840
Views
16
CrossRef citations to date
0
Altmetric
Review

Adverse Drug Reactions of Acetylcholinesterase Inhibitors in Older People Living with Dementia: A Comprehensive Literature Review

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 927-949 | Published online: 04 Sep 2021

Abstract

The rising of global geriatric population has contributed to increased prevalence of dementia. Dementia is a neurodegenerative disease, which is characterized by progressive deterioration of cognitive functions, such as judgment, language, memory, attention and visuospatial ability. Dementia not only has profoundly devastating physical and psychological health outcomes, but it also poses a considerable healthcare expenditure and burdens. Acetylcholinesterase inhibitors (AChEIs), or so-called anti-dementia medications, have been developed to delay the progression of neurocognitive disorders and to decrease healthcare needs. AChEIs have been widely prescribed in clinical practice for the treatment of Alzheimer’s disease, which account for 70% of dementia. The rising use of AChEIs results in increased adverse drug reactions (ADRs) such as cardiovascular and gastrointestinal adverse effects, resulting from overstimulation of peripheral cholinergic activity and muscarinic receptor activation. Changes in pharmacokinetics (PK), pharmacodynamics (PD) and pharmacogenetics (PGx), and occurrence of drug interactions are said to be major risk factors of ADRs of AChEIs in this population. To date, comprehensive reviews in ADRs of AChEIs have so far been scarcely studied. Therefore, we aimed to recapitulate and update the diverse aspects of AChEIs, including the mechanisms of action, characteristics and risk factors of ADRs, and preventive strategies of their ADRs. The collation of this knowledge is essential to facilitate efforts to reduce ADRs of AChEIs.

Introduction

Globally, the number of older population aged 60 years or over was 962 million in 2017 and will almost double to reach 2.1 billion by 2050.Citation1,Citation2 The rising geriatric population results in an exponential increase in incidence of neurodegenerative disorders such as dementia.Citation3,Citation4 Worldwide, the population of people living with dementia was estimated at 50 million in 2017 and is predicted to increase to 131.5 million by 2050.Citation5,Citation6 Dementia is described as symptoms related to a cluster of major neurocognitive disorders or conditions which are usually manifested as slowly progressive decline of multiple cortical functions including orientation, comprehension, memory, language, learning skills and problem-solving ability.Citation7 The most common type of dementia is AD (50–75%), followed by vascular dementia (20%), dementia with Lewy bodies (5%) and finally frontotemporal lobar dementia (5%).Citation8Citation10 In AD, the progressive loss of cholinergic neurons in the basal forebrain leads to a decrease in acetylcholine (ACh) which is essential in cognition and neuroprotection.Citation11 Dementia has a devastating impact on healthcare infrastructures in economic and medical aspects. This neurodegenerative disease is one of the leading causes of death and contributors to premature disability and dependency burdens.Citation5,Citation12,Citation13 With increased disability, dementia could be overwhelming for caregivers and families, leading to increased healthcare needs.Citation3,Citation12,Citation14Citation17 Appropriate management, including non-pharmacological and pharmacological therapies, are necessary to delay worsening of symptoms and to reduce healthcare burdens.Citation18 Anti-dementia medications are being used worldwide, especially in Alzheimer’s disease (AD), which is the most common type of dementia.Citation19 One-fourth of older people with dementia are prescribed anti-dementia medications which are classified into two classes: Acetylcholinesterase inhibitors (AChEIs) and N-Methyl-D-aspartate (NMDA) receptor antagonists.Citation20,Citation21 AChEIs were the first pharmacological treatment approved by the US Food and Drug Administration (FDA) for AD and have been reported to be used in 10–20% of dementia patients.Citation22Citation25

The aging population usually have multiple other chronic diseases as well as behavioral and psychological symptoms of dementia (BPSD),Citation12,Citation22,Citation26Citation35 resulting in the concurrent use of five or more medications or polypharmacy.Citation26 The exposure of 82–98% of people with dementia to polypharmacy was reported in previous studies.Citation36Citation38 This can lead to a greater risk of undesirable or harmful reactions to medications or adverse drug reactions (ADRs).Citation39Citation41 The alterations in pharmacokinetics (PK), pharmacodynamics (PD) and pharmacogenetics (PGx) of AChEIs also result in higher risk of AChEIs’ ADR.Citation42Citation47 Over the last decades, there has been an increase in the reports of AChEI-induced ADRs with 70% being severe and up to 2.3% being fatal ADRs.Citation48Citation50

Therefore, the significance of the paper is to facilitate effort to address the issue of AChEI-induced ADRs among older patients with dementia. We aim to review and update the diverse aspects of AChEIs such as the mechanisms of action, characteristics and risk factors of ADRs, and preventive strategies of their ADRs.

Search Strategy

PubMed, Scopus and Web of Science databases were searched for relevant articles published in English from January 1, 1976 until March 31, 2021. The search terms were “donepezil”, “galantamine”, “rivastigmine”, “acetylcholinesterase inhibitors”, “dementia”, “Alzheimer’s disease”, “older adults”, “mechanism”, “pharmacokinetics”, “pharmacodynamics”, “pharmacogenetics”, “adverse drug reactions”, “drug-drug interactions”, “prevention”. Google Scholar was searched using main keywords for any additional studies.

Acetylcholinesterase Inhibitors

Mechanism of Acetylcholinesterase Inhibitors

ACh is mostly hydrolyzed by acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE).Citation51 Both enzymes are responsible for rapid hydrolysis of ACh in synaptic clefts, producing the products: Choline and acetate. AChE predominates in the human brain whereas BuChE is widely distributed in peripheral nervous system (PNS) and other organs such as intestine, heart and liver.Citation52Citation54 In the brain, AChE levels are usually high in synapses while BuChE is distributed in glial cells.Citation53 In AD patients, BuChE has progressively increased activity in particular part of the brain such as hippocampal area and has raised accumulation of Aβ-aggregation and neurofibrillary tangles, resulting in the reduction of ACh.Citation52,Citation55Citation61 Therefore, a class of AChEIs is developed to block both AChE and BuChE in the synaptic clefts to reduce the degradation of Ach.Citation19 Furthermore, one AChEI has a pharmacological property for modulation of muscarinic or nicotinic receptors, contributing to enhancement of cholinergic activity.Citation62 AChEIs increase cholinergic activities to improve and sustain cognitive functions and ADLs as well as to make better psycho-behavioral symptoms in dementia patients.Citation22 However, AChEIs also inhibit rapid hydrolysis of ACh in PNS including sympathetic autonomic nervous system, and parasympathetic and preganglionic sympathetic neurons. This leads to peripheral adverse outcomes, such as diarrhea, nausea and vomiting, dizziness, and muscle cramping.Citation63

The first-generation of AChEIs such as tacrine, velnacrine, and physostigmine were removed from the market because of high incidence rates of potential drug interactions and serious side effects.Citation64 Three second-generation AChEIs were rapidly developed to replace the first-generation AChEIs. Donepezil, galantamine and rivastigmine have been approved by the FDA for the treatment of AD. Donepezil (58.4%) is the most frequently prescribed AChEIs, followed by rivastigmine (13.6%) and galantamine (12.4%).Citation22,Citation65Citation67 Donepezil in oral form and rivastigmine transdermal patches have received regulatory approval for the treatment of all stages of AD ranging from mild to severe.Citation19,Citation22,Citation68Citation80 There is no significant difference between the efficacy of these AChEIs in terms of improving psychometric and cognitive scales.Citation81 However, three AChEIs differ in both PK and PD properties,Citation82Citation84 as shown in Supplementary Table 1.

Donepezil

Donepezil was the first AChEI approved by the FDA for AD treatment in 1996. It is a piperidine-based reversible inhibitor of AChE.Citation82,Citation85 Donepezil is 500-fold selective for AChE inhibition in comparison with BuChE.Citation86,Citation87 The oral bioavailability is 100% and time to peak plasma concentration (Tmax) approximates 3–5 hours following a single-dose administration.Citation88,Citation89 Plasma albumin binding is 75% and volume of distribution (Vd) is 12 L/kg.Citation88Citation90 Donepezil readily transports across the blood brain barrier (BBB), resulting in 7-fold higher concentration in the brain compared with plasma. Cytochrome P450 2D6 (CYP2D6) is accounted for the major Phase I metabolism (90%) and the rest by Cytochrome P450 3A4 (CYP3A4).Citation89,Citation91 6-O-desmethyl donepezil (6DD) is the end product of Phase II metabolism which is excreted via kidney route.Citation88,Citation92 The average apparent plasma clearance is approximately 0.13–0.19 L/h/kg. According to its elimination half-life (70 hours), it takes around 15 days to reach the steady state. Then, it is conveniently administered as once daily.Citation88,Citation89 Both 5 mg and 10 mg once daily administration for 24 weeks could improve cognitive and quality of life scales in mild to moderate AD patients.Citation93Citation95 The initial dose should be administered initially with 5 mg/day, followed by slow-dose titration every 4–6 weeks along with the clinical status monitoring until reaching the maximum dose of 10 mg for mild to moderate AD.Citation64,Citation95 For severe AD, the maximum daily dose of donepezil is 23 mg once daily.Citation76

Rivastigmine

Rivastigmine was approved by the FDA to be marketed in 1997. Rivastigmine is classified as a carbamate substance.Citation81,Citation82,Citation85 Its mechanism of action is a slow reversible or pseudo-irreversible inhibition of both AChE and BuChE.Citation85,Citation96,Citation97 The oral bioavailability is poor, approximate 40% with Tmax ranging from 0.5 to 2 hours following oral single-dose administration.Citation88,Citation98 Plasma protein binding is 40% and Vd is 1.8–2.7 L/kg.Citation88,Citation99,Citation100 Rivastigmine easily passes through the BBB to exert activity in the brain.Citation101,Citation102 Intestinal esterase is the major enzyme responsible for first pass metabolism and the rest is minimally metabolized by liver cytochrome P450.Citation88 The main metabolite is NAP 226–90 which is rapidly excreted by renal system.Citation88 The plasma clearance of rivastigmine is estimated to be 1.5 L/h/kg. Its half-life is short, nearly 1.5 hours. Therefore, twice-daily dosing is recommended in clinical practice.Citation88,Citation100 Several double-blind controlled studies showed significant improvement in cognitive and global functions with 6 month-treatment.Citation103,Citation104 Clinical studies pointed out the effective doses of rivastigmine to be 6 to 12 mg per day.Citation64,Citation105 Rivastigmine is recommended to start at the dose of 1.5 mg twice-daily as capsules or liquid formulations and slowly titrate up to 6 mg twice-daily at intervals of every 2 to 4 weeks.Citation81,Citation103 In terms of other efficacy of rivastigmine, the improvement of peripheral insulin resistance has not been reported.Citation106 Transdermal patch is another preparation which delivers rivastigmine constantly into the blood circulation without level fluctuation.Citation71,Citation97,Citation107 The therapeutic dose of transdermal patch delivering rivastigmine is 4.6 mg per 24 hours to 13.3 mg per 24 hours in clinical practice.Citation107,Citation108 Rivastigmine patch is suggested to be started at 4.6 mg per 24 hours for at least for 4 weeks and then to be increased to 9.5 mg per 24 hours. After a minimum of 6 months a dose of 9.5 mg per 24 hours, 13.3 mg per 24 hours is recommended for well-tolerated patients with progressive cognitive decline.Citation109

Galantamine

Galantamine was approved by the FDA in 2000 for the treatment of AD.Citation110 This agent is a tertiary alkaloid-based compound that acts as both rapidly reversible-competitive inhibitor of AChE and a positive allosteric modulator of nicotinic acetylcholine receptors.Citation62,Citation85,Citation110 The oral bioavailability of galantamine ranges from 85 to 100% with rapid absorption.Citation88,Citation111 The Tmax is approximately 52 minutes following a single oral administration. Unlike donepezil, protein binding of galantamine is less than 50% and the mean Vd is 2.64 L/kg.Citation88,Citation111 This medication is demethylated and oxidized by CYP2D6 and CYP3A4. The active metabolite of galantamine is sanguinine or O-desmethyl galantamine.Citation88,Citation112 Galantamine goes through glucuronidation forming a water-soluble metabolite which is excreted via the renal route.Citation88,Citation111,Citation112 The total plasma clearance of galantamine is 0.34 L/h/kg.Citation113 Due to its short half-life of 6–8 hours, twice-daily dosing is recommended.Citation88,Citation111,Citation113 The formulation of galantamine consists of both immediate-release tablets and extended-release capsules. The efficacy of both extended and immediate release tablets of galantamine was studied in a randomized, double-blind, placebo-controlled trails, using doses titrating up to 16 or 24 mg per day for 6 months duration of treatment in patients with mild to moderate AD.Citation104,Citation114,Citation115 These studies demonstrated a significant improvement in cognitive and neuropsychiatric scales.Citation114 The recommended therapeutic dose of galantamine is 8 mg per day and gradually escalates every 4 weeks up to a maximum daily dose of 24 mg.Citation116

Therefore, the safety and effectiveness of AChEIs should be evaluated in older population in whom adverse reactions may be serious.Citation117 In terms of efficacy and effectiveness assessment of AChEIs, the common tools for cognitive evaluation includes Mini-Mental-State Examination (MMSE),Citation118 Alzheimer’s Disease Assessment Scale-Cognitive subscale (ADAS-Cog),Citation119 and Severe Impairment Battery (SIB).Citation120 Other tests are used to measures functional status and psycho-behavioral symptoms are the Basic and Instrumental Activity of Daily LivingCitation121Citation123 and Neuropsychiatric inventory (NPI),Citation124 respectively. According to previous clinical practice guideline based on systematic reviews,Citation104,Citation125,Citation126 AChEIs treatment for dementia contributed to marginally significant improvement of cognitive function, functional and global status, and psycho-behavioural symptoms.Citation117,Citation125,Citation127 In mild to moderate AD patients, meta-analyses on AChEIs have revealed the results with cognitive improvements on 1.5 points in MMSE and 2.5 points in ADAS-cog, comparing to the placebo.Citation19 The Pooled data presented an improvement of 0.1 standard deviations of ADLsCitation104,Citation125 and 2 of 144 points in NPI.Citation128 Besides cognitive and behavioural improvement, AChEIs have positive effects on balance and gait function without orthostatic hypotension.Citation129 However, there is limited evidence of AChEI efficacy and effectiveness in severe dementia, advanced age and long-term treatment.Citation19,Citation130,Citation131

Adverse Drug Reactions of Acetylcholinesterase Inhibitors

The prevalence of AChEI-induced ADRs tends upward significantly in older population with dementia.Citation48 In a 16-year period study, the number of AChEI-induced ADRs increased from 1924 ADRs in 1998 to 2961 ADRs in 2013.Citation48 Most reported cases are serious ADRs (50–70%) of which 2.3% are fatal ADRs.Citation48Citation50 AChEIs have a dose-related toxicity and a narrow therapeutic index. Therefore, the prevalence of ADRs has an upward trend with an increasing dose.Citation48 Most ADRs of AChEIs are described as type A reactions which are associated with dose and altered PK and PD. However, most type A reactions are potentially preventable In a recent study, preventable ADRs from prescription and administration errors were presented in 2.0% of all serious cases.Citation48 According to the mechanism of AChEI action, overstimulation of central and peripheral muscarinic and nicotinic receptors may contribute to diarrhea, nausea, vomiting, vagotonic effects (bradycardia, heart block, syncope), tremor, insomnia, urinary incontinence, and seizure.Citation63,Citation132Citation135 Common ADRs induced by AChEIs are principally neuropsychiatric (17%), gastrointestinal (16.2%), and cardiovascular (11.2%) in natureCitation49 as a result of overstimulation of peripheral cholinergic activity and muscarinic receptor activation, as revealed in Supplementary Table 2.Citation48,Citation72,Citation83,Citation132,Citation133

Gastrointestinal Adverse Effects

Oral administration of AChEIs increases gastric acid secretion of hydrochloric acid and internal propulsion which lead to the increase of gastrointestinal adverse effects, namely gastrointestinal ulceration and bleeding, especially for the concomitant use of AChEI and NSAIDs.Citation136,Citation137 Commonly reported gastrointestinal adverse effects are abdominal pain, nausea, vomiting, diarrhea, and poor appetite.Citation72,Citation133,Citation138Citation141 The increase of gastrointestinal side effects is associated with the rapid escalation of AChEI dose.Citation142

Cardiovascular Adverse Effects

Both conduction and sinus node function gradually deteriorate with advanced age. Moreover, AChEI increases the availability of choline in the heart and vagotonic effects via muscarinic receptors.Citation143,Citation144 Cardiovascular side effects are some of the most common peripheral adverse cholinergic effects. Therefore, older adults treated with AChEIs are at greater risk of life-threatening conduction dysfunction such as sinoatrial and atrioventricular block,Citation140,Citation145,Citation146 severe sinus bradycardiaCitation147 and QT interval prolongation with torsades de pointes (TdP).Citation148Citation150 Wandering atrial pacemaker (WAP) is another uncommon cardiac side effect in patients treated with donepezil. This condition is an atrial arrhythmia which presents with at least three distinctly different P wave morphologies.Citation151 Negative chronotropic effects contribute to detrimental health outcomes including syncope, pacemaker insertion, falls, fractures, hospitalization.Citation147,Citation152Citation154 However, there is controversy that AChEIs result in negative chronotropic effects.Citation155Citation158 Therefore, older people receiving AChEIs should be routinely asked regarding syncope histories and be evaluated for arrhythmia or bradycardia by physical examination and electrocardiogram.Citation159 Concomitant use of AChEIs and drug-induced QT prolongation such as beta-blockers, antiarrhythmic drugs and antipsychotics should be closely monitored by physicians and pharmacists.Citation159 In contrast, AChEIs treatment may be correlated with lower risk of cardiovascular events.Citation158

Neurological and Psychological Adverse Effects

Neurological side effects mainly result from excessive activation of nicotinic receptors. Common neurological adverse effects are dizziness, dyskinesia, convulsion, muscle cramps, insomnia, and vivid dream. The epileptic seizure is a very rare neurological adverse effects induced by AChEIs.Citation160,Citation161 From previous report, patient with mild AD treated with 10 mg donepezil once daily for 3 weeks presented convulsions during the treatment.Citation160 Moreover, AChEI-induced seizures may result from nutritional and metabolic disorders such as hyponatremia.Citation161 The vivid dream results from the disorder of brainstem cholinergic systems in processing rapid eye movement sleep. One characteristic of the vivid dream is extremely realistic. Vivid dreams usually appear in patients treated with donepezil in the evening owing to peak plasma concentration at night.Citation63,Citation162 Psychiatric adverse outcomes in older adults treated with AChEIs may include worsening of hallucination, anxiety, aggression, and confusion.Citation163,Citation164 Psychiatric problems are usually presented in dementia patients treated with high doses of AChEIs.

Respiratory Adverse Effects

Bronchospasm was presented as a pulmonary side effect after AChEI administration.Citation165 Therefore, patients with a history of bronchoconstriction should be closely monitored during treatment.Citation165 Furthermore, nasal problems could be presented among patients treated with AChEI.Citation165 There is no report of respiratory failure from AChEIs for dementia treatment.

Genitourinary Adverse Effects

Urinary incontinence may occur after treatment with AChEIs, in particular for galantamine.Citation166 The mechanism is related to nicotinic Ach receptor stimulation at the neuromuscular junction, resulting in an increased peripheral ACh.

Dermatological Adverse Effects

Rivastigmine could be used in the form of a skin patch. The most common skin adverse reaction is irritant contact dermatitis as a local skin reaction which is not associated with an immunological process. Its manifestation is localized erythema and itching.Citation167,Citation168 These symptoms usually resolve within 48 hours after patch removal. As a rare dermatological adverse reaction, allergic contact dermatitis is delayed type-IV immunologic reaction and manifests as erythema, vesicles and edema appearing more than 48 hours after rivastigmine patch removal. The life-threatening skin adverse reaction called Stevens-Johnson Syndrome (SJS) can occur in patients treated with oral or dermal administration and was reported in patients treated with galantamine.Citation138,Citation167Citation169

Uncommon Adverse Effects

According to post-marketing surveillance, a rare dystonic reaction called Pisa syndrome has been reported in patients receiving AChEIs. This syndrome is described as tonic flexion of the head and trunk one side accompanied by slight axial rotation.Citation170 The pathophysiology of the syndrome results from dopaminergic-cholinergic imbalance. Pisa syndrome was reported in a patients receiving 9 mg per day rivastigmine for 2 years.Citation171,Citation172 However, this abnormal syndrome disappeared when the drug dose was decreased.Citation171 Rhabdomyolysis and neuroleptic malignant syndromes are uncommon side effects which have been reported in older adults receiving donepezil.Citation173Citation175 Furthermore, hemolytic anemia, syndrome of inappropriate antidiuretic hormone (SIADH),Citation161 and severe hepatitisCitation176 also present as uncommon adverse effects in clinical practice.

AChEI-induced adverse effects may provide chance for prescribing cascades. AChEIs activate muscarinic receptors in urinary tract, leading to strong contraction of detrusor muscle and urinary incontinence. Therefore, bladder anticholinergic agents such as oxybutynin, tolterodine, trospium and solifenacin are usually used to relieve urge incontinence which called AChEI-induced urinary incontinence prescribing cascade.Citation177 Another common prescribing cascade is AChEI-induced rhinorrhea which is concomitant use of rhinorrhea medications to relieve side effects of AChEIs. The rhinorrhea medications consist of antihistamine, nasal anticholinergics and nasal glucocorticoids.Citation178 These co-medications may contribute to negative side effects. Therefore, physicians should consider dose reduction of AChEIs instead of adding other medications to treat adverse effects of AChEIs.Citation179

Factors Associated with Adverse Drug Reactions of Acetylcholinesterase Inhibitors in Older Adults with Dementia

In geriatric patients with dementia, changes in PK and PD are major risk factors of ADRs. In terms of PK, hepatic and renal functions usually decline in these patients, resulting in decreased drug elimination. Older patients are vulnerable to get an uneventful ADRs from these medications according to their sensitivity to the pharmacodynamic effects.Citation180 Additionally, patients with AD are prone to be sensitive to ADRs as a result of increased BBB permeability and decreased P-gp activity in the brain.Citation43,Citation101,Citation102,Citation181,Citation182 Polypharmacy is common in aging populations and is an important risk factor for drug-related problems (DRPs) such as potentially inappropriate medications (PIMs), drug–drug interactions (DDIs), ADRs and poor compliance.Citation183,Citation184 ADRs derived from DDIs, PIMs or poor compliance are often reported in older patients with dementia.Citation183,Citation184 ADRs are major causes of hospitalization, morbidity and mortality in older people with dementia.Citation185,Citation186

Changes in Pharmacokinetics

PK is what an individual’s body does to a medication after its administration, and refers to absorption, distribution, metabolism and excretion.Citation42Citation47 In geriatric population, the alteration of absorption does not lead to major adverse effects whereas changes in distribution, metabolism and excretion play important roles in clinical outcomes. The alterations of PK and PD of AChEIs among older people living with dementia were presented in and .

Table 1 The Changes in Pharmacokinetics of Acetylcholinesterase Inhibitors Among Older Adults Living with Dementia

Table 2 The Changes in Pharmacokinetics and Pharmacodynamics of Acetylcholinesterase Inhibitors Among Older Adults Living with Dementia

Absorption

Age-related gastrointestinal tract changes often affect the oral absorption. Hypochlorhydria in older adults alleviate the degree of absorption of weakly basic drugs. Furthermore, reduced splanchnic blood flow and gastrointestinal motility as well as delayed gastric emptying time result in longer staying of drug in the gastrointestinal tract and delaying absorption of the drug. Older adults treated with donepezil presented a significant increase in mean Tmax but not in plasma level concentration contributing to slower donepezil’s absorption.Citation86,Citation90 In contrast, the bioavailability and absorption of rivastigmine have no significant change with advancing age.Citation187 Concomitant administration of galantamine with food delays Tmax by 1.5 hours and slows its absorption rate but does not affect the extent of absorption.Citation74 A moderate food effect was found in previous studies of rivastigmine.Citation54 Food slows the absorption of rivastigmine and reduces Tmax by 30%.Citation54,Citation188 Therefore, the coadministration of food and galantamine or rivastigmine is indicated to reduce cholinergic adverse effects such as nausea and vomiting. Conversely, food intake has no significant effect on the absorption of donepezil.Citation54,Citation189

Rivastigmine could also be administered via a skin patch. Age-related changes in skin includes atrophy of epidermis and dermis and decreased blood perfusion, leading to reduced drug absorption via the skin.Citation190 Nevertheless, age-related changes to drug absorption have minimal effects on the pharmacotherapy of dermal medications.

Distribution

Many factors affect volume of distribution. Patients with dementia are more likely to experience malnutrition and frailty as a result of inability to feed by themselves, changes in feeding behaviours, and difficulty with swallowing.Citation191 Changes in the body composition of older adults with dementia also occur including, 10–15% reduction in total body water, 25–30% reduction in muscle mass, and a 25–30% relative increase in body fat.Citation192,Citation193 The aging and frailty processes in this group of patients also contributes to a 10–20% reduction in serum albumin concentration which plays a major role in plasma protein binding.Citation193Citation196 Medication that predominately binds albumin such as donepezil (75% bind to albumin), a reduction in albumin binding may contribute to the rising of unbound fraction being pharmacologically active, resulting in greater potency and toxicity.Citation88,Citation90 Furthermore, donepezil may displace other high-protein binding medications such as warfarin, benzodiazepine and valproate, leading to an increased unbound form of these medications and serious adverse effects. Due to age-related changes, the Vd throughout the whole body of donepezil is substantially increased by approximately 40%, resulting in a prolonged half-life.Citation90,Citation197

Metabolism

Liver CYP enzymes system plays a major role in drug metabolism and may be affected by increasing age. CYP2C19 functions are reduced with age while other isoenzymes show minimal reduction or no change.Citation45 In contrast, there is no significant change in phase II metabolism, especially conjugation in older adults. However, phase II metabolism and downregulation of the transporter pathway of AChEIs are decreased in frail older adults, leading to a greater risk of drug toxicity.Citation198Citation200 The decrease of drug metabolism in the geriatric population, especially in phase I metabolism, results from a 30% and 40% reduction in liver mass and in hepatic blood flow, respectively.Citation201Citation205 The reduction in drug metabolism may account for decreased hepatic clearance, prolonged half-life and increased dose-dependent ADRs. In terms of AChEIs, there are diverse pharmacological properties and differences of clinical outcomes. Data from clinical trials of geriatric patients with AD reveal that the steady-state concentrations of galantamine are 40% higher than those in a healthy younger population as a result of reduced galantamine’s metabolism.Citation88,Citation111,Citation112,Citation138 Based on a population pharmacokinetic analysis, the hepatic clearance of donepezil and of rivastigmine has a tendency to decrease with increasing age.Citation88,Citation89,Citation91,Citation187 Apart from age-related changes in metabolism, most older adults with dementia have multiple chronic diseases including hepatic diseases or cirrhosis, which may lead to decreased hepatic function and drug metabolism. The clearance of both galantamine and rivastigmine was reduced by 25% and 65%, respectively in patients with moderate hepatic impairment (Child-Pugh score of 7–9).Citation88,Citation111,Citation121 Hence, dose adjustment is recommended for these populations. The use of galantamine for such patients should be initiated with a low dose (4 mg per day) and slowly titrated to a maximum daily dose (16 mg per day).Citation88,Citation111,Citation138 However, no data is available on the use of galantamine or rivastigmine in patients with severe hepatic impairment (Child-Pugh score of 10–15).Citation88,Citation111,Citation112,Citation138 Consequently, the use of galantamine or rivastigmine in patients with severe hepatic impairment is contraindicated in clinical practice.Citation138 A recent study showed a 20% reduction in the clearance of donepezil in dementia patients with cirrhosis.Citation206 However, there is no clinically significant alteration in the PK of donepezil in AD patients with moderate or severe hepatic impairment.Citation206,Citation207 This may explain why dose modification of donepezil is not required.

Excretion

After metabolism, most substances are transformed to products that are readily excreted via the kidneys. As a results of age-related physiological changes, the reduction in renal blood flow (50%), renal mass and size (20–30%), and number of nephrons (60%), lead to a decline in drug excretion and drug half-life prolongation.Citation208 Apart from metabolism changes, dosage adjustment should be done based on renal function which is calculated from laboratory measurement (serum creatinine) by using a mathematical equation including the Cockcroft-Gault (CG) formula to ensure proper drug dose for older adults.Citation45,Citation46,Citation209 However, serum creatinine level in older frail individuals may not accurately present renal function because of decreased muscle mass.Citation209 Older AD patients presented a 30% reduction in renal clearance of galantamine, compared with healthy individuals.Citation210 As a consequence of increasing age and frailty, the clearance rate of galantamine, rivastigmine and donepezil in older patients with AD is reduced, compared to healthy individuals.Citation138,Citation210 The clearance of galantamine and rivastigmine is decreased by 25% and 64%, respectively in AD patients with moderate renal impairment.Citation138 This PK alteration may necessitate dose modification and close monitoring to avoid adverse outcomes.Citation138 A total daily dose of galantamine should not exceed 16 mg in patients with moderate renal decline or creatinine clearance 9–59 mL/minCitation138 whereas specific-dose adjustment of rivastigmine is not indicated.Citation211 Nevertheless, the use of galantamine is not recommended given the insufficient data for patients with severe renal impairment or creatinine clearance less than 9 mL/min.Citation138 On the contrary, donepezil disposition is not affected by renal dysfunction. The renal clearance of donepezil in patients with moderate to severe renal impairment has no difference to sex- and age-matched healthy population despite donepezil and its metabolites are mostly excreted by kidneys. In a population pharmacokinetic study of AD patients with moderate to severe renal impairment, there is no clinically significant change of PK or PD parameters of donepezil, compared with healthy population. Therefore, dose adjustments are not necessary in AD patients with renal impairment.Citation64,Citation207

As a result of decreased elimination of rivastigmine, dose adjustments with close monitoring should be done. Nevertheless, no study has been reported for rivastigmine transdermal patches in AD patients with renal or hepatic impairment. Therefore, rivastigmine transdermal patches should be avoided in AD patients with severe renal or hepatic impairment.Citation107,Citation108

Changes in the Blood-Brain Barrier

The BBB is a highly selective semipermeable layer of endothelial cells which limits the access of water-soluble and large molecules transporting from blood circulation into the brain parenchyma. Older adults with dementia have changes in the permeability and integrity of the BBB, as presented in . BBB mechanism includes reabsorption of CSF and efflux pumps for molecules such as p-glycoprotein (P-gp) which assists the maintenance of hemostasis in the brain and in the clearance of beta-amyloid.Citation43,Citation101,Citation102 P-gp is a phosphorylated protein encoded by multidrug resistance gene 1 (MDR1) and belongs to the family of ATP-binding cassette (ABC) membrane transporters.Citation102,Citation212 It is located on the apical surface of endothelial cells and is involved in limiting the transfer of small molecules into the brain.Citation213,Citation214 With aging process and dementia, levels and activity of P-gp have a tendency to decline.Citation101,Citation102,Citation181,Citation215 Furthermore, micro-disruption of the BBB is found in patients with dementia, contributing to increased allowance of some medications across BBB around the disruption areas.Citation181 These changes may lead to increased permission of AChEIs to the brain as a predisposing factor of AChEI-induced ADRs in this population.Citation43

Changes in Pharmacodynamics

By definition, PD is described as what medication does to the body such as receptor binding and chemical interaction.Citation42Citation47 The changes of PD are difficult to predict and evaluate in individuals. In the aging process, the sensitive affinity of receptors for particular medications may change. Moreover, the number of receptor sites may alter and may impact on the efficacy of many medications. The geriatric population is more susceptible to certain central nervous system (CNS) adverse outcomes of AChEIs due to increased permeability of the BBB and decreased P-gp activity.Citation101,Citation102,Citation181,Citation182,Citation215 Furthermore, high sensitivity to cholinergic receptors in the brain and the reduction in homeostasis are found in the older adult population.Citation45,Citation46,Citation216 These alterations result in an elevated responses to AChEIs and contribute to PNS and CNS cholinergic ADRs, as presented in . However, changes in the PD of AChEIs in older patients with dementia have not been extensively explored.

Changes in Pharmacogenetics

Pharmacogenetics is defined as genetic variations in individuals which contribute to different responses to medications. PGx plays a major role in ADRs and therapeutic failures (TFs). Polymorphism of CYP enzymes for AChEIs results in PK and PD difference.Citation84,Citation217 In terms of AChEIs, PGx of encoded gene on P-gp, CYP2D6, and CYP3A4 plays an important role in PK of donepezil and galantamine.Citation218 Interesting studies presented genetic variations of single nucleotide polymorphisms (SNP) in cholinergic markers on AChE and BuChE which have effects on clinical responses to AChEIs as well.Citation82,Citation219 Moreover, polymorphism in the gene encoding choline acetyltransferase (ChAT), acetylcholine biosynthetic enzyme, and a genetic variation of paraoxonase-1 (PON-1) 192Q/R (rs662) which influences the activity of this arylesterase, are involved as the prognostic indicators of response to AChEIs.Citation220,Citation221 Pharmacogenetic considerations for AChEIs should be heeded because they could help predict drug toxicity and efficacy in individuals. In recent decades, genetic polymorphism on CYP2D6 genotype was increasingly studied in various populations.Citation222Citation225 CYP2D6 phenotypes are categorized into four types of metabolizers: Poor metabolizers (PMs), intermediate metabolizers (IMs), extensive metabolizers (EMs), and ultra-rapid metabolizers (UMs). PMs have functional deficiency of CYP2D6 due to mutated allele of CYP2D6. EMs have normal functions of CYP2D6 while UMs have a very low concentration of AChEI owing to multiple copies of CYP2D6 gene. IMs metabolize medications with a rate between PMs and EMs.Citation222,Citation223,Citation225 According to PGx of CYP2D6 (PGX-CYP2D6), approximately 30% of older AD patients have poor metabolite of galantamine and donepezil.Citation226 This situation can be explained by the phenotypic profile of CYP2D6 genotypes being associated with the presence of the APOE-4 allele.Citation227Citation229 Furthermore, the prevalence of each CYP2D6 polymorphism differs according to race and ethnicity.Citation84,Citation230 In Caucasian populations, PMs, IMs, EMs and UMs account for approximately 5–10%, 10–17%, 70–80% and 3–5% of individuals, respectively.Citation231,Citation232 Asians, Africans and African Americans have a greater percentage of reduced-function of CYP2D6 (50%), compared with Caucasians (26%).Citation233 CYP3A4 polymorphism is not responsible for the variation in metabolism of donepezil and galantamine. The effect of genetic variation in ATP-binding cassette sub-family B member 1 (ABCB1) on membrane transporter P-gp plays an important role in donepezil transporters across the BBB and in the clearance of amyloid β (Aβ) peptide related to APOE, ABCB1 gene polymorphisms which have an impact on distribution, excretion, and absorption of donepezil.Citation102,Citation212,Citation234,Citation235

Drug–Drug Interactions

DDI is defined as the pharmacological activities of one drug changed by the concomitant administration of another medication.Citation236 Generally, drug interactions are responsible for 20% to 30% of ADRs. Over 30% of reported ADRs caused by AChEIs result from DDIs.Citation237 The major risk factors for DDIs are polypharmacy and age-related PK and PD changes.Citation238,Citation239 DDIs are classified into two types: PK and PD drug interactions. By definition, PK drug interaction involves one medication altering the absorption, distribution, transport, metabolism or excretion of another medication.Citation240 PD drug interaction is defined as one medication changing the response to another medication.Citation240 CYP enzymes-mediated and transporter-mediated PK drug interactions as well as synergistic or antagonistic PD drug interactions are common DDIs among dementia patients treated with AChEIs.Citation241Citation243 Inducers and inhibitors of CYP2D6 and CYP3A4 enzyme play important roles in the mechanism of PK drug interactions of donepezil and galantamine.Citation226,Citation244 P-gp inducers and inhibitors are involved in transporter-mediated PK drug interactions of donepezil, which is considered a weak P-gp substrate.Citation245

Potent CYP2D6 and CYP3A4 inhibitors such as antidepressants (paroxetine, fluoxetine), and antifungal drugs (ketoconazole) contribute to increased plasma concentration of donepezil and galantamine, as shown in .Citation138,Citation242,Citation246Citation249 The adverse outcomes may be hypercholinergic effects of AChEIs, such as bradycardia, diarrhea and hypersalivation. However, there is no significant CYP2D6 and CYP3A4 inducers of donepezil and galantamine. In terms of transporter-mediated PK drug interactions, PK of donepezil is affected by P-gp inhibitors and inducers. Most medications, which are transported by P-gp, are also metabolized by CYP3A4.Citation214,Citation245,Citation250 Many P-gp inhibitors and inducers are also inhibitors and inducers of CYP3A4. Therefore, many DDIs are associated with inhibition or induction of both CYP3A4 and P-gp.Citation250 The most common P-gp inhibitors in patients with dementia are antibiotics (azithromycin, clarithromycin, erythromycin), cardiovascular medications (carvedilol, verapamil) and antiplatelets (cilostazol, ticagrelor), resulting in the rising of donepezil plasma concentration.Citation250Citation252 There was the clinical report of cardiotoxicity owing to coadministration of donepezil and cilostazol.Citation252 Due to P-gp interaction with cilostazol, the concentration of donepezil in the heart tissue was increased, leading QT prolongation.Citation252 In the case of P-gp inducers, the plasma concentration of donepezil is decreased by carbamazepine, phenobarbital, phenytoin and rifampicin,Citation250Citation252 as presented in .

Table 3 Common CYP Enzymes-Mediated Pharmacokinetic Drug Interactions of Acetylcholinesterase Inhibitors in Older Adults Living with Dementia

Table 4 Common Transporter-Mediated Pharmacokinetic Drug Interactions of Acetylcholinesterase Inhibitors in Older Adults Living with Dementia

Pharmacoepidemiological studies in people with dementia have revealed that anticholinergics, antidepressants, antipsychotics, non-steroidal anti-inflammatory drugs (NSAIDs), and cardiovascular drugs are common co-medications with AChEIs, resulting in PD drug interactions.Citation237,Citation243,Citation253,Citation254 Synergistic PD drug interactions of AChEIs with cholinomimetics or cholinergic agonists have additional cholinergic effects such as hypersalivation, diarrhea, nausea, and vomiting, as presented in .Citation255Citation257 Many antagonistic PD drug interactions of AChEIs are related to changes in PD from advancing age and to dementia processes. In the aging process, a reduction in the number of cholinergic and dopaminergic neurons and dopamine D2 receptors are reported. Therefore, the uses of anticholinergics and antipsychotics which affect cholinergic and dopaminergic neurotransmitters, potentially interfere with the activity of cholinesterase inhibitors and can cause adverse clinical outcomes.Citation253,Citation254,Citation258,Citation259 The clinical report described rigidity, parkinsonism and immobilization in AD patients treated with donepezil and risperidone which these adverse symptoms resolved after risperidone was discontinued.Citation260 Furthermore, concomitant use of beta-blockers, calcium channel blockers or antiarrhythmics in older patients with dementia treated with AChEIs may result in adverse cardiovascular effects such as bradyarrhythmia, heart block, syncope and QT prolongation,Citation63,Citation243,Citation261 as presented in .

Table 5 Pharmacodynamic Interactions in Older Adults with Dementia

Principles for Prescribing Acetylcholinesterase Inhibitors

Recommendations for Prescribing Acetylcholinesterase Inhibitors

AChEI should be initiated at a low efficient dose and titrated slowly upward. The starting dose of donepezil is 5 mg once daily. Donepezil dosage should not be adjusted too quickly because the time to reach the steady state is within 15 days. Therefore, donepezil should be slowly titrated after the first dose is started over 4–6 weeks. Older adults with moderate to severe AD could slowly titrate the donepezil dose to 23 mg per day,Citation262 as presented in Supplementary Table 1. However, gastrointestinal complaints and poor appetite may be reported in patients receiving high donepezil doses.Citation75,Citation139,Citation262,Citation263 Among patients with mild to moderate hepatic insufficiency, a low dose (5 mg daily) consumption of donepezil is safe and the use of its doubling dose should be monitored.Citation264 Galantamine is a daily oral medication ranging from 8 to 24 mg per day,Citation116 as shown in Supplementary Table 1. Galantamine doses must be adjusted for people with moderate hepatic impairment. Furthermore, galantamine should not be recommended to patients with severe liver and kidney dysfunction. Rivastigmine may be a good choice for older demented patients exposed to polypharmacy to reduce the incidence of PK drug interactions related to CYP enzymes. Rivastigmine prescriptions start at 1.5 mg and could be gradually increased to 6 mg twice a day,Citation64,Citation81,Citation103,Citation105 as shown in Supplementary Table 1. Rivastigmine transdermal patches are usually recommended for dementia patients with severe gastrointestinal side effects from oral administration. Transdermal patches have a long half-life and are easily applied only once a day. The dose of rivastigmine in a patch can be titrated from 4.6 mg per 24 hours to 13.3 mg per 24 hours.Citation107Citation109 However, some patients treated with a transdermal patch may develop dermatological side effects such as pruritus or an allergic reaction. Therefore, patients should be examined for adverse skin reactions during treatment.Citation108

Contraindication and Caution in the Prescribing of Acetylcholinesterase Inhibitors

Allergic reaction to the medication itself and chemicals in the same structural group is an absolute contraindication. Donepezil is a piperidine-based compound.Citation81,Citation82,Citation85 Rivastigmine is derived from carbamate compound whereas galantamine is belonged to alkaloid substance.Citation62,Citation81,Citation82,Citation85 Rivastigmine patches are contraindicated for patients with suggestive allergic contact dermatitis.Citation107Citation109 Galantamine is also contraindicated for patients with severe hepatic and renal dysfunction.Citation212 Additionally, patients with sick sinus syndrome (SSS) and second or third heart block should avoid using AChEIs.Citation138,Citation140,Citation141 AChEIs should be used with cautions in people with severe hepatic impairment, severe obstructive pulmonary disease, active gastrointestinal ulcers or bleeding, seizure and significant conduction abnormalities such as supraventricular conduction problems, and arrhythmias.Citation138,Citation140,Citation141 However, older patients treated with AChEIs rarely develop cholinergic crisis in the clinical practice.

Recommendations for Discontinuation of Acetylcholinesterase Inhibitors

The continuous use of AChEIs should be often weighted the risks and the benefits. According to the Food and Drug Administration Adverse Event Reporting System database, serious ADRs related to AChEIs were reported, especially in long-term treatment.Citation135,Citation136,Citation179,Citation213,Citation265,Citation266 Advanced dementia patients may be offered unnecessary treatment that may not provide positive effects, resulting in increased adverse outcomes.Citation267 Therefore, many studies offered recommendations to discontinue AChEI treatment in particular conditions to optimize medication prescribing.Citation268 The common reason for deprescribing AChEIs were lack of response, significantly impaired functional status, severe cognitive impairment (MMSE score < 10), and side effects.Citation268 Besides minimized medication prescribing, the benefits of AChEI discontinuation are improved medication non-adherence, and reduced DDI, medication management burden and cost of medications.Citation269 The discontinuation of AChEIs should be slowly tapered the dose by halving the previous dose and stepping down to the lowest available dose.Citation269 The abrupt cessation should be done in patients with experiencing ADRs. After discontinuation, physicians should closely monitor the withdrawal symptoms and the changes of cognitive function, psycho-behavioral symptoms and functional status.Citation269

Strategies to Prevent Adverse Drug Reactions of Acetylcholinesterase Inhibitors

Many strategies have been developed and implemented to prevent ADRs in patients using AChEIs, as shown in . Minimizing effective dose is required to reduce the occurrence of adverse outcomes. The “start low go slow” strategy is widely recommended as the lowest initial dose, slow-dose titration and close monitoring.Citation270,Citation271 The dose adjustment of AChEIs is recommended according to the alteration of PK or PD.Citation47,Citation270,Citation272Citation275 Furthermore, older patients usually have comorbidities for which multiple medications are taken, resulting in DRPs including potential DDIs, drug–disease interactions, inappropriate medications and medication non-adherence.Citation270,Citation272Citation274,Citation276 Thus, comprehensive medication reviews and optimizing medications prescribing are necessary to address DRPs.Citation275 Another potential strategy could be using tools such as the Micromedex Drug Interaction DatabaseCitation277 and the 2019 American Geriatrics Society Beers criteriaCitation278 to evaluate DDIs and PIMs, respectively.Citation238,Citation279 The discontinuation of AChEIs in older adults with particular circumstances including lack of treatment response, severe cognitive function, significantly impaired functional status, could have reduced DDIs and PIMs.Citation268 Moreover, computerized alert systems for screening prescriptions and flagging DDIs and PIMs could also prevent ADRs.Citation275,Citation280,Citation281 Medication non-adherence is another major DRP in older adults, resulting from language barriers, complex regimens and physiological changes including cognitive impairment, visual and auditory problems and bone-joint deformities.Citation282Citation286 Many techniques could provide benefits to people with medication non-adherence; for example, readily openable containers, clearly written instructions in large print, the simple possible dosage regimens and supporting technology (alarm clock and drug calendar).Citation287,Citation288

Table 6 Prevention Strategies for Adverse Drug Reactions of Acetylcholinesterase Inhibitors

Conclusions

AChEIs have been widely prescribed to delay worsening of cognitive functions and psycho-behavioral problems in older people living with dementia. In the aging population, age-related PK and PD changes, and multiple comorbidities lead to altered pharmacological responses and increased ADRs. Furthermore, geriatric people are more likely to be sensitive to pharmacological toxicity. The most common negative effects of AChEIs are adverse neuropsychiatric, gastrointestinal, and cardiovascular outcomes. Thus, prescribing of AChEIs for dementia treatment should carefully consider both risks and benefits. The discontinuation of AChEIs in older people with particular circumstances such as lack of treatment response, severe cognitive impairment and side effects, could reduce DRPs. Many strategies have been developed to prevent adverse effects. The “start low go slow” strategy as well as comprehensive medication review are highly recommended to address ADRs.

Abbreviation

ABCB1, ATP-binding cassette sub-family B member 1; Aβ, amyloid β; Ach, acetylcholine; AChE, acetylcholinesterase; AChEIs, acetylcholinesterase inhibitors; AD, Alzheimer’s disease; ADRs, adverse drug reactions; AGS Beers Criteria, American Geriatrics Society Beers Criteria; BBB, blood brain barrier; BPSD, behavioral and psychological symptoms; BuChE, butyrylcholinesterase; CG, Cockcroft-Gault; ChAT, choline acetyltransferase; CNS, central nervous system; CSF, cerebrospinal fluid; CYP, cytochrome P450; CYP2D6, cytochrome P450 2D6; CYP3A4, cytochrome P450 3A4; DDIs, drug–drug interactions; DRPs, Drug-related problems; Ems, extensive metabolisers; FDA, Food and Drug Administration; GI, gastrointestinal; IMs, intermediate metabolisers; MDR1, multidrug resistance gene 1; nAChRs, nicotinic acetylcholine receptors; NMDA, N-Methyl-D-aspartate; NSAIDs, non-steroidal anti-inflammatory drugs; PD, pharmacodynamics; P-gp, p-glycoprotein; PIMs, potentially inappropriate medications; PGx, pharmacogenetics; PGx-CYP2D6, pharmacogenetics of CYP2D6; PK, pharmacokinetics; PMs, poor metabolisers; PNS, peripheral nervous system; PON-1, paraoxonase-1; SIADH, syndrome of inappropriate antidiuretic hormone; SJS, Stevens-Johnson Syndrome; SNP, single nucleotide polymorphism; SSS, sick sinus syndrome; TdP, torsades de pointes; TFs, therapeutic failures; Tmax, time to peak plasma concentration; Ums, ultra-rapid metabolisers; Vd, volume of distribution; WAP, wandering atrial pacemaker; 6DD, 6-O-desmethyl donepezil.

Author Contributions

All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; agreed to submit to the current journal; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.

Acknowledgments

The authors would like to thank Leila Shafiee Hanjani, Centre for Health Services Research, Faculty of Medicine, The University of Queensland, for providing valuable advice and comments.

Disclosure

The authors declare no potential conflicts of interest regarding this work.

Additional information

Funding

The authors received no financial support for the research.

References

  • World Health Organization. World report on ageing and health. Geneva: World Health Organization; 2015. Available from http://www.who.int/ageing/events/world-report-2015-launch. Accessed November 22, 2020.
  • Wilson T. An introduction to population projections for Australia.Australian Population Studies.2019;3(1):40–56.
  • Prince MJ, Wimo A, Guerchet MM, Ali GC, Wu Y-T, Prina M. World Alzheimer Report 2015; The Global Impact of Dementia: An analysis of prevalence, incidence, cost and trends. London: Alzheimer's Disease International, 2015. Available from: http://www.alz.co.uk/research/world-report-2015. Accessed November 22, 2020.
  • Ahmadi-Abhari S, Guzman-Castillo M, Bandosz P, et al. Temporal trend in dementia incidence since 2002 and projections for prevalence in England and Wales to 2040: modelling study. BMJ. 2017;358:j2856. doi:10.1136/bmj.j2856
  • World Health Organization. Dementia. Geneva: World Health Organization; 2019. Available from: http://www.who.int/news-room/fact-sheets/detail/dementia. Accessed November 21, 2020.
  • Alzheimer’s Disease International. Dementia Statistics. London: Alzheimer’s Disease International; 2017. Available from: https://www.alz.co.uk/research/statistics. Accessed November 21, 2020.
  • ICD-10. Organic, including symptomatic, mental disorders (F00-F09); 2016. Available from: https://icd.who.int/browse10/2016/en#/F00-F09. Accessed November 24, 2020..
  • Dementia Australia 2018a. Types of Dementia. Australia: Dementia Australia; 2018. Available from: https://www.dementia.org.au/information/about-dementia/types-of-dementia. Accessed December 2, 2020.
  • Dementia Australia 2018c. Alzheimer’s disease. Australia: Dementia Australia; 2018. Available from: https://www.dementia.org.au/about-dementia/types-of-dementia/alzheimer-disease. Accessed December 2, 2020.
  • Alzheimer’s Association. 2016 Alzheimer’s Disease Facts and Figures. Alzheimers Dement. 2016;12(4):459–509.
  • Epperly T, Dunay MA, Boice JL. Alzheimer Disease: pharmacologic and Nonpharmacologic Therapies for Cognitive and Functional Symptoms. Am Fam Physician. 2017;95(12):771–778.
  • Australian Institute of Health and Welfare 2012. Dementia in Australia. Cat. no. AGE 70. Canberra: AIHW; 2012. Available from: https://www.aihw.gov.au/getmedia/13995.pdf.aspx. Accessed November 25, 2020.
  • Australian Bureau of Statistics 2019. Causes of Death, Australia, 2019, Catalogue No. 3303.0, ABS. Canberra: Australian Bureau of Statistics (ABS); 2019. Available from: http://www.abs.gov.au/ausstats/[email protected]/mf/3303.0. Accessed December 2, 2020.
  • Winblad B, Amouyel P, Andrieu S, et al. Defeating Alzheimer’s disease and other dementias: a priority for European science and society. Lancet Neurol. 2016;15(5):455–532.
  • Malone DC, McLaughlin TP, Wahl PM, et al. Burden of Alzheimer’s disease and association with negative health outcomes. Am J Manag Care. 2009;15(8):481–488.
  • Sharma S, Mueller C, Stewart R, et al. Predictors of falls and fractures leading to hospitalization in people with dementia: a representative cohort study. J Am Med Dir Assoc. 2018;19(7):607–612. doi:10.1016/j.jamda.2018.03.009
  • The National Centre for Social and Economic Modelling NATSEM (2016) Economic Cost of Dementia in Australia 2016–2056; 2017 Feb. Available from: http://www.dementia.org.au/files/NATIONAL/documents/The-economic-cost-of-dementia-in-Australia-2016-to-2056.pdf. Accessed November 12, 2020.
  • Dyer SM, Harrison SL, Laver K, et al. An overview of systematic reviews of pharmacological and non‐pharmacological interventions for the treatment of behavioral and psychological symptoms of dementia. Int Psychogeriatr. 2017;30(03):1‐15.
  • Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst Rev. 2006;1:CD005593.
  • O’Brien JT, Holmes C, Jones M, et al. Clinical practice with anti-dementia drugs: a revised (third) consensus statement from the British Association for Psychopharmacology. J Psychopharmacol. 2017;31(2):147–168. doi:10.1177/0269881116680924
  • Rabins PV, Rummans T, Schneider LS, et al. Practice Guideline for the Treatment of Patients with Alzheimer’s Disease and Other Dementias. 2nd ed. USA: American Psychiatric Association; 2014. doi:10.1176/appi.books.9780890423967.152139
  • Australian Institute of Health and Welfare 2019. Dispensing patterns for anti-dementia medications 2016–17. Cat. no. AGE 95. Canberra: AIHW; 2019. Available from: https://www.aihw.gov.au/reports/dementia/dispensing-patterns-for-anti-dementia-medications/contents. Accessed November 20, 2020.
  • Calvó-Perxas L, Turró-Garriga O, Vilalta-Franch J, et al. Trends in the Prescription and Long-Term Utilization of Antidementia Drugs Among Patients with Alzheimer’s Disease in Spain: a Cohort Study Using the Registry of Dementias of Girona. Drugs Aging. 2017;34(4):303–310. doi:10.1007/s40266-017-0446-x
  • Moraes FS, Souza MLC, Lucchetti G, Lucchetti ALG. Trends and disparities in the use of cholinesterase inhibitors to treat Alzheimer’s disease dispensed by the Brazilian public health system - 2008 to 2014: a nation-wide analysis. Arq Neuropsiquiatr. 2018;76(7):444–451. doi:10.1590/0004-282x20180064
  • Pariente A, Helmer C, Merliere Y, Moore N, Fourrier-Réglat A, Dartigues JF. Prevalence of cholinesterase inhibitors in subjects with dementia in Europe. Pharmacoepidemiol Drug Saf. 2008;17(7):655–660. doi:10.1002/pds.1613
  • Clague F, Mercer SW, McLean G, Reynish E, Guthrie B. Comorbidity and polypharmacy in people with dementia: insights from a large, population-based cross-sectional analysis of primary care data. Age Ageing. 2017;46(1):33–39.
  • Parsons C. Polypharmacy and inappropriate medication use in patients with dementia: an underresearched problem. Ther Adv Drug Saf. 2017;8(1):31–46. doi:10.1177/2042098616670798
  • Hoffmann F, van den Bussche H, Wiese B, et al. Impact of geriatric comorbidity and polypharmacy on cholinesterase inhibitors prescribing in dementia. BMC Psychiatry. 2011;11:190. doi:10.1186/1471-244X-11-190
  • Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350(mar02 7):h369. doi:10.1136/bmj.h369
  • Masopust J, Protopopová D, Vališ M, et al. Treatment of behavioral and psychological symptoms of dementias with psychopharmaceuticals: a review. Neuropsychiatr Dis Treat. 2018;14:1211–1220. doi:10.2147/NDT.S163842
  • Gabryelewicz T. Pharmacological treatment of behavioral symptoms in dementia patients. Przegl Lek. 2014;71(4):215–220.
  • Andersen F, Viitanen M, Halvorsen DS, Straume B, Engstad TA. Co-morbidity and drug treatment in Alzheimer’s disease. A cross sectional study of participants in the dementia study in northern Norway. BMC Geriatr. 2011;11:58. doi:10.1186/1471-2318-11-58
  • Fereshtehnejad SM, Johnell K, Eriksdotter M. Anti-dementia drugs and co-medication among patients with Alzheimer’s disease: investigating real-world drug use in clinical practice using the Swedish Dementia Quality Registry (SveDem). Drugs Aging. 2014;31(3):215–224. doi:10.1007/s40266-014-0154-8
  • Crugel M, Paton G, Singh P, et al. Antipsychotics in people with dementia: frequency of use and rationale for prescribing in a UK mental health service. Psychiatrist. 2012;36(5):165–169. doi:10.1192/pb.bp.111.034579
  • Seitz DP, Adunuri N, Gill SS, Gruneir A, Herrmann N, Rochon P. Antidepressants for agitation and psychosis in dementia. Cochrane Database Syst Rev. 2011;2:CD008191.
  • Kable A, Fullerton A, Fraser S, et al. Comparison of potentially inappropriate medications for people with dementia at admission and discharge during an unplanned admission to hospital: results from the SMS dementia study. Healthcare (Basel). 2019;7(1):8. doi:10.3390/healthcare7010008
  • Molist-Brunet N, Sevilla-Sa´nchez D, Ambla´s-Novellas J, et al. Optimizing drug therapy in patients with advanced dementia: a patient-centered approach. Eur Geriatr Med. 2014;5(1):66–71. doi:10.1016/j.eurger.2013.10.011
  • von Renteln-kruse W, Neumann L, Klugmann B, et al. Geriatric patients with cognitive impairment: patient characteristics and treatment results on a specialized ward. Dtsch Arztebl Int. 2015;112(7):103–112.
  • Maher RL Jr, Hanlon JT, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13(1):57–65. doi:10.1517/14740338.2013.827660
  • Rodrigues MC, Oliveira C. Drug-drug interactions and adverse drug reactions in polypharmacy among older adults: an integrative review. Rev Lat Am Enfermagem. 2016;24:e2800. doi:10.1590/1518-8345.1316.2800
  • Wastesson JW, Morin L, Tan ECK, Johnell K. An update on the clinical consequences of polypharmacy in older adults: a narrative review. Expert Opin Drug Saf. 2018;17(12):1185–1196. doi:10.1080/14740338.2018.1546841
  • Reeve E, Trenaman SC, Rockwood K, Hilmer SN. Pharmacokinetic and pharmacodynamic alterations in older people with dementia. Expert Opin Drug Metab Toxicol. 2017;13(6):651–668. doi:10.1080/17425255.2017.1325873
  • Mehta DC, Short JL, Hilmer SN, Nicolazzo JA. Drug access to the central nervous system in Alzheimer’s disease: preclinical and clinical insights. Pharm Res. 2015;32(3):819–839.
  • Massoud L, Agha HA, Taleb M. Pharmacokinetic and pharmacodynamic changes in elderly people. World j Pharm Med. 2017;3(11):14–23.
  • Hutchison LC, O’Brien CE. Changes in pharmacokinetics and pharmacodynamics in the elderly patient. J Pharm Prac. 2007;20:4–12. doi:10.1177/0897190007304657
  • Mangoni AA, Jackson SH. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14. doi:10.1046/j.1365-2125.2003.02007.x
  • Wooten JM. Pharmacotherapy considerations in elderly adults. South Med J. 2012;105(8):437–445. doi:10.1097/SMJ.0b013e31825fed90
  • Kröger E, Mouls M, Wilchesky M, et al. Adverse Drug Reactions Reported with Cholinesterase Inhibitors: an Analysis of 16 Years of Individual Case Safety Reports from VigiBase. Ann Pharmacother. 2015;49(11):1197–1206. doi:10.1177/1060028015602274
  • Pariente A, Sanctussy DJ, Miremont-Salamé G, et al. Factors associated with serious adverse reactions to cholinesterase inhibitors: a study of spontaneous reporting. CNS Drugs. 2010;24(1):55–63. doi:10.2165/11530300-000000000-00000
  • Ali TB, Schleret TR, Reilly BM, Chen WY, Abagyan R. Adverse Effects of Cholinesterase Inhibitors in Dementia, According to the Pharmacovigilance Databases of the United-States and Canada. PLoS One. 2015;10(12):e0144337.
  • Silva T, Reis J, Teixeira J, Borges F. Alzheimer’s disease, enzyme targets and drug discovery struggles: from natural products to drug prototypes. Ageing Res Rev. 2014;15:116–145.
  • Guillozet AL, Smiley JF, Mash DC, Mesulam MM. Butyrylcholinesterase in the life cycle of amyloid plaques. Ann Neurol. 1997;42(6):909–918. doi:10.1002/ana.410420613
  • Giacobini E. Selective inhibitors of butyrylcholinesterase: a valid alternative for therapy of Alzheimer’s disease? Drugs Aging. 2001;18(12):891–898. doi:10.2165/00002512-200118120-00001
  • Weinstock M. Selectivity of cholinesterase inhibition: clinical implications for the treatment of Alzheimer’s disease. CNS Drugs. 1999;12(4):307–323. doi:10.2165/00023210-199912040-00005
  • Mesulam MM, Geula C. Butyrylcholinesterase reactivity differentiates the amyloid plaques of aging from those of dementia. Ann Neurol. 1994;36(5):722–727. doi:10.1002/ana.410360506
  • Op Den Velde W, Stam FC. Some cerebral proteins and enzyme systems in Alzheimer’s presenile and senile dementia. J Am Geriatr Soc. 1976;24(1):12–16. doi:10.1111/j.1532-5415.1976.tb03247.x
  • Perry EK, Perry RH, Blessed G, Tomlinson BE. Changes in brain cholinesterases in senile dementia of Alzheimer type. Neuropathol Appl Neurobiol. 1978;4(4):273–277. doi:10.1111/j.1365-2990.1978.tb00545.x
  • Geula C, Darvesh S. Butyrylcholinesterase, cholinergic neurotransmission and the pathology of Alzheimer’s disease. Drugs Today (Barc). 2004;40(8):711–721. doi:10.1358/dot.2004.40.8.850473
  • Arendt T, Bruckner MK, Lange M, Bigl V. Changes in acetylcholinesterase and butyrylcholinesterase in Alzheimer’s disease resemble embryonic development – a study of molecular forms. Neurochem Int. 1992;21(3):381–396. doi:10.1016/0197-0186(92)90189-X
  • Davies P. Neurotransmitter-related enzymes in senile dementia of the Alzheimer type. Brain Res. 1979;171(2):319–327. doi:10.1016/0006-8993(79)90336-6
  • Wright CI, Geula C, Mesulam MM. Neurological cholinesterases in the normal brain and in Alzheimer’s disease: relationship to plaques, tangles, and patterns of selective vulnerability. Ann Neurol. 1993;34(3):373–384. doi:10.1002/ana.410340312
  • Maelicke A. Allosteric modulation of nicotinic receptors as a treatment strategy for Alzheimer’s disease. Dement Geriatr Cogn Disord. 2000;11(Suppl 1):11–18. doi:10.1159/000051227
  • Gauthier S. Cholinergic adverse effects of cholinesterase inhibitors in Alzheimer’s disease: epidemiology and management. Drugs Aging. 2001;18(11):853–862. doi:10.2165/00002512-200118110-00006
  • Thompson S, Lanctôt KL, Herrmann N. The benefits and risks associated with cholinesterase inhibitor therapy in Alzheimer’s disease. Expert Opin Drug Saf. 2004;3(5):425–440.
  • Patel M, Joshi A, Suthar J, Desai S. Drug utilization pattern in patients with different types of dementia in Western India. Int J Alzheimers Dis. 2014;2014:435202.
  • Li Q, He S, Chen Y, et al. Donepezil-based multi-functional cholinesterase inhibitors for treatment of Alzheimer’s disease. Eur J Med Chem. 2018;158:463–477. doi:10.1016/j.ejmech.2018.09.031
  • Cheewakriengkrai L, Gauthier S. A 10-year perspective on donepezil. Expert Opin Pharmacother. 2013;14(3):331–338. doi:10.1517/14656566.2013.760543
  • FDA approved drug products. Available from: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm. Accessed 19 November 2020.
  • Howard R, McShane R, Lindesay J, et al. Donepezil and memantine for moderate-to-severe Alzheimer’s disease. N Engl J Med. 2012;366(10):893–903. doi:10.1056/NEJMoa1106668
  • Rodda J, Carter J. Cholinesterase inhibitors and memantine for symptomatic treatment of dementia. BMJ. 2012;344:e2986. doi:10.1136/bmj.e2986
  • Birks JS, Grimley EJ. Rivastigmine for Alzheimer’s disease. Cochrane Database Syst Rev. 2015;4:CD001191.
  • Tan CC, Yu JT, Wang HF, et al. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2014;41(2):615–631. doi:10.3233/JAD-132690
  • Di Santo SG, Prinelli F, Adorni F, et al. A meta-analysis of the efficacy of donepezil, rivastigmine, galantamine, and memantine in relation to severity of Alzheimer’s disease. J Alzheimers Dis. 2013;35(2):349–361. doi:10.3233/JAD-122140
  • Tsoi KK, Chan JY, Chan FC, et al. Monotherapy is good enough for patients with mild-to moderate Alzheimer’s disease: a network meta-analysis of 76 randomized controlled trials. Clin Pharmacol Ther. 2019;105(1):121–130. doi:10.1002/cpt.1104
  • Deardorff WJ, Feen E, Grossberg GT. The Use of Cholinesterase Inhibitors Across All Stages of Alzheimer’s Disease. Drugs Aging. 2015;32(7):537–547. doi:10.1007/s40266-015-0273-x
  • Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high-dose (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24 week, randomized, double-blind study. Clin Ther. 2010;32(7):1234–1251. doi:10.1016/j.clinthera.2010.06.019
  • Sabbagh M, Cummings J, Christensen D, et al. Evaluating the cognitive effects of donepezil 23 mg/ d in moderate and severe Alzheimer’s disease: analysis of effects of baseline features on treatment response. BMC Geriatr. 2013;13:56. doi:10.1186/1471-2318-13-56
  • Molinuevo JL, Frölich L, Grossberg GT, et al. Responder analysis of a randomized comparison of the 13.3 mg/24 h and 9.5 mg/24 h rivastigmine patch. Alzheimers Res Ther. 2015;7(1):9. doi:10.1186/s13195-014-0088-8
  • Farlow MR, Grossberg GT, Sadowsky CH, Meng X, Somogyi MA. 24-week, randomized, controlled trial of rivastigmine patch 13.3 mg/24 h versus 4.6 mg/24 h in severe Alzheimer’s dementia. CNS Neurosci Ther. 2013;19(10):745–752. doi:10.1111/cns.12158
  • Farlow MR, Grossberg G, Gauthier S, Meng X, Olin JT. The ACTION study: methodology of a trial to evaluate safety and efficacy of a higher dose rivastigmine transdermal patch in severe Alzheimer’s disease. Curr Med Res Opin. 2010;26(10):2441–2447. doi:10.1185/03007995.2010.513849
  • Colović MB, Krstić DZ, Lazarević-Pašti TD, Bondžić AM, Vasić VM. Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol. 2013;11(3):315–335.
  • Noetzli M, Eap CB. Pharmacodynamic, pharmacokinetic and pharmacogenetic aspects of drugs used in the treatment of Alzheimer’s disease. Clin Pharmacokinet. 2013;52(4):225–241. doi:10.1007/s40262-013-0038-9
  • Campos C, Rocha NB, Vieira RT, et al. Treatment of Cognitive Deficits in Alzheimer’s disease: a psychopharmacological review. Psychiatr Danub. 2016;28(1):2–12.
  • Cacabelos R. Pharmacogenomics and therapeutic prospects in Alzheimer’s disease. Expert Opin Pharmacother. 2005;6(12):1967–1987. doi:10.1517/14656566.6.12.1967
  • Lane RM, Potkin SG, Enz A. Targeting acetylcholinesterase and butyrylcholinesterase in dementia. Int J Neuropsychopharmacol. 2006;9(1):101–124. doi:10.1017/S1461145705005833
  • Heydorn WE. Donepezil (E2020): a new acetylcholinesterase inhibitor. Review of its pharmacology, pharmacokinetics, and utility in the treatment of Alzheimer’s disease. Expert Opin Investig Drugs. 1997;6(10):1527–1535. doi:10.1517/13543784.6.10.1527
  • Kosasa T, Kuriya Y, Matsui K, Yamanishi Y. Inhibitory effects of donepezil hydrochloride (E2020) on cholinesterase activity in brain and peripheral tissues of young and aged rats. Eur J Pharmacol. 1999;386(1):7–13. doi:10.1016/S0014-2999(99)00741-4
  • Gauthier S. Acetylcholinesterase inhibitors in the treatment of Alzheimer’s disease. Exp Opin Invest Drugs. 1999;8:1511–1520. doi:10.1517/13543784.8.10.1511
  • Tiseo PJ, Rogers SL, Friedhoff LT. Pharmacokinetic and pharmacodynamic profile of donepezil HCl following evening administration. Br J Clin Pharmacol. 1998;46(Suppl 1):13–18. doi:10.1046/j.1365-2125.1998.0460s1013.x
  • Ohnishi A, Mihara M, Kamakura H, et al. Comparison of the pharmacokinetics of E2020, a new compound for Alzheimer’s disease, in healthy young and elderly subjects. J Clin Pharmacol. 1993;33(11):1086–1091. doi:10.1002/j.1552-4604.1993.tb01945.x
  • Coin A, Pamio MV, Alexopoulos C, et al. Donepezil plasma concentrations, CYP2D6 and CYP3A4 phenotypes, and cognitive outcome in Alzheimer’s disease. Eur J Clin Pharmacol. 2016;72(6):711–717. doi:10.1007/s00228-016-2033-1
  • Riyath AA, Ammar BB, Faris TA. In vitro kinetic study of donepezil N-oxide metabolites. Irq J Pharm. 2011;11:1–9.
  • Winblad B, Engedal K, Soininen H, et al. Donepezil Nordic Study Group. A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology. 2001;57(3):489–495. doi:10.1212/WNL.57.3.489
  • Mohs RC, Doody RS, Morris JC, et al.; “312” Study Group. A 1-year, placebo-controlled preservation of function survival study of donepezil in AD patients. Neurology. 2001;57(3):481–488. doi:10.1212/WNL.57.3.481
  • Rogers SL, Farlow MR, Doody RS, et al.; Donepezil Study Group. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer’s disease. Neurology. 50;1998:136–145. doi:10.1212/WNL.50.1.136
  • Potocnik F. Dementia. S Afr J Psychiatr. 2013;19(3):141–152.
  • Jann MW. Rivastigmine, a new generation cholinesterase inhibitor for the treatment of Alzheimer’s disease. Pharmacotherapy. 2000;20(1):1–12. doi:10.1592/phco.20.1.1.34664
  • Hossain M, Jhee SS, Shiovitz T, et al. Estimation of the absolute bioavailability of rivastigmine in patients with mild to moderate dementia of the Alzheimer’s type. Clin Pharmacokinet. 2002;41(3):225–234. doi:10.2165/00003088-200241030-00006
  • Obburu JV, Tammara V, Lesko L, et al. Pharmacokinetic, pharmacodynamic modeling of rivastigmine, a cholinesterase inhibitor, in patients with Alzheimer’s disease. J Clin Pharmacol. 2001;41(10):1082–1090. doi:10.1177/00912700122012689
  • Spencer CM, Noble S. Rivastigmine: a review of its use in Alzheimer’s disease. Drugs Aging. 1998;13(5):391–411. doi:10.2165/00002512-199813050-00005
  • Banks WA. Drug delivery to the brain in Alzheimer’s disease: consideration of the blood-brain barrier. Adv Drug Deliv Rev. 2012;64(7):629–639. doi:10.1016/j.addr.2011.12.005
  • van Assema DM, Lubberink M, Bauer M, et al. Blood-brain barrier P-glycoprotein function in Alzheimer’s disease. Brain. 2012;135(Pt 1):181–189. doi:10.1093/brain/awr298
  • Rösler M, Anand R, Cicin-Sain A, et al. Efficacy and safety of rivastigmine in patients with Alzheimer’s disease: international randomised controlled trial. BMJ. 1999;318(7184):633–638. doi:10.1136/bmj.318.7184.633
  • Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379–397. doi:10.7326/0003-4819-148-5-200803040-00009
  • Farlow MR. Update on rivastigmine. Neurologist. 2003;9(5):230–234. doi:10.1097/01.nrl.0000087724.73783.5f
  • Isik AT, Bozoglu E, Eker D. aChE and BuChE inhibition by rivastigmin have no effect on peripheral insulin resistance in elderly patients with Alzheimer disease. J Nutr Health Aging. 2012;16(2):139–141. doi:10.1007/s12603-011-0095-4
  • Greenspoon J, Herrmann N, Adam DN. Transdermal rivastigmine: management of cutaneous adverse events and review of the literature. CNS Drugs. 2011;25(7):575–583. doi:10.2165/11592230-000000000-00000
  • Amanatkar HR, Grossberg GT. Transdermal rivastigmine in the treatment of Alzheimer’s disease: current and future directions. Expert Rev Neurother. 2014;14(10):1119–1125. doi:10.1586/14737175.2014.955852
  • Lefevre G, Sedek G, Jhee SS, et al. Pharmacokinetics and pharmacodynamics of the novel Daily rivastigmine transdermal patch compared with twice-daily capsules in Alzheimer’s disease patients. Clin Pharmacol Ther. 2008;83(1):106–114. doi:10.1038/sj.clpt.6100242
  • Sramek JJ, Frackiewicz EJ, Cutler NR. Review of the acetylcholinesterase inhibitor galantamine. Expert Opin Investig Drugs. 2000;9(10):2393–2402. doi:10.1517/13543784.9.10.2393
  • Bickel U, Thomsen T, Weber W, et al. Pharmacokinetics of galanthamine in humans and corresponding cholinesterase inhibition. Clin Pharmacol Ther. 1991;50(4):420–428. doi:10.1038/clpt.1991.159
  • Bachus R, Bickel U, Thomsen T, Roots I, Kewitz H. The O-demethylation of the antidementia drug galanthamine is catalysed by cytochrome P450 2D6. Pharmacogenetics. 1999;9:661–668. doi:10.1097/00008571-199912000-00001
  • Kewitz H. Pharmacokinetics and metabolism of galanthamine. Drugs Today. 1997;33:265–272. doi:10.1358/dot.1997.33.4.425052
  • Raskind MA, Peskind ER, Wessel T, Yuan W; The Galantamine USA-1 Study Group. Galantamine in [Alzheimer’s disease]. A 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology. 2000;54:2261–2268. doi:10.1212/WNL.54.12.2261
  • Zhao Q, Brett M, Van ON, et al. Galantamine pharmacokinetics, safety, and tolerability profiles are similar in healthy Caucasian and Japanese subjects. J Clin Pharmacol. 2002;42(9):1002–1010. doi:10.1177/0091270002042009007
  • Tayeb HO, Yang HD, Price BH, Tarazi FI. Pharmacotherapies for Alzheimer’s disease: beyond cholinesterase inhibitors. Pharmacol Ther. 2012;134(1):8–25.
  • Buckley JS, Salpeter SR. A Risk-Benefit Assessment of Dementia Medications: systematic Review of the Evidence. Drugs Aging. 2015;32(6):453–467. doi:10.1007/s40266-015-0266-9
  • O’Bryant SE, Humphreys JD, Smith GE, et al. Detecting dementia with the mini-mental state examination in highly educated individuals. Arch Neurol. 2008;65(7):963–967. doi:10.1001/archneur.65.7.963
  • Doraiswamy PM, Kaiser L, Bieber F, et al. The Alzheimer’s Disease Assessment Scale: evaluation of psychometric properties and patterns of cognitive decline in multicenter clinical trials of mild to moderate Alzheimer’s disease. Alzheimer Dis Assoc Disord. 2001;15(4):174–183. doi:10.1097/00002093-200110000-00003
  • Panisset M, Roudier M, Saxton J, et al. Severe impairment battery. A neuropsychological test for severely demented patients. Arch Neurol. 1994;51(1):41–45. doi:10.1001/archneur.1994.00540130067012
  • Collin C, Wade DT, Davies S, Horne V. The Barthel ADL Index: a reliability study. Int Disabil Stud. 1988;10(2):61–63. doi:10.3109/09638288809164103
  • Wade DT, Collin C. The Barthel ADL Index: a standard measure of physical disability? Int Disabil Stud. 1988;10(2):64–67. doi:10.3109/09638288809164105
  • Lawton MP, Brody EM. Assessment of older people: self-maintaining and instrumental activities of daily living. Gerontologist. 1969;9(3):179–186. doi:10.1093/geront/9.3_Part_1.179
  • Cummings JL, Mega M, Gray K, et al. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994;44(12):2308–2314. doi:10.1212/WNL.44.12.2308
  • Qaseem A, Snow V, Cross JT, et al. Current pharmacologic treatment of dementia: a clinical practice guideline from the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med. 2008;148(5):370–378. doi:10.7326/0003-4819-148-5-200803040-00008
  • Santaguida PS, Raina P, Booker L, et al. Pharmacological treatment of dementia. Evid Rep Technol Assess (Summ). 2004;97:1–16.
  • Tricco AC, Ashoor HM, Soobiah C, et al. Comparative Effectiveness and Safety of Cognitive Enhancers for Treating Alzheimer’s Disease: systematic Review and Network Metaanalysis. J Am Geriatr Soc. 2018;66(1):170–178. doi:10.1111/jgs.15069
  • Trinh NH, Hoblyn J, Mohanty S, et al. Efficacy of cholinesterase inhibitors in the treatment of neuropsychiatric symptoms and functional impairment in Alzheimer disease: a meta- analysis. JAMA. 2003;289(2):210–216. doi:10.1001/jama.289.2.210
  • Isik AT, Soysal P, Usarel C. Effects of Acetylcholinesterase Inhibitors on Balance and Gait Functions and Orthostatic Hypotension in Elderly Patients With Alzheimer Disease. Am J Alzheimers Dis Other Demen. 2016;31(7):580–584. doi:10.1177/1533317516666195
  • Winblad B, Kilander L, Eriksson S, et al. Donepezil in patients with severe Alzheimer’s disease: double-blind, parallel-group, placebo-controlled study. Lancet. 2006;367(9516):1057–1065. doi:10.1016/S0140-6736(06)68350-5
  • Froelich L, Andreasen N, Tsolaki M, et al. Long-term treatment of patients with Alzheimer’s disease in primary and secondary care: results from an international survey. Curr Med Res Opin. 2009;25(12):3059–3068. doi:10.1185/03007990903396626
  • Lockhart IA, Mitchell SA, Kelly S. Safety and tolerability of donepezil, rivastigmine and galantamine for patients with Alzheimer’s disease: systematic review of the “real-world” evidence. Dement Geriatr Cogn Disord. 2009;28:389–403. doi:10.1159/000255578
  • Hansen RA, Gartlehner G, Webb AP, Morgan LC, Moore CG, Jonas DE. Efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. Clin Interv Aging. 2008;3:211–225.
  • Kröger E, Van Marum R, Souverein P, Carmichael PH, Egberts T. Treatment with rivastigmine or galantamine and risk of urinary incontinence: results from a Dutch database study. Pharmacoepidemiol Drug Saf. 2015;24:276–285. doi:10.1002/pds.3741
  • Colovic MB, Krstic DZ, Lazarevic-Pasti TD, Bondzic AM, Vasic VM. Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol. 2013;11:315–335. doi:10.2174/1570159X11311030006
  • Galligan JJ, Burks TF. Cholinergic neurons mediate intestinal propulsion in the rat. J Pharm Exper Ther. 1986;238(2):594–598.
  • Lewin MJ. Cellular mechanisms and inhibitors of gastric acid secretion. Drugs Today (Barc). 1999;35(10):743–752. doi:10.1358/dot.1999.35.10.561693
  • Full prescription information. Razadyne (galantamine). Available from: https://www.janssenmd.com/pdf/razadyne/PI-Razadyne-RazadyneER.pdf. Assessed November 22, 2020.
  • Soysal P, Isik AT, Stubbs B, et al. Acetylcholinesterase inhibitors are associated with weight loss in older people with dementia: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2016;87(12):1368–1374. doi:10.1136/jnnp-2016-313660
  • Full prescription information. Aricept (donepezil). Available from: https://www.labeling.pfizer.com/ShowLabeling.aspx?id=510. Accessed November 22, 2020.
  • Full prescription information. Exelon (rivastigmine). Available from: https://www.pharma.us.novartis.com/product/pi/pdf/exelon.pdf. Accessed November 22, 2020.
  • Cutler NR, Anand R, Hartman RD, et al. Antiemetic therapy for Alzheimer’s patients receiving the cholinesterase inhibitor SDZ ENA 713 [abstract]. Clin Pharmacol Ther. 1998;63(2):188.
  • Kistler PM, Sanders P, Fynn SP, et al. Electrophysiologic and electroanatomic changes in the human atrium associated with age. J Am Coll Cardiol. 2004;44(1):109–116. doi:10.1016/j.jacc.2004.03.044
  • Jones SA, Lancaster MK, Boyett MR. Ageing-related changes of connexins and conduction within the sinoatrial node. J Physiol (Lond). 2004;560(Pt 2):429–437. doi:10.1113/jphysiol.2004.072108
  • Jane Newby V, Anne Kenny R, McKeith IG. Donepezil and cardiac syncope: case report. Int J Geriatr Psychiatry. 2004;19(11):1110–1112. doi:10.1002/gps.1183
  • Shahani L. Donepezil-associated sick sinus syndrome. J Neuropsychiatry Clin Neurosci. 2014;26(1):E5. doi:10.1176/appi.neuropsych.12110270
  • Park-Wyllie LY, Mamdani MM, Li P, Gill SS, Laupacis A, Juurlink DN. Cholinesterase inhibitors and hospitalization for bradycardia: a population-based study. PLoS Med. 2009;6(9):e1000157. doi:10.1371/journal.pmed.1000157
  • Tanaka A, Koga S, Hiramatsu Y. Donepezil-induced adverse side effects of cardiac rhythm: 2 cases report of atrioventricular block and Torsade de Pointes. Intern Med. 2009;48(14):1219–1223. doi:10.2169/internalmedicine.48.2181
  • Takaya T, Okamoto M, Yodoi K, et al. Torsades de Pointes with QT prolongation related to donepezil use. J Cardiol. 2009;54(3):507–511. doi:10.1016/j.jjcc.2009.03.011
  • Hadano Y, Ogawa H, Wakeyama T, et al. Donepezil-induced torsades de pointes without QT prolongation. J Cardiol Cases. 2013;8(2):e69–e71. doi:10.1016/j.jccase.2013.05.004
  • Celik IE, Acar B, Çay S. An unusual cardiovascular adverse effect of donepezil. Intern Med J. 2015;45(8):877–878. doi:10.1111/imj.12827
  • Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern Med. 2009;169(9):867–873. doi:10.1001/archinternmed.2009.43
  • Kim DH, Brown RT, Ding EL, Kiel DP, Berry SD. Dementia medications and risk of falls, syncope, and related adverse events meta-analysis of randomized controlled trials. J Am Geriatr Soc. 2011;59(6):1019–1031. doi:10.1111/j.1532-5415.2011.03450.x
  • Hernandez RK, Farwell W, Cantor MD, Lawler EV. Cholinesterase inhibitors and hospitalizations for bradycardia. J Am Geriatr Soc. 2009;57:1997–2003. doi:10.1111/j.1532-5415.2009.02488.x
  • Isik AT, Yildiz GB, Bozoglu E, et al. Cardiac safety of donepezil in elderly patients with Alzheimer disease. Intern Med. 2012;51(6):575–578. doi:10.2169/internalmedicine.51.6671
  • Isik AT, Soysal P, Yay A. Which rivastigmine formula is better for heart in elderly patients with Alzheimer’s disease: oral or patch? Am J Alzheimers Dis Other Demen. 2014;29(8):735–738. doi:10.1177/1533317514536598
  • Isik AT, Bozoglu E, Yay A, et al. Which cholinesterase inhibitor is the safest for the heart elderly patients with Alzheimer’s disease? Am J Alzheimers Dis Other Demen. 2012;27(3):171–174. doi:10.1177/1533317512442999
  • Isik AT, Soysal P, Stubbs B, et al. Cardiovascular Outcomes of Cholinesterase Inhibitors in Individuals with Dementia: a Meta-Analysis and Systematic Review. J Am Geriatr Soc. 2018;66(9):1805–1811. doi:10.1111/jgs.15415
  • Rowland JP, Rigby J, Harper AC, et al. Cardiovascular monitoring with acetylcholinesterase inhibitors: a clinical protocol. Adv Psychiatr Treat. 2007;13:178–184. doi:10.1192/apt.bp.106.002725
  • Babic T, Zurak N. Convulsions induced by donepezil. J Neurol Neurosurg Psychiatry. 1999;66(3):410. doi:10.1136/jnnp.66.3.410
  • Shareef J, Joseph J, Adithi K. A single case report on hyponatremia seizure induced by acetylcholinesterase inhibitors. Int J Pharm Pharm Sci. 2017;9(7):165–166. doi:10.22159/ijpps.2017v9i7.19057
  • Ross JS, Shua-Haim JR. Aricept-induced nightmares in Alzheimer’s disease: 2 case reports. J Am Geriatr Soc. 1998;46(1):119–120. doi:10.1111/j.1532-5415.1998.tb01033.x
  • Bouman WP, Pinner G. Violent behavior associated with donepezil. Am J Psychiatry. 1998;155(11):1626–1627. doi:10.1176/ajp.155.11.1626a
  • Yorston GA, Gray R. Hypnopompic hallucinations with donepezil. J Psychopharmacol. 2000;14(3):303–304. doi:10.1177/026988110001400315
  • Wong CW. Pharmacotherapy for Dementia: a Practical Approach to the Use of Cholinesterase Inhibitors and Memantine. Drugs Aging. 2016;33(7):451–460. doi:10.1007/s40266-016-0372-3
  • Hashimoto M, Imamura T, Tanimukai S, Kazui H, Mori E. Urinary incontinence: an unrecognised adverse effect with donepezil. Lancet. 2000;356(9229):568. doi:10.1016/S0140-6736(00)02588-5
  • Ale I, Lachapelle JM, Maibach HI. Skin tolerability associated with transdermal drug delivery systems: an overview. Adv Ther. 2009;26(10):920–935. doi:10.1007/s12325-009-0075-9
  • Wohlrab J, Kreft B, Tamke B. Skin tolerability of transdermal patches. Expert Opin Drug Deliv. 2011;8(7):939–948. doi:10.1517/17425247.2011.574689
  • Osada T, Watanabe N, Asano N, Adachi Y, Yamamura K. Adverse drug events affecting medication persistence with rivastigmine patch application. Patient Prefer Adherence. 2018;12:1247–1252. doi:10.2147/PPA.S166680
  • Zannas AS, Okuno Y, Doraiswamy PM. Cholinesterase inhibitors and Pisa syndrome: a pharmacovigilance study. Pharmacotherapy. 2014;34(3):272–278. doi:10.1002/phar.1359
  • Hsu CW, Lee Y, Lee CY, Lin PY. Reversible Pisa Syndrome Induced by Rivastigmine in a Patient With Early-Onset Alzheimer Disease. Clin Neuropharmacol. 2017;40(3):147–148. doi:10.1097/WNF.0000000000000215
  • Miyaoka T, Seno H, Yamamori C, et al. Pisa syndrome due to a cholinesterase inhibitor (donepezil): a case report. J Clin Psychiatry. 2001;62(7):573–574. doi:10.4088/JCP.v62n07d13
  • Fleet JL, McArthur E, Patel A, et al. Risk of rhabdomyolysis with donepezil compared with rivastigmine or galantamine: a population-based cohort study. CMAJ. 2019;191(37):e1018- e1024. doi:10.1503/cmaj.190337
  • Health Canada. Summary Safety Review - ARICEPT (donepezil) - Risk of Rhabdomyolysis and Neuroleptic Malignant Syndrome [Internet]. Drug Heal Prod. 2015. Available from. http://www.hc-sc.gc.ca/dhp-mps/medeff/reviews-examens/aricept-eng.php#fnb1
  • Ekinci F, Soyaltin UE, Ugur MC, Develi A, Akar H. A rare case of Rhabdomyolysis probably due to donepezil. J Clin Case Rep. 2014;4:465.
  • Verrico MM, Nace DA, Towers AL. Fulminant chemical hepatitis possibly associated with donepezil and sertraline therapy. J Am Geriatr Soc. 2000;48:1659–1663. doi:10.1111/j.1532-5415.2000.tb03879.x
  • Trenaman SC, Bowles SK, Kirkland S, et al. An examination of three prescribing cascades in a cohort of older adults with dementia. BMC Geriatr. 2021;21(1):297. doi:10.1186/s12877-021-02246-2
  • Vouri SM, Possinger MC, Usmani S, et al. Evaluation of the Potential Acetylcholinesterase Inhibitor-Induced Rhinorrhea Prescribing Cascade. J Am Geriatr Soc. 2020;68(2):440–441. doi:10.1111/jgs.16224
  • Gill SS, Mamdani M, Naglie G, et al. A prescribing cascade involving cholinesterase inhibitors and anticholinergic drugs. Arch Intern Med. 2005;165(7):808–813. doi:10.1001/archinte.165.7.808
  • Bowie MW, Slattum PW. Pharmacodynamics in older adults: a review. Am J Geriatr Pharmacother. 2007;5:263–303. doi:10.1016/j.amjopharm.2007.10.001
  • Maher D, Ailabouni N, Mangoni AA, Wiese MD, Reeve E. Alterations in drug disposition in older adults: a focus on geriatric syndromes. Expert Opin Drug Metab Toxicol. 2021;17(1):41–52. doi:10.1080/17425255.2021.1839413
  • Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvascular disease - systematic review and meta-analysis. Neurobiol Aging. 2009;30(3):337–352. doi:10.1016/j.neurobiolaging.2007.07.015
  • Nobili A, Garattini S, Mannucci PM. Multiple diseases and polypharmacy in the elderly: challenges for the internist of the third millennium. J Comorbidity. 2011;1:28–44. doi:10.15256/joc.2011.1.4
  • Onder G, Marengoni A, Russo P, et al. Advanced age and medication prescription: more years, less medications? A Nationwide report from the Italian Medicines Agency. J Am Med Dir Assoc. 2016;17(2):168–172. doi:10.1016/j.jamda.2015.08.009
  • Gustafsson M, Sjölander M, Pfister B, Jonsson J, Schneede J, Lövheim H. Drug-related hospital admissions among old people with dementia. Eur J Clin Pharmacol. 2016;72(9):1143–1153. doi:10.1007/s00228-016-2084-3
  • Pirmohamed M, James S, Meakin S, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ. 2004;329(7456):15–19. doi:10.1136/bmj.329.7456.15
  • Desai AK, Grossberg GT. Rivastigmine for Alzheimer’s disease. Expert Rev Neurother. 2005;5(5):563–580. doi:10.1586/14737175.5.5.563
  • Desai AK, Grossberg GT. Review of rivastigmine and its clinical applications in Alzheimer’s disease and related disorders. Expert Opin Pharmacother. 2001;2(4):653–666. doi:10.1517/14656566.2.4.653
  • Román GC, Rogers SJ. Donepezil: a clinical review of current and emerging indications. Expert Opin Pharmacother. 2004;5(1):161–180. doi:10.1517/14656566.5.1.161
  • Kurz A, Farlow M, Lefèvre G. Pharmacokinetics of a novel transdermal rivastigmine patch for the treatment of Alzheimer’s disease: a review. Int J Clin Pract. 2009;63(5):799–805. doi:10.1111/j.1742-1241.2009.02052.x
  • Wysokiński A, Sobów T, Kłoszewska I, Kostka T. Mechanisms of the anorexia of aging-a review. Age (Dordr). 2015;37(4):9821. doi:10.1007/s11357-015-9821-x
  • Thompson CM, Johns DO, Sonawane B, et al. Database for physiologically based pharmacokinetic (PBPK) modeling: physiological data for healthy and health-impaired elderly. J Toxicol Environ Health B Crit Rev. 2009;12(1):1–24. doi:10.1080/10937400802545060
  • Hughes VA, Frontera WR, Wood M, et al. Longitudinal muscle strength changes in older adults: influence of muscle mass, physical activity, and health. J Gerontol a Biol Sci Med Sci. 2001;56(5):B209–217. doi:10.1093/gerona/56.5.B209
  • Hubbard RE, O’Mahony MS, Calver BL, Woodhouse KW. Nutrition, inflammation, and leptin levels in aging and frailty. J Am Geriatr Soc. 2008;56(2):279–284. doi:10.1111/j.1532-5415.2007.01548.x
  • Reeve E, Wiese MD, Mangoni AA. Alterations in drug disposition in older adults. Expert Opin Drug Metab Toxicol. 2015;11(4):491–508. doi:10.1517/17425255.2015.1004310
  • Le Couteur DG, Blyth FM, Creasey HM, et al. The association of alanine transaminase with aging, frailty, and mortality. J Gerontol a Biol Sci Med Sci. 2010;65(7):712–717. doi:10.1093/gerona/glq082
  • Dooley M, Lamb HM. Donepezil: a review of its use in Alzheimer’s disease. Drugs Aging. 2000;16(3):199–226. doi:10.2165/00002512-200016030-00005
  • Hubbard RE, O’Mahony MS, Calver BL, Woodhouse KW. Plasma esterases and inflammation in ageing and frailty. Eur J Clin Pharmacol. 2008;64(9):895–900. doi:10.1007/s00228-008-0499-1
  • Renton KW. Regulation of drug metabolism and disposition during inflammation and infection. Expert Opin Drug Metab Toxicol. 2005;1(4):629–640. doi:10.1517/17425255.1.4.629
  • Aitken AE, Richardson TA, Morgan ET. Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev Pharmacol Toxicol. 2006;46:123–149. doi:10.1146/annurev.pharmtox.46.120604.141059
  • Wynne H. Drug metabolism and ageing. J Br Menopause Soc. 2005;11(2):51–56. doi:10.1258/136218005775544589
  • Zoli M, Magalotti D, Bianchi G, et al. Total and functional hepatic blood flow decrease in parallel with ageing. Age Ageing. 1999;28(1):29–33. doi:10.1093/ageing/28.1.29
  • Wynne HA, Cope LH, Mutch E, Rawlins MD, Woodhouse KW, James OFW. The effect of age upon liver volume and apparent liver blood flow in healthy man. Hepatology. 1989;9(2):297–301. doi:10.1002/hep.1840090222
  • Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab Rev. 2009;41(2):67–76. doi:10.1080/03602530902722679
  • Butler JM, Begg EJ. Free drug metabolic clearance in elderly people. Clin Pharmacokinet. 2008;47(5):297–321. doi:10.2165/00003088-200847050-00002
  • Tiseo PJ, Vargas R, Perdomo CA, Friedhoff LT. An evaluation of the pharmacokinetics of donepezil HCl in patients with impaired hepatic function. Br J Clin Pharmacol. 1998;46 Suppl 1(Suppl 1):51–55. doi:10.1046/j.1365-2125.1998.0460s1051.x
  • Tiseo PJ, Foley K, Friedhoff LT. An evaluation of the pharmacokinetics of donepezil HCl in patients with moderately to severely impaired renal function. Br J Clin Pharmacol. 1998;46 Suppl 1(Suppl1):56–60. doi:10.1046/j.1365-2125.1998.0460s1056.x
  • Fliser D, Zeier M, Nowack R, Ritz E. Renal functional reserve in healthy elderly subjects. J Am Soc Nephrol. 1993;3(7):1371–1377. doi:10.1681/ASN.V371371
  • Viallon A, Guyomarch P, Marjollet O, et al. Creatinine clearance and drug prescriptions for the elderly. A study of 419 patients older than 70 years admitted through the emergency department. Presse Med. 2006;35:413–417. doi:10.1016/S0755-4982(06)74605-X
  • Farlow MR. Clinical pharmacokinetics of galantamine. Clin Pharmacokinet. 2003;42(15):1383–1392. doi:10.2165/00003088-200342150-00005
  • Gottwald MD, Rozanski RI. Rivastigmine, a brain-region selective acetylcholinesterase inhibitor for treating Alzheimer’s disease: review and current status. Expert Opin Investig Drugs. 1999;8(10):1673–1682. doi:10.1517/13543784.8.10.1673
  • Hrycyna CA. Molecular genetic analysis and biochemical characterization of mammalian P- glycoproteins involved in multidrug resistance. Semin Cell Dev Biol. 2001;12(3):247–256. doi:10.1006/scdb.2000.0250
  • Alavijeh MS, Chishty M, Qaiser MZ, Palmer AM. Drug metabolism and pharmacokinetics, the blood-brain barrier, and central nervous system drug discovery. NeuroRx. 2005;2(4):554–571. doi:10.1602/neurorx.2.4.554
  • van Waterschoot RA, Schinkel AH. A critical analysis of the interplay between cytochrome P450 3A and P-glycoprotein: recent insights from knockout and transgenic mice. Pharmacol Rev. 2011;63(2):390–410. doi:10.1124/pr.110.002584
  • van Assema DM, Lubberink M, Rizzu P, et al. Blood-brain barrier P-glycoprotein function in healthy subjects and Alzheimer’s disease patients: effect of polymorphisms in the ABCB1 gene. EJNMMI Res. 2012;2(1):57. doi:10.1186/2191-219X-2-57
  • Trifirò G, Spina E. Age-related changes in pharmacodynamics: focus on drugs acting on central nervous and cardiovascular systems. Curr Drug Metab. 2011;12(7):611–620. doi:10.2174/138920011796504473
  • Bishop JR. Pharmacogenetics. Handb Clin Neurol. 2018;147:59–73.
  • Noetzli M, Guidi M, Ebbing K, et al. Population pharmacokinetic approach to evaluate the effect of CYP2D6, CYP3A, ABCB1, POR and NR1I2 genotypes on donepezil clearance. Br J Clin Pharmacol. 2014;78(1):135–144. doi:10.1111/bcp.12325
  • Darvesh S, Hopkins DA, Geula C. Neurobiology of butyrylcholinesterase. Nat Rev Neurosci. 2003;4(2):131–138. doi:10.1038/nrn1035
  • Harold D, Macgregor S, Patterson CE, et al. A single nucleotide polymorphism in CHAT influences response to acetylcholinesterase inhibitors in Alzheimer’s disease. Pharmacogenet Genomics. 2006;16(2):75–77. doi:10.1097/01.fpc.0000189799.88596.04
  • Pola R, Flex A, Ciaburri M, et al. Responsiveness to cholinesterase inhibitors in Alzheimer’s disease: a possible role for the 192 Q/R polymorphism of the PON-1 gene. Neurosci Lett. 2005;382(3):338–341. doi:10.1016/j.neulet.2005.03.027
  • Varsaldi F, Miglio G, Scordo MG, et al. Impact of the CYP2D6 polymorphism on steady-state plasma concentrations and clinical outcome of donepezil in Alzheimer’s disease patients. Eur J Clin Pharmacol. 2006;62(9):721–726. doi:10.1007/s00228-006-0168-1
  • Pilotto A, Franceschi M, D’Onofrio G, et al. Effect of a CYP2D6 polymorphism on the efficacy of donepezil in patients with Alzheimer disease. Neurology. 2009;73(10):761–767. doi:10.1212/WNL.0b013e3181b6bbe3
  • Albani D, Martinelli Boneschi F. Replication study to confirm the role of CYP2D6 polymorphism rs1080985 on donepezil efficacy in Alzheimer’s disease patients. J Alzheimers Dis. 2012;30(4):745–749. doi:10.3233/JAD-2012-112123
  • Seripa D, Bizzarro A, Pilotto A, et al. Role of cytochrome P4502D6 functional polymorphisms in the efficacy of donepezil in patients with Alzheimer’s disease. Pharmacogenet Genomics. 2011;21(4):225–230. doi:10.1097/FPC.0b013e32833f984c
  • Campbell NL, Skaar TC, Perkins AJ, et al. Characterization of hepatic enzyme activity in older adults with dementia: potential impact on personalizing pharmacotherapy. Clin Interv Aging. 2015;10:269–275. doi:10.2147/CIA.S65980
  • Cacabelos R, Martínez R, Fernández-Novoa L, et al. Genomics of Dementia: APOE- and CYP2D6-Related Pharmacogenetics. Int J Alzheimers Dis. 2012;2012:518901.
  • Liu M, Zhang Y, Huo YR, et al. Influence of the rs1080985 Single Nucleotide Polymorphism of the CYP2D6 Gene and APOE Polymorphism on the Response to Donepezil Treatment in Patients with Alzheimer’s Disease in China. Dement Geriatr Cogn Dis Extra. 2014;4(3):450–456. doi:10.1159/000367596
  • Choi SH, Kim SY, Na HR, et al. Effect of ApoE genotype on response to donepezil in patients with Alzheimer’s disease. Dement Geriatr Cogn Disord. 2008;25(5):445–450. doi:10.1159/000124752
  • Cacabelos R. Influence of pharmacogenetic factors on Alzheimer’s disease therapeutics. Neurodegener Dis. 2008;5(3–4):176–178. doi:10.1159/000113695
  • Zanger UM, Raimundo S, Eichelbaum M. Cytochrome P450 2D6: overview and update on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch Pharmacol. 2004;369(1):23–37. doi:10.1007/s00210-003-0832-2
  • Zhou SF. Polymorphism of human cytochrome P450 2D6 and its clinical significance: part I. Clin Pharmacokinet. 2009;48(11):689–723.
  • Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3:229–243. doi:10.1517/14622416.3.2.229
  • Martinelli-Boneschi F, Giacalone G, Magnani G, et al. Pharmacogenomics in Alzheimer’s disease: a genome-wide association study of response to cholinesterase inhibitors. Neurobiol Aging. 2013;34(6):1711.e7–13. doi:10.1016/j.neurobiolaging.2012.12.008
  • Lu J, Fu J, Zhong Y, et al. Association between ABCA1 gene polymorphisms and the therapeutic response to donepezil therapy in Han Chinese patients with Alzheimer’s disease. Brain Res Bull. 2018;140:1–4. doi:10.1016/j.brainresbull.2018.03.014
  • Triplitt C. Drug interactions of medications commonly used in diabetes. Diabetes. 2006;19(4):202–211.
  • Tavassoli N, Sommet A, Lapeyre-Mestre M, Bagheri H, Montrastruc JL. Drug interactions with Cholinesterase inhibitors: an analysis of the French pharmacovigilance database and a comparison of two national drug formularies (Vidal, British National Formulary). Drug Saf. 2007;30(11):1063–1071. doi:10.2165/00002018-200730110-00005
  • Delafuente JC. Understanding and preventing drug interactions in elderly patients. Crit Rev Oncol Hematol. 2003;48(2):133–143. doi:10.1016/j.critrevonc.2003.04.004
  • Johnell K, Klarin I. The relationship between number of drugs and potential drug-drug interactions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed Drug Register. Drug Saf. 2007;30(10):911–918. doi:10.2165/00002018-200730100-00009
  • Gallicano K, Drusano G. Introduction to drug interactions. In: Piscitelli SC, Rodvold K, editors. Drug Interactions in Infectious Diseases. 2nd ed. Totowa: Humana Press Inc; 2005:1–12.
  • Pasqualetti G, Tognini S, Calsolaro V, et al. Potential drug-drug interactions in Alzheimer patients with behavioral symptoms. Clin Interv Aging. 2015;10:1457–1466.
  • Magnuson TM, Keller BK, Burke WJ. Extrapyramidal side effects in a patient treated with risperidone plus donepezil. Am J Psychiatry. 1998;155(10):1458–1459. doi:10.1176/ajp.155.10.1458a
  • Mehrpouya M, Ataei S, Nili-Ahmadabadi A. Potential drug interactions with cholinesterase inhibitors in Alzheimer patients: a guideline for Neurologists. J Appl Pharm Sci. 2017;7(1):223–226. doi:10.7324/JAPS.2017.70134
  • Lin JH, Lu AYH. Inhibition and induction of cytochrome P450 and the clinical implications. Clin Pharmacokinet. 1998;35(5):361–390. doi:10.2165/00003088-199835050-00003
  • McEneny-King A, Edginton AN, Pp R. Investigating the binding interactions of the anti- Alzheimer’s drug donepezil with CYP3A4 and P-glycoprotein. Bioorg Med Chem Lett. 2015;25(2):297–301. doi:10.1016/j.bmcl.2014.11.046
  • Spina E, Scordo MG, D’Arrigo C. Metabolic drug interactions with new psychotropic agents. Fundam Clin Pharmacol. 2003;17(5):517–538. doi:10.1046/j.1472-8206.2003.00193.x
  • Tiseo PJ, Perdomo CA, Friedhoff LT. Concurrent administration of donepezil HCl and ketoconazole: assessment of pharmacokinetic changes following single and multiple doses. Br J Clin Pharmacol. 1998;46(Suppl 1):30–34. doi:10.1046/j.1365-2125.1998.0460s1030.x
  • Defilippi JL, Crismon ML. Drug interactions with cholinesterase inhibitors. Drugs Aging. 2003;20(6):437–444. doi:10.2165/00002512-200320060-00003
  • Carrier L. Donepezil and paroxetine: possible drug interaction. J Am Geriatr Soc. 1999;47(8):1037. doi:10.1111/j.1532-5415.1999.tb01306.x
  • Finch A, Pillans P. P-glycoprotein and its role in drug-drug interactions. Aust Prescr. 2014;37:137–139. doi:10.18773/austprescr.2014.050
  • US Food and Drug Administration. Clinical drug interaction studies-Cytochrome P450 enzyme- and transporter-mediated drug interactions guidance for industry; January 2020. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/clinical-drug-interaction-studies-cytochrome-p450-enzyme-and-transporter-mediated-drug-interactions. Accessed on November 5, 2020.
  • Takeuchi R, Shinozaki K, Nakanishi T, Tamai I. Local Drug-Drug Interaction of Donepezil with Cilostazol at Breast Cancer Resistance Protein (ABCG2) Increases Drug Accumulation in Heart. Drug Metab Dispos. 2016;44(1):68–74. doi:10.1124/dmd.115.066654
  • Johnell K, Fastbom J. Concurrent use of anticholinergic drugs and cholinesterase inhibitors: register-based study of over 700,000 elderly patients. Drugs Aging. 2008;25:871–877. doi:10.2165/00002512-200825100-00006
  • Modi A, Weiner M, Craig BA, Sands LP, Rosenman MB, Thomas J. Concomitant use of anticholinergics with acetylcholinesterase inhibitors in Medicaid recipients with dementia and residing in nursing homes. J Am Geriatr Soc. 2009;57:1238–1244. doi:10.1111/j.1532-5415.2009.02258.x
  • Walker C, Perks D. Do you know about donepezil and succinylcholine? Anaesthesia. 2002;57:1041. doi:10.1046/j.1365-2044.2002.00117.x
  • Sprung J, Castellani WJ, Srinivasan V, Udayashankar S. The effects of donepezil and neostigmine in a patient with unusual pseudocholinesterase activity. Anesth Analg. 1998;87(5):1203–1205.
  • Sánchez Morillo J, Demartini Ferrari A, Roca de Togores López A. Interacción entre donepezilo y bloqueantes musculares en la enfermedad de Alzheimer [Interaction of donepezil and muscular blockers in Alzheimer’s disease]. Rev Esp Anestesiol Reanim. 2003;50(2):97–100.
  • Carcenac D, Martin-Hunyadi C, Kiesmann M, Demuynck-Roegel C, Alt M, Kuntzmann F. Syndrome extrapyramidal sous donepezil [Extra-pyramidal syndrome induced by donepezil]. Presse Med. 2000;29(18):992–993.
  • Shimizu S, Mizuguchi Y, Sobue A, Fujiwara M, Morimoto T, Ohno Y. Interaction between anti- Alzheimer and antipsychotic drugs in modulating extrapyramidal motor disorders in mice. J Pharmacol Sci. 2015;127(4):439–445. doi:10.1016/j.jphs.2015.03.004
  • Bentue-Ferrer D, Tribut O, Polard E, Allain H. Clinically significant drug interactions with cholinesterase inhibitors: a guide for neurologists. CNS Drugs. 2003;17:947–963. doi:10.2165/00023210-200317130-00002
  • [No authors listed]. Bradycardia due to cholinesterase inhibitors: identify adverse effects and take them into account. Prescrire Int. 2011;20(115):95.
  • Hong YJ, Han HJ, Youn YC, et al. Safety and tolerability of donepezil 23 mg with or without intermediate dose titration in patients with Alzheimer’s disease taking donepezil 10 mg: a multicenter, randomized, open-label, parallel-design, three-arm, prospective trial. Alzheimers Res Ther. 2019;11(1):37. doi:10.1186/s13195-019-0492-1
  • Christensen DD. Higher-dose (23 mg/day) donepezil formulation for the treatment of patients with moderate-to-severe Alzheimer’s disease. Postgrad Med. 2012;124(6):110–116. doi:10.3810/pgm.2012.11.2589
  • Reyes JF, Vargas R, Kumar D, et al. Steady-state pharmacokinetics, pharmacodynamics and tolerability of donepezil hydrochloride in hepatically impaired patients. Br J Clin Pharmacol. 2004;58(Suppl 1):9–17. doi:10.1111/j.1365-2125.2004.01802.x
  • Ali TB, Schleret TR, Reilly BM, et al. Adverse effects of cholinesterase inhibitors in dementia, according to the pharmacovigilance databases of the United-States and Canada. PLoS One. 2015;10(12):e0144337. doi:10.1371/journal.pone.0144337
  • Sheffrin M, Miao Y, Boscardin WJ, et al. Weight loss associated with cholinesterase inhibitors in individuals with dementia in a national healthcare system. J Am Geriatr Soc. 2015;63(8):1512–1518. doi:10.1111/jgs.13511
  • Vetrano DL, Tosato M, Colloca G, et al. Polypharmacy in nursing home residents with severe cognitive impairment: results from the SHELTER Study. Alzheimers Dement. 2013;9(5):587–593. doi:10.1016/j.jalz.2012.09.009
  • Renn BN, Asghar-Ali AA, Thielke S, et al. A Systematic Review of Practice Guidelines and Recommendations for Discontinuation of Cholinesterase Inhibitors in Dementia. Am J Geriatr Psychiatry. 2018;26(2):134–147. doi:10.1016/j.jagp.2017.09.027
  • Reeve E, Farrell B, Thompson W, et al. Deprescribing cholinesterase inhibitors and memantine In dementia: guideline summary. Med J Aust. 2019;210(4):174–179. doi:10.5694/mja2.50015
  • Corsonello A, Pedone C, Incalzi RA. Age-related pharmacokinetic and pharmacodynamic changes and related risk of adverse drug reactions. Curr Med Chem. 2010;17(6):571–584. doi:10.2174/092986710790416326
  • Gurwitz JH, Field TS, Harrold LR, et al. Incidence and preventability of adverse drug events among older persons in the ambulatory setting. JAMA. 2003;289(9):1107–1116. doi:10.1001/jama.289.9.1107
  • Eshetie TC, Nguyen TA, Gillam MH, Kalisch Ellett LM. A narrative review of problems with medicines use in people with dementia. Expert Opin Drug Saf. 2018;17(8):825–836. doi:10.1080/14740338.2018.1497156
  • Pfister B, Jonsson J, Gustafsson M. Drug-related problems and medication reviews among old people with dementia. BMC Pharmacol Toxicol. 2017;18(1):52. doi:10.1186/s40360-017-0157-2
  • Wucherer D, Thyrian JR, Eichler T, et al. Drug-related problems in community-dwelling primary care patients screened positive for dementia. Int Psychogeriatrics. 2017;29(11):1857–1868. doi:10.1017/S1041610217001442
  • Mallet L, Spinewine A, Huang A. The challenge of managing drug interactions in elderly people. Lancet. 2007;370(9582):185–191. doi:10.1016/S0140-6736(07)61092-7
  • Lau DT, Mercaldo ND, Harris AT, Trittschuh E, Shega J, Weintraub S. Polypharmacy and potentially inappropriate medication use among community-dwelling elders with dementia. Alzheimer Dis Assoc Disord. 2010;24(1):56–63. doi:10.1097/WAD.0b013e31819d6ec9
  • Micromedex® HealthCare Series (internet database) Version 2.0. Greenwood Village, Colorado: Thomson Healthcare Inc; 2013. Available from: http://www.micromedexsolutions.com. Accessed November 22, 2020.
  • By the 2019 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2019 updated AGS Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2019;67(4):674–694.
  • Gustafsson M, Sjölander M, Pfister B, Schneede J, Lövheim H. Effects of Pharmacists’ Interventions on Inappropriate Drug Use and Drug-Related Readmissions in People with Dementia-A Secondary Analysis of a Randomized Controlled Trial. Pharmacy (Basel). 2018;6(1):7. doi:10.3390/pharmacy6010007
  • Heikkilä T, Lekander T, Raunio H. Use of an online surveillance system for screening drug interactions in prescriptions in community pharmacies. Eur J Clin Pharmacol. 2006;62(8):661–665. doi:10.1007/s00228-006-0149-4
  • Horn JR, Hansten PD, Chan LN. Proposal for a new tool to evaluate drug interaction cases. Ann Pharmacother. 2007;41(4):674–680. doi:10.1345/aph.1H423
  • Le Couteur DG, Robinson M, Leverton A, et al. Adherence, persistence and continuation with cholinesterase inhibitors in Alzheimer’s disease. Australas J Ageing. 2012;31(3):164–169. doi:10.1111/j.1741-6612.2011.00564.x
  • Cotrell V, Wild K, Bader T. Medication management and adherence among cognitively impaired older adults. J Gerontol Soc Work. 2006;47(3–4):31–46. doi:10.1300/J083v47n03_03
  • Thiruchselvam T, Naglie G, Moineddin R, et al. Risk factors for medication nonadherence in older adults with cognitive impairment who live alone. Int J Geriatr Psychiatry. 2012;27(12):1275–1282. doi:10.1002/gps.3778
  • Maxwell CJ, Stock K, Seitz D, Herrmann N. Persistence and adherence with dementia pharmacotherapy: relevance of patient, provider, and system factors. Can J Psychiatry. 2014;59(12):624–631. doi:10.1177/070674371405901203
  • Elliott RA, Goeman D, Beanland C, Koch S. Ability of older people with dementia or cognitive impairment to manage medicine regimens: a narrative review. Curr Clin Pharmacol. 2015;10(3):213–221. doi:10.2174/1574884710666150812141525
  • Arlt S, Lindner R, Rösler A, von Renteln-kruse W. Adherence to medication in patients with dementia: predictors and strategies for improvement. Drugs Aging. 2008;25(12):1033–1047. doi:10.2165/0002512-200825120-00005
  • Kröger E, Tatar O, Vedel I, et al. Improving medication adherence among community-dwelling seniors with cognitive impairment: a systematic review of interventions. Int J Clin Pharm. 2017;39(4):641–656. doi:10.1007/s11096-017-0487-6