, Khan BA, et al. Use of anticholinergics and the risk of cognitive impairment in an African American population. Neurology. 2010;75(2):152–9.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Fox C, Richardson K, Maidment ID, Savva GM, Matthews FE, Smithard D, et al. Anticholinergic medication use and cognitive impairment in the older population: The medical research council cognitive function and ageing study. J Am Geriatr Soc. 2011;59(8):1477–83.PubMedCrossRef Fox C, Richardson K, Maidment ID, Savva GM, Matthews FE, Smithard D, et al. Anticholinergic medication use and cognitive impairment in the older population: The medical research council cognitive function and ageing study. J Am Geriatr Soc. 2011;59(8):1477–83.PubMedCrossRef
34.
Zurück zum Zitat Cai X, Campbell N, Khan B, Callahan C, Boustani M. Long-term anticholinergic use and the aging brain. Alzheimers Dement. 2013;9(4):377–85.PubMedCrossRef Cai X, Campbell N, Khan B, Callahan C, Boustani M. Long-term anticholinergic use and the aging brain. Alzheimers Dement. 2013;9(4):377–85.PubMedCrossRef
35.
Zurück zum Zitat Carriere I, Fourrier-Reglat A, Dartigues JF, Rouaud O, Pasquier F, Ritchie K, et al. Drugs with anticholinergic properties, cognitive decline, and dementia in an elderly general population: the 3-city study. Arch Intern Med. 2009;169(14):1317–24.PubMedPubMedCentralCrossRef Carriere I, Fourrier-Reglat A, Dartigues JF, Rouaud O, Pasquier F, Ritchie K, et al. Drugs with anticholinergic properties, cognitive decline, and dementia in an elderly general population: the 3-city study. Arch Intern Med. 2009;169(14):1317–24.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Duran CE, Azermai M, Vander Stichele RH. Systematic review of anticholinergic risk scales in older adults. Eur J Clin Pharmacol. 2013;69(7):1485–96.PubMedCrossRef Duran CE, Azermai M, Vander Stichele RH. Systematic review of anticholinergic risk scales in older adults. Eur J Clin Pharmacol. 2013;69(7):1485–96.PubMedCrossRef
37.
Zurück zum Zitat Salahudeen MS, Duffull SB, Nishtala PS. Impact of anticholinergic discontinuation on cognitive outcomes in older people: a systematic review. Drugs Aging. 2014;31(3):185–92.PubMedCrossRef Salahudeen MS, Duffull SB, Nishtala PS. Impact of anticholinergic discontinuation on cognitive outcomes in older people: a systematic review. Drugs Aging. 2014;31(3):185–92.PubMedCrossRef
38.
Zurück zum Zitat Chew ML, Mulsant BH, Pollock BG, Lehman ME, Greenspan A, Mahmoud RA, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc. 2008;56(7):1333–41.PubMedCrossRef Chew ML, Mulsant BH, Pollock BG, Lehman ME, Greenspan A, Mahmoud RA, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc. 2008;56(7):1333–41.PubMedCrossRef
39.
Zurück zum Zitat Kersten H, Molden E, Tolo IK, Skovlund E, Engedal K, Wyller TB. Cognitive effects of reducing anticholinergic drug burden in a frail elderly population: a randomized controlled trial. J Gerontol A Biol Sci Med Sci. 2013;68(3):271–8.PubMedCrossRef Kersten H, Molden E, Tolo IK, Skovlund E, Engedal K, Wyller TB. Cognitive effects of reducing anticholinergic drug burden in a frail elderly population: a randomized controlled trial. J Gerontol A Biol Sci Med Sci. 2013;68(3):271–8.PubMedCrossRef
40.
Zurück zum Zitat Kersten H, Molden E, Willumsen T, Engedal K, Bruun Wyller T. Higher anticholinergic drug scale (ADS) scores are associated with peripheral but not cognitive markers of cholinergic blockade. Cross sectional data from 21 Norwegian nursing homes. Br J Clin Pharmacol. 2013;75(3):842–9.PubMedPubMedCentral Kersten H, Molden E, Willumsen T, Engedal K, Bruun Wyller T. Higher anticholinergic drug scale (ADS) scores are associated with peripheral but not cognitive markers of cholinergic blockade. Cross sectional data from 21 Norwegian nursing homes. Br J Clin Pharmacol. 2013;75(3):842–9.PubMedPubMedCentral
41.
Zurück zum Zitat Lampela P, Lavikainen P, Garcia-Horsman JA, Bell JS, Huupponen R, Hartikainen S. Anticholinergic drug use, serum anticholinergic activity, and adverse drug events among older people: a population-based study. Drugs Aging. 2013;30(5):321–30.PubMedCrossRef Lampela P, Lavikainen P, Garcia-Horsman JA, Bell JS, Huupponen R, Hartikainen S. Anticholinergic drug use, serum anticholinergic activity, and adverse drug events among older people: a population-based study. Drugs Aging. 2013;30(5):321–30.PubMedCrossRef
42.
Zurück zum Zitat Kashyap M, Belleville S, Mulsant BH, Hilmer SN, Paquette A, le Tu M, et al. Methodological challenges in determining longitudinal associations between anticholinergic drug use and incident cognitive decline. J Am Geriatr Soc. 2014;62(2):336–41.PubMedCrossRef Kashyap M, Belleville S, Mulsant BH, Hilmer SN, Paquette A, le Tu M, et al. Methodological challenges in determining longitudinal associations between anticholinergic drug use and incident cognitive decline. J Am Geriatr Soc. 2014;62(2):336–41.PubMedCrossRef
43.
Zurück zum Zitat Juliebo V, Bjoro K, Krogseth M, Skovlund E, Ranhoff AH, Wyller TB. Risk factors for preoperative and postoperative delirium in elderly patients with hip fracture. J Am Geriatr Soc. 2009;57(8):1354–61.PubMedCrossRef Juliebo V, Bjoro K, Krogseth M, Skovlund E, Ranhoff AH, Wyller TB. Risk factors for preoperative and postoperative delirium in elderly patients with hip fracture. J Am Geriatr Soc. 2009;57(8):1354–61.PubMedCrossRef
44.
Zurück zum Zitat Drag LL, Wright SL, Bieliauskas LA. Prescribing practices of anticholinergic medications and their association with cognition in an extended care setting. J Appl Gerontol. 2012;31(2):239–59.CrossRef Drag LL, Wright SL, Bieliauskas LA. Prescribing practices of anticholinergic medications and their association with cognition in an extended care setting. J Appl Gerontol. 2012;31(2):239–59.CrossRef
45.
Zurück zum Zitat Mangoni AA, van Munster BC, Woodman RJ, de Rooij SE. Measures of anticholinergic drug exposure, serum anticholinergic activity, and all-cause postdischarge mortality in older hospitalized patients with hip fractures. Am J Geriatr Psychiatry. 2013;21(8):785–93.PubMedCrossRef Mangoni AA, van Munster BC, Woodman RJ, de Rooij SE. Measures of anticholinergic drug exposure, serum anticholinergic activity, and all-cause postdischarge mortality in older hospitalized patients with hip fractures. Am J Geriatr Psychiatry. 2013;21(8):785–93.PubMedCrossRef
46.
Zurück zum Zitat Kalisch Ellett LM, Pratt NL, Ramsay EN, Barratt JD, Roughead EE. Multiple anticholinergic medication use and risk of hospital admission for confusion or dementia. J Am Geriatr Soc. 2014;62(10):1916–22.PubMedCrossRef Kalisch Ellett LM, Pratt NL, Ramsay EN, Barratt JD, Roughead EE. Multiple anticholinergic medication use and risk of hospital admission for confusion or dementia. J Am Geriatr Soc. 2014;62(10):1916–22.PubMedCrossRef
47.
Zurück zum Zitat Agar M, Currow D, Plummer J, Seidel R, Carnahan R, Abernethy A. Changes in anticholinergic load from regular prescribed medications in palliative care as death approaches. Palliat Med. 2009;23(3):257–65.PubMedCrossRef Agar M, Currow D, Plummer J, Seidel R, Carnahan R, Abernethy A. Changes in anticholinergic load from regular prescribed medications in palliative care as death approaches. Palliat Med. 2009;23(3):257–65.PubMedCrossRef
48.
Zurück zum Zitat Yeh Y-C, Liu C-L, Peng L-N, Lin M-H, Chen L-K. Potential benefits of reducing medication-related anticholinergic burden for demented older adults: A prospective cohort study. Geriatr Gerontol Int. 2013;13(3):694–700.PubMedCrossRef Yeh Y-C, Liu C-L, Peng L-N, Lin M-H, Chen L-K. Potential benefits of reducing medication-related anticholinergic burden for demented older adults: A prospective cohort study. Geriatr Gerontol Int. 2013;13(3):694–700.PubMedCrossRef
49.
Zurück zum Zitat Lowry E, Woodman RJ, Soiza RL, Mangoni AA. Clinical and demographic factors associated with antimuscarinic medication use in older hospitalized patients. Hosp Pract (1995). 2011;39(1):30–6.CrossRef Lowry E, Woodman RJ, Soiza RL, Mangoni AA. Clinical and demographic factors associated with antimuscarinic medication use in older hospitalized patients. Hosp Pract (1995). 2011;39(1):30–6.CrossRef
50.
Zurück zum Zitat Koshoedo S, Soiza RL, Purkayastha R, Mangoni AA. Anticholinergic drugs and functional outcomes in older patients undergoing orthopaedic rehabilitation. Am J Geriatr Pharmacother. 2012;10(4):251–7.PubMedCrossRef Koshoedo S, Soiza RL, Purkayastha R, Mangoni AA. Anticholinergic drugs and functional outcomes in older patients undergoing orthopaedic rehabilitation. Am J Geriatr Pharmacother. 2012;10(4):251–7.PubMedCrossRef
51.
Zurück zum Zitat Huang K-H, Chan Y-F, Shih H-C, Lee C-Y. Relationship between Potentially Inappropriate Anticholinergic Drugs (PIADs) and Adverse Outcomes among Elderly Patients in Taiwan. J Food Drug Anal. 2012;20(4):930–7. Huang K-H, Chan Y-F, Shih H-C, Lee C-Y. Relationship between Potentially Inappropriate Anticholinergic Drugs (PIADs) and Adverse Outcomes among Elderly Patients in Taiwan. J Food Drug Anal. 2012;20(4):930–7.
52.
Zurück zum Zitat Kolanowski A, Fick DM, Campbell J, Litaker M, Boustani M. A preliminary study of anticholinergic burden and relationship to a quality of life indicator, engagement in activities, in nursing home residents with dementia. J Am Med Dir Assoc. 2009;10(4):252–7.PubMedPubMedCentralCrossRef Kolanowski A, Fick DM, Campbell J, Litaker M, Boustani M. A preliminary study of anticholinergic burden and relationship to a quality of life indicator, engagement in activities, in nursing home residents with dementia. J Am Med Dir Assoc. 2009;10(4):252–7.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Campbell N, Perkins A, Hui S, Khan B, Boustani M. Association between prescribing of anticholinergic medications and incident delirium: a cohort study. J Am Geriatr Soc. 2011;59 Suppl 2:S277–81.PubMedPubMedCentralCrossRef Campbell N, Perkins A, Hui S, Khan B, Boustani M. Association between prescribing of anticholinergic medications and incident delirium: a cohort study. J Am Geriatr Soc. 2011;59 Suppl 2:S277–81.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Fox C, Livingston G, Maidment ID, Coulton S, Smithard DG, Boustani M, et al. The impact of anticholinergic burden in Alzheimer's dementia-the LASER-AD study. Age Ageing. 2011;40(6):730–5.PubMedCrossRef Fox C, Livingston G, Maidment ID, Coulton S, Smithard DG, Boustani M, et al. The impact of anticholinergic burden in Alzheimer's dementia-the LASER-AD study. Age Ageing. 2011;40(6):730–5.PubMedCrossRef
55.
Zurück zum Zitat Koyama A, Steinman M, Ensrud K, Hillier TA, Yaffe K. Long-term Cognitive and Functional Effects of Potentially Inappropriate Medications in Older Women. J Gerontol A: Biol Med Sci. 2014;69(4):423–9.CrossRef Koyama A, Steinman M, Ensrud K, Hillier TA, Yaffe K. Long-term Cognitive and Functional Effects of Potentially Inappropriate Medications in Older Women. J Gerontol A: Biol Med Sci. 2014;69(4):423–9.CrossRef
56.
Zurück zum Zitat Koyama A, Steinman M, Ensrud K, Hillier TA, Yaffe K. Ten-year trajectory of potentially inappropriate medications in very old women: importance of cognitive status. J Am Geriatr Soc. 2013;61(2):258–63.PubMedPubMedCentralCrossRef Koyama A, Steinman M, Ensrud K, Hillier TA, Yaffe K. Ten-year trajectory of potentially inappropriate medications in very old women: importance of cognitive status. J Am Geriatr Soc. 2013;61(2):258–63.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Shah RC, Janos AL, Kline JE, Yu L, Leurgans SE, Wilson RS, et al. Cognitive decline in older persons initiating anticholinergic medications. PLoS One. 2013;8(5):e64111.PubMedPubMedCentralCrossRef Shah RC, Janos AL, Kline JE, Yu L, Leurgans SE, Wilson RS, et al. Cognitive decline in older persons initiating anticholinergic medications. PLoS One. 2013;8(5):e64111.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Kidd AC, Musonda P, Soiza RL, Butchart C, Lunt CJ, Pai Y, et al. The relationship between total anticholinergic burden (ACB) and early in-patient hospital mortality and length of stay in the oldest old aged 90 years and over admitted with an acute illness. Arch Gerontol Geriatr. 2014;59(1):155–61.PubMedCrossRef Kidd AC, Musonda P, Soiza RL, Butchart C, Lunt CJ, Pai Y, et al. The relationship between total anticholinergic burden (ACB) and early in-patient hospital mortality and length of stay in the oldest old aged 90 years and over admitted with an acute illness. Arch Gerontol Geriatr. 2014;59(1):155–61.PubMedCrossRef
Metadaten
Titel
Anticholinergic burden quantified by anticholinergic risk scales and adverse outcomes in older people: a systematic review
verfasst von
Mohammed Saji Salahudeen
Stephen B Duffull
Prasad S Nishtala
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Geriatrics / Ausgabe 1/2015
Elektronische ISSN: 1471-2318
DOI
https://doi.org/10.1186/s12877-015-0029-9

Weitere Artikel der Ausgabe 1/2015

BMC Geriatrics 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.