Buscar en
Neurología (English Edition)
Toda la web
Inicio Neurología (English Edition) Pharmacogenetic studies in Alzheimer disease
Journal Information
Vol. 37. Issue 4.
Pages 287-303 (May 2022)
Visits
2673
Vol. 37. Issue 4.
Pages 287-303 (May 2022)
Review article
Open Access
Pharmacogenetic studies in Alzheimer disease
Estudios farmacogenéticos en la enfermedad de Alzheimer
Visits
2673
T. Zúñiga Santamaríaa,b, P. Yescas Gómezb, I. Fricke Galindoc, M. González Gonzálezd, A. Ortega Vázqueze, M. López Lópeze,
Corresponding author
mlopez@correo.xoc.uam.mx

Corresponding author.
a Maestría en Ciencias Farmacéuticas, Universidad Autónoma Metropolitana, Unidad Xochimilco, Coyoacán (México D.F.), Mexico
b Departamento de Neurogenética, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Tlalpan (México D.F.), Mexico
c Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Xochimilco, Coyoacán (México D.F.), Mexico
d Unidad de Cognición y Conducta, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Tlalpan (México D.F.), Mexico
e Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Coyoacán (México D.F.), Mexico
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Tables (5)
Table 1. Pharmacogenetic studies of disease-modifying drugs for Alzheimer disease.
Table 2. Genes included in pharmacogenetic studies of disease-modifying drugs for Alzheimer disease.
Table 3. Key aspects of pharmacogenetic studies evaluating clinical response to disease-modifying drugs for Alzheimer disease.
Table 4. Main tests used to assess cognitive impairment and functional status in pharmacogenetic studies into Alzheimer disease.
Table 5. Specifications of the methods used to determine plasma concentrations of disease-modifying drugs for Alzheimer disease in the pharmacogenetic studies reviewed.
Show moreShow less
Abstract
Introduction

Alzheimer disease (AD) is the most common cause of dementia and is considered one of the main causes of disability and dependence affecting quality of life in elderly people and their families. Current pharmacological treatment includes acetylcholinesterase inhibitors (donepezil, galantamine, rivastigmine) and memantine; however, only one-third of patients respond to treatment. Genetic factors have been shown to play a role in this inter-individual variability in drug response.

Development

We review pharmacogenetic reports of AD-modifying drugs, the pharmacogenetic biomarkers included, and the phenotypes evaluated. We also discuss relevant methodological considerations for the design of pharmacogenetic studies into AD. A total of 33 pharmacogenetic reports were found; the majority of these focused on the variability in response to and metabolism of donepezil. Most of the patients included were from Caucasian populations, although some studies also include Korean, Indian, and Brazilian patients. CYP2D6 and APOE are the most frequently studied biomarkers. The associations proposed are controversial.

Conclusions

Potential pharmacogenetic biomarkers for AD have been identified; however, it is still necessary to conduct further research into other populations and to identify new biomarkers. This information could assist in predicting patient response to these drugs and contribute to better treatment decision-making in a context as complex as ageing.

Keywords:
Alzheimer disease
Pharmacogenetics
Acetylcholinesterase inhibitors
APOE
CYP2D6
ABCB1
Resumen
Introducción

La enfermedad de Alzheimer (EA) es la primera causa de demencia y una de las principales causas de discapacidad y dependencia que afecta la calidad de vida de los adultos mayores y de sus familiares. En la actualidad el manejo farmacológico disponible incluye a los fármacos inhibidores de la acetilcolinesterasa (donepezilo, galantamina, rivastigmina) y a la memantina. Sin embargo, se ha reportado que solo un tercio de los pacientes responden al tratamiento. Se ha evidenciado que los factores genéticos pueden explicar parte de la variabilidad en la respuesta a estos fármacos.

Desarrollo

En esta revisión se incluyen los estudios farmacogenéticos de fármacos modificadores de EA, los farmacogenes analizados y los fenotipos que fueron evaluados, además de las consideraciones metodológicas que es importante tomar en cuenta en este tipo de estudios. Se encontraron 33 reportes de farmacogenética de EA en los que principalmente se ha estudiado la variabilidad en la respuesta y en el metabolismo de donepezilo. La población más estudiada es la caucásica, aunque también han sido investigados coreanos, indios y brasileños. Los biomarcadores más estudiados son CYP2D6 y APOE. Los resultados de las asociaciones son controversiales.

Conclusiones

Se han identificado posibles biomarcadores farmacogenéticos para el tratamiento de EA; sin embargo, se requieren más estudios farmacogenéticos en otras poblaciones que no han sido investigadas, así como profundizar en la identificación de los biomarcadores. Este conocimiento podría ayudar a predecir la respuesta a fármacos modificadores de EA y contribuiría a tomar mejores decisiones en el tratamiento de la enfermedad en un contexto tan complejo como es el envejecimiento.

Palabras clave:
Enfermedad de Alzheimer
Farmacogenética
Inhibidores de acetilcolinesterasa
APOE
CYP2D6
ABCB1
Full Text
IntroductionDefinition, incidence, and issues of Alzheimer disease

Population ageing has given rise to a constant increase in the prevalence of chronic degenerative diseases, including such neurodegenerative diseases as dementia.1

Dementia is characterised by progressive, irreversible decline of behavioural and cognitive function (attention, orientation, memory, language, visuospatial function, executive function, and praxis), which progressively affects the quality of life of both the patients and their family and primary caregivers, contributing to the status of dementia as one of the leading causes of disability and dependence.2 For these reasons, the World Health Organization and the G8 have designated dementia a public health priority (in 2012 and 2014, respectively).1,3,4

The prevalence of dementia ranges from 5% to 10% among the population aged over 65, with the figure doubling every 5 years, reaching 25%-50% among those aged over 85.5

Alzheimer disease (AD), the most frequent cause of dementia, accounts for 60% of known cases and is defined as a neurodegenerative disease characterised by reduced neuronal function, which leads to synaptic loss and neuronal death; this causes lasting cognitive impairment, affects functional capacity, and progressively gives rise to dependence and disability.6–8

The number of patients with AD worldwide is estimated at 46 million, according to a 2015 report by Alzheimer's Disease International,1 and is projected to increase to 131.5 million by 2050. According to the report, nearly two-thirds of patients with dementia live in developing countries (including Latin American and Caribbean countries), and these regions are expected to present a greater increase in dementia cases in the coming years.1 Compared to more developed regions, access to specialised care in developing countries is limited. However, it should be noted that both in developed and in developing countries, an individual's social setting is an important risk factor for disease progression, and social services are essential in dementia care.

Since the discovery of amyloid-β peptide (Aβ) and tau protein, the main components of amyloid plaques and neurofibrillary tangles, research into AD pathophysiology has yielded crucial information on the pathogenic molecular changes occurring at the neuronal level. Although these pathological changes constitute a sine qua non for the diagnosis of AD and are sufficient to cause cognitive and behavioural symptoms in some patients (memory loss and executive dysfunction affecting the activities of daily living), numerous risk factors are reported in patients developing symptoms after the age of 75 years.7,8 However, the pathophysiology of AD is very complex and much about the disease is yet to be understood; therefore, the development of novel drugs to cure AD or persistently slow or stop its progression has to date been unsuccessful.

Most cases of AD are sporadic and of multifactorial origin, with onset after the age of 65; the ɛ4 allele of the APOE gene has been identified as a significant risk factor in these cases.9 Familial forms follow an autosomal dominant pattern of inheritance, and onset is early. The main causal mutations affect the genes encoding amyloid precursor protein (APP), presenilin-1 (PSEN1), and presenilin-2 (PSEN2).

Two essential elements in the proper management of AD are early diagnosis and selection of the optimal treatment for each patient. Differential diagnosis of AD is complex, requiring imaging and neuropsychological studies, clinical assessment, and interviews with the primary caregiver or a family member. In 1984, the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association (NINCDS-ADRDA) published the first diagnostic criteria for AD.10 These were updated in 2007 with the criteria proposed by Dubois et al.,11 and later revised in 2011 by the National Institute on Aging/Alzheimer Association; the latest version is known as the NIA-AA guidelines.12

In familial cases, genetic testing assists in accurate diagnosis of the disease, which is important for starting treatment early and providing genetic counselling to family members at risk of developing AD.13,14

Available treatments and variability in response

Two types of treatment are currently available for AD. Non-pharmacological treatment includes memory training, social and mental stimulation, music therapy, aromatherapy, and physical exercise programmes, among other approaches. This strategy aims to slow the progression of cognitive and functional impairment; to conserve and promote the preserved cognitive, functional, and social skills and capacities; to restore unused cognitive abilities; to prevent disconnection from the environment; and to strengthen social relationships.2 Studies evaluating the efficacy of non-pharmacological treatment show that it is cost-effective and has positive results in terms of delaying institutionalisation and cognitive function; this results in improved quality of life for patients and caregivers.15,16

This review only addresses pharmacological treatment, which comprises a small number of drugs used in the management of dementia. Currently, the only drugs approved by international regulators are 3 acetylcholinesterase inhibitors (donepezil, galantamine, and rivastigmine) and memantine. The efficacy of these drugs is variable and considered to be poor to moderate. A recent study calculated the rate of response to 3 of these drugs at 27.8%.17 Furthermore, AD treatments are associated with numerous adverse reactions, including diarrhoea, nausea, instability, vomiting, weight loss, stomach ulcers, syncope, and generalised seizures; in patients presenting these reactions, it may be necessary to withdraw treatment, limiting the therapeutic options available.18–20 Pharmacogenetic analysis may assist in explaining how genetic variability between patients contributes to these differences in treatment response and in the metabolism of drugs, including disease-modifying drugs.

This represents an opportunity for the development of novel diagnostic and therapeutic models for dementia, which may aim to preserve patients’ functional capacity and improve their quality of life. The challenges before us are novel, complex, and interesting, and represent a new vision of diagnosis and treatment, involving the tools of molecular biology and pharmacogenetics. Thus, there is a clear need to include analysis of molecular biomarkers in the early diagnosis of AD to assist in differential diagnosis, as well as in AD prevention and care.

This review describes the pharmacogenetic studies analysing disease-modifying drugs for AD and the pharmacogenes and phenotypes analysed, and addresses important methodological considerations to be taken into account in these studies. We performed a systematic review of articles published before February 2018 on the PubMed and PharmGKB databases, using the search terms “Alzheimer,” “pharmacogenetics,” “pharmacogenomics,” “donepezil,” “galantamine,” “rivastigmine,” and “memantine.”

Pharmacogenetics of disease-modifying drugs for Alzheimer disease

Between 75% and 85% of variability in treatment response to donepezil and other acetylcholinesterase inhibitors metabolised by cytochrome P450 (CYP) enzymes is thought to be explained by pharmacogenetic factors.21 Taking these factors into account may help to improve the safety and efficacy of AD treatment.

Acetylcholinesterase inhibitors and memantine function through distinct metabolic pathways (Fig. 1). Donepezil and galantamine are metabolised in the liver, mainly by the enzymes CYP3A4, CYP2D6, and CYP1A2, whereas the biotransformation of rivastigmine occurs through the process of carbamylation. Hepatic metabolism of memantine is limited, and a considerable proportion of the drug is renally cleared. In terms of pharmacodynamics, the acetylcholinesterase inhibitors present different affinities for acetylcholinesterase (ACHE) and butyrylcholinesterase (BCHE): donepezil and galantamine inhibit ACHE more than BCHE, whereas rivastigmine inhibits both enzymes equally.21,22 ACHE is one of the most important enzymes in nerve functioning and response, as it catalyses acetylcholine hydrolysis in both the autonomic and peripheral nervous systems. BCHE hydrolyses both butyrylcholine and acetylcholine, and is distributed extensively in the liver, lungs, heart, and brain.23 The binding of drugs to ACHE and BCHE inhibits acetylcholine hydrolysis in the hippocampus, increasing the concentration of the neurotransmitter in the synaptic space and preventing it from binding to postsynaptic cholinergic receptors. This contributes to the improvement of cognitive symptoms, as cholinergic neurotransmission, which is involved in memory, attention, and emotion, is reported to be severely affected in AD.22

Figure 1.

Proteins that interact with disease-modifying drugs for Alzheimer disease.

ACHE: acetylcholinesterase; BCHE: butyrylcholinesterase; BBB: blood–brain barrier; CHAT: choline acetyltransferase; DPZ: donepezil; GAL: galantamine; MEM: memantine; NMDA: N-methyl-D-aspartate; RIV: rivastigmine.

(0.15MB).

Memantine, in turn, is a moderate-affinity N-methyl-d-aspartate (NMDA) receptor antagonist that inhibits the pathological effect of elevated glutamate levels, which lead to the neuronal death and cell damage that characterise AD.22

P-glycoprotein has been shown to play a significant role in transporting donepezil across the blood–brain barrier.24

Pharmacogenetic studies into disease-modifying drugs for Alzheimer disease

Table 1 summarises key information from pharmacogenetic studies into disease-modifying drugs for AD. We identified 33 articles, with donepezil being the most frequently studied drug (included in 24 studies); only 3 studies included patients treated with memantine. The most frequently studied population was white Europeans, although other studies were performed with Korean, Indian, and Brazilian populations, among others. Most studies evaluated the association between pharmacogenetic variants and response to disease-modifying drugs for AD; 8 also analysed pharmacokinetic parameters and plasma concentrations of the drugs.

Table 1.

Pharmacogenetic studies of disease-modifying drugs for Alzheimer disease.

Drug  Population studied (nGene  Phenotype evaluated  Conclusions  Ref. 
DonepezilItalian (42)  CYP2D6  PC and response  CYP2D6 variants influence PC of donepezil and treatment response.  42 
Italian (127)  CYP2D6 and APOE  Response  CYP2D6 variant rs1080985 may influence donepezil efficacy in patients with mild or moderate AD.  26 
White (57)  CYP2D6  Response  CYP2D6 variants may influence donepezil efficacy.  27 
White (80)  APOE  Response  APOE genotype and sex do not predict response to donepezil in patients with AD.  52 
White (128) and Arab (1)  CYP2D6, CYP3A4/5/7, POR, NR1I2, and ABCB1  Pharmacokinetic parameters  Functional alleles of CYP2D6 contribute to variability in donepezil disposition.  53 
Italian (54)  CYP3A4, CYP3A5, and ABCB1  PC and response  CYP3A4 and CYP3A5 variants do not affect donepezil metabolism or response to the treatment. ABCB1 variants may influence donepezil disposition and response to the treatment.  54 
Italian (81)  APOE  Response  APOE ɛ4 predicts adequate response to donepezil.  55 
French-Canadian (Quebec) (367)  APOE and BCHE  Response  In carriers of the APOE ɛ4 and BCHE-Ka variants, symptoms improved with donepezil.  56 
Bulgarian, Czech, Russian, Slovakian, South African, Ukrainian, British, and North American (335)  APOE  Response  APOE genotype did not affect patient response to donepezil.  57 
Korean (51)  APOE  Response  Carriers of APOE ɛ4 may respond better to donepezil.  58 
Brazilian (42)  APOE and CYP2D6  PC and response  Treatment response was associated with PC of donepezil but not with APOE or CYP2D6 genotype.  59 
White Polish (88)  APOE and CYP2D6  Response  APOE and CYP2D6 variants do not affect response to donepezil.  60 
Non-Hispanic white (574)  BCHE  Response  The BCHE-K variant is associated with deleterious changes in cognitive performance in patients treated with donepezil.  61 
GalantamineEuropean (27)  CYP2D6, CYP3A, POR, and ABCB1  Steady-state PC  CYP2D6 influences PC of galantamine.  62 
European (39)  APOE  Response  Sex may predict response to treatment with acetylcholinesterase inhibitors better than APOE63 
White (569)  APOE  Response  APOE genotype showed no effect on galantamine efficacy.  64 
RivastigmineMainly white (490)  APOE and BCHE  Response  BCHE genotype showed an association with clinical aspects of AD.  65 
Mainly European (367)  APOE  Response  Rivastigmine has considerable, qualitatively similar benefits in APOE ɛ4 carriers and non-carriers.  66 
Taiwanese (63)  APOE  PC and response  Cognitive improvement may be associated with higher PC of rivastigmine, lower baseline MMSE or CDR-SB scores, and absence of the APOE ɛ4 allele.  67 
Donepezil, galantamine, and rivastigmineItalian (171)  CYP2D6, BCHE, and APOE  Response  Individualised treatment based on CYP2D6 and BCHE is not beneficial in AD.  68 
Brazilian (97)  APOE and CYP2D6  Response  Pharmacogenetic factors are associated with treatment response.  17 
Korean (158)  CHAT, SLC5A7, and ACHE  Response  A CHAT variant influences response to AD drugs.  69 
Brazilians of European, Asian, and African origin (177)  APOE and CHRNA7  Response  Variant rs6494223 of CHRNA7 and APOE ɛ4 may be useful for understanding response to treatment with acetylcholinesterase inhibitors.  70 
Italian (176)  GWASa  Response  PRKCE and NBEA polymorphisms may be biomarkers of treatment response or AD severity.  71 
European (165)  APOE and BCHE  Response  Genotyping of APOE and BCHE may inform therapeutic decision-making for patients with AD.  28 
European (121)  CHAT  Response  Variant rs733722 is a potential biomarker of response to acetylcholinesterase inhibitors in patients with AD.  72 
Taiwanese (223)  CHRNA7  Response  CHRNA7 variants are associated with better response to acetylcholinesterase inhibitors.  73 
Italian (169)  CHRNA7  Response  CHRNA7 variants were not found to be associated with response to treatment with acetylcholinesterase.  74 
Donepezil and rivastigmineItalian (471)  ACHE, BCHE, and CHAT  Response  The variants studied do not influence response to treatment with donepezil or rivastigmine.  25 
White (114)  BCHE  Response  An association was found between BCHE variants and AD treatment, especially with rivastigmine.  29 
Rivastigmine and memantineKorean (146)  BCHE  Response  BCHE-K genotype is associated with less marked response to treatment with rivastigmine and memantine, especially in patients carrying the APOE ɛ4 allele.  30 
North Indian (36)  CYP2D6, CYP3A4, CYP2C9/19, UGT2B7, UGT1A6, UGT1A9  PC and response  Poor metabolisers of UGT2B7 polymorphism treated with rivastigmine and memantine presented higher PC of the drugs and poorer treatment response.  75 
Memantine  European (108)  SLC22A1/2/5, SLC47A1, ABCB1, NR1I2, NR1I3, RXR, PPARA  Pharmacokinetic parameters  NR1I2 variant rs1523130 affects memantine clearance.  76 

CDR-SB: Clinical Dementia Rating – Sum of Boxes; PC: plasma concentration; AD: Alzheimer disease; GWAS: genome-wide association study; MMSE: Mini-Mental State Examination; Ref.: reference.

a

This study did not evaluate the association between treatment response and a specific gene; rather, a GWAS was performed in patients who responded to treatment with acetylcholinesterase inhibitors.

The majority of the positive associations reported were found between CYP2D6 variants and plasma concentrations of acetylcholinesterase inhibitors and the response to these drugs. Results from studies into the association between treatment response and variants of APOE and BCHE were more controversial. Although few studies included patients treated with memantine, variants of UGT2B7, BCHE, and NR1I2 are reported to be associated with pharmacokinetic parameters and with variation in response to the drug (Table 1).

The pharmacogenes considered in the studies reviewed include ABCB1, ACHE, APOE, BCHE, CHAT, CHRNA7, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5, CYP3A7, NR1I2, NR1I3, POR, PPAR, RXR, SLC22A1/2/5, SLC47A1, SLC5A7, UGT1A6, UGT1A9, and UGT2B7. Descriptions of these genes are given in Table 2. The APOE gene has mainly been studied in relation to the risk of developing AD. Carriers of the ɛ4 allele are reported to be at greater risk of AD compared to those with the ɛ3 allele; the ɛ2 allele has been identified as a protective factor.9 The ACHE, BCHE, CHAT, CHRNA7, and SLC5A7 genes participate directly in acetylcholine formation and metabolism.25 Other genes analysed in the pharmacogenetic studies reviewed are involved in the transport, metabolism, and inhibition of acetylcholinesterase inhibitors (e.g., ABCB1, CYP2D6, CYP3A4, CYP3A5, and CYP3A7) or in the transcription and expression of these genes (e.g., POR, NR1I2, NR1I3, RXR, and PPAR).

Table 2.

Genes included in pharmacogenetic studies of disease-modifying drugs for Alzheimer disease.

Symbol  Location  Descriptiona 
ABCB1  7q21.12  ATP Binding Cassette Subfamily B Member 1. Efflux transporter also known as P-glycoprotein, which transports xenobiotics out of the cell 
ACHE  7q22  Acetylcholinesterase. Responsible for hydrolysis of acetylcholine in the synapses of cholinergic neurons to terminate signalling 
APOE  19q13.2  Apolipoprotein E. Binds to a specific cellular receptor in the liver and peripheral tissue. It is essential in the normal catalysis of triglyceride-rich lipoproteins. 
BCHE  3q26.1-q26.2  Butyrylcholinesterase. A cholinesterase belonging to the carboxylesterase type B/lipase family of proteins. Involved in the detoxification of such toxic substances as organophosphorous compounds and in the metabolism of substances including cocaine, heroin, and acetylsalicylic acid 
CHAT  10q11.2  Choline acetyltransferase. Enzyme that catalyses acetylcholine biosynthesis 
CHRNA7  15q14  Cholinergic receptor nicotinic alpha 7 subunit. Receptor belonging to a superfamily of ligand-gated ion channels involved in synaptic transmission 
CYP2C9  10q24  Cytochrome P450 family 2 subfamily C member 9. Monooxygenase that catalyses several reactions involved in drug metabolism (phenytoin, tolbutamide, ibuprofen, and S-warfarin) and the synthesis of cholesterol, steroids, and other lipids 
CYP2C19  10q24  Cytochrome P450 family 2 subfamily C member 19. Monooxygenase involved in the synthesis of cholesterol, steroids, and other lipids and in the metabolism of omeprazol, diazepam, and some barbiturates 
CYP2D6  22q13.1  Cytochrome P450 family 2 subfamily D member 6. Monooxygenase involved in the synthesis of cholesterol, steroids, and other lipids and in the metabolism of antidepressants, antipsychotics, analgesics, antitussives, beta-blockers, antiarrhythmic drugs, and antiemetics 
CYP3A4  7q21.1  Cytochrome P450 family 3 subfamily A member 4. Monooxygenase involved in the metabolism of steroids, carcinogens, and approximately half of all common drugs, such as acetaminophen, codeine, cyclosporin A, diazepam, and erythromycin 
CYP3A5  7q21.1  Cytochrome P450 family 3 subfamily A member 5. Monoxygenase involved in the metabolism of drugs and such hormones as testosterone and progesterone 
CYP3A7  7q22.1  Cytochrome P450 family 3 subfamily A member 7. Monooxygenase that hydrolyses testosterone and dehydroepiandrosterone 3-sulfate, involved in oestriol formation during pregnancy 
NR1I2  3q12-q13.3  Nuclear receptor subfamily 1 group I member 2. Transcription factor characterised by presence of a ligand-binding domain and a DNA-binding domain. Regulates the transcription of CYP3A4 
NR1I3  1q23.3  Nuclear receptor subfamily 1 group I member 3. Regulates the metabolism of xenobiotic and endobiotic compounds. Regulates the transcription of genes involved in drug metabolism and bilirubin clearance, for example members of the cytochrome P450 family 
POR  7q11.2  Cytochrome P450 oxidoreductase. Oxidoreductase that binds to 2 cofactors, FAD and FMN, which donate electrons from NADPH to all microsomal P450 enzymes 
PPARA  22q13.31  Peroxisome proliferator-activated receptor alpha. Regulates the expression of genes involved in cell proliferation and differentiation, and the inflammatory and immune responses 
RXR  9q34.3  Retinoid X receptor. Nuclear receptor participating in the biological effects of retinoids due to its involvement in retinoic acid-mediated gene activation 
SLC22A1, SLC22A2, and SLC22A5  6q25.3  Solute carrier family 22 members 1, 2, and 5. Play an important role in the clearance of endogenous small organic cations, drugs, and environmental toxins 
SLC47A1  17p11.2  Solute carrier family 47 member 1. Transporter protein of unknown function 
SLC5A7  2q12  Solute carrier family 5 member 7. High-affinity sodium- and chlorine-dependent transporter that mediates choline uptake for the synthesis of acetylcholine in cholinergic neurons 
UGT1A6 and UGT1A9  2q37  UDP-glucuronosyltransferase family 1 members A6 and A9. Involved in the glucuronidation of small lipophilic molecules including steroids, bilirubin, hormones, and drugs, to convert them into hydrophilic metabolites 
UGT2B7  4q13  UDP-glucuronosyltransferase family 2 member B7. Involved in the conjugation and clearance of endogenous and xenobiotic compounds. Presents a specific affinity to oestriol and 3,4-catechol oestrogen. 

ATP: adenosine triphosphate; FAD: flavin adenine dinucleotide; FMN: flavin mononucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; UDP: uridine diphosphate.

a

Source: NCBI http://www.ncbi.nlm.nih.gov/gene/.

Key considerations in pharmacogenetic research into Alzheimer diseaseEvaluating clinical response

In order to study the association between genetic variants and patients’ response to disease-modifying drugs for AD, we need an established set of clinical parameters and neuropsychological tests to evaluate cognition after treatment onset. The pharmacogenetic studies identified in the literature search use criteria established by the British National Institute for Clinical Excellence (NICE) and by the United States Food and Drug Administration (FDA).

Some studies based on the NICE recommendations consider “responders” to be those patients not presenting deterioration in cognitive function after at least 6 months of treatment. This definition of treatment response is established by equal or better score on the Mini-Mental State Examination (MMSE) or the Alzheimer's Disease Assessment Scale-Cognitive section (ADAS-Cog) scales as compared to baseline values, and an improvement in functional status as measured with the Katz Activities of Daily Living or the Lawton-Brody Instrumental Activities of Daily Living scales.26,27 Most studies focus on MMSE scores, although some use other tests to determine response to disease-modifying drugs (Table 3).28–30

Table 3.

Key aspects of pharmacogenetic studies evaluating clinical response to disease-modifying drugs for Alzheimer disease.

Drug  Time of treatment response evaluation  Tests used  Response criteria  ADR  Ref. 
DonepezilAverage of 9 months (range, 3-40) after treatment onset  MMSE, CDR, IADL, ADL, CIBIC-Plus  Changes in scores on the tests used  Evaluated but not specified  42 
6 months  ADAS-Cog, MMSE, CDR, IADL, ADL  Responders: no deterioration in cognition, according to ADAS-Cog and MMSE scores, or in functional status, according to ADL or IADL scores (NICE criteria)  Identified (nausea, vomiting, bradycardia, abdominal pain, diarrhoea, dizziness, orthostatic hypotension)  26,27 
36 weeks  ADAS-Cog, MMSE, IADL, CGIC  FDA recommendations  Headache, rhinitis, muscle cramps, and cholinergic system symptoms  52 
9 months (range, 3-40)  MMSE, CDR, ADL, CIBIC-Plus  Poorer MMSE score reflected cognitive deterioration.  Evaluated but not specified  54 
12-16 months  MMSE, verbal and visual memory, visual attention, and inductive reasoning  Responders: improvement or no change on neuropsychological tests  Not specified  55 
3 years  ADAS-Cog  Changes against baseline ADAS-Cog score  Not specified  56 
4, 8, and 12 weeks  ADAS-Cog  Responders: decreased ADAS-Cog score  Not specified  57 
48 weeks  ADAS-Cog  Smaller increase in ADAS-Cog score  Not specified  58 
12 months  MMSE  Responders: MMSE score equal, higher, or 1 point lower than at baseline  Not specified  59 
6 and 9 months  MMSE, CDT, IADL  Responders: improvement or no change in cognitive impairment and functional and behavioural improvement  Not specified  60 
6, 12, 18, 24, 30, and 36 months  MMSE, CDR-SB  Changes in MMSE and CDR-SB scores  Not specified  61 
Rivastigmine3 to 4 years  MMSE, ADAS-Cog, ADCS-ADL  Changes in assessment scale scores  Not specified  65 
26 weeks  ADAS-Cog, MMSE  Changes in ADAS-Cog score vs baseline  Anorexia, diarrhoea, nausea, vomiting  66 
6 months  MMSE, CDR-SB  Responders: equal or better MMSE and CDR-SB scores  Not specified  67 
Galantamine3 months  MMSE  Responders: equal or better MMSE score  Not specified  63 
3-12 months  DAD and ADAS-Cog  Responders: increase in ADAS-Cog score. Changes in DAD scores were also considered.  Not specified  64 
Donepezil, galantamine, and rivastigmine12 months  MMSE, ADL, IADL  Responders: equal or better MMSE score  Identified (epigastric pain, diarrhoea, irritability, vertigo, dizziness, abdominal pain, nausea, cardiovascular events, rash, depression, perspiration, hallucination, falls, agitation, insomnia)  68 
26 weeks  K-MMSE, CDR  Not specified  Evaluated but not specified  69 
2 years  MMSE  Not specified  Not specified  70 
Median 10.2 months (6-18)  MMSE  Responders: MMSE score equal, greater, or 1 point lower in the second assessment vs baseline  Not specified  71 
3-24 months  MMSE, Hachinski ischaemia score, Barthel Index for Activities of Daily Living, Global Deterioration Scale  Caregiver's impression of response  Evaluated but not specified  28 
Mean 15 months  MMSE  Responders: decrease in MMSE score  Not specified  72 
3, 6, and 12 months  MMSE  Responders: ≥ 2 point improvement on the MMSE at 12 months  Not specified  17 
6 months  MMSE  Responders: ≥ 2 point improvement on the MMSE between baseline and the second assessment  Evaluated but not specified  73 
6 months  MMSE  Responders: improvement or no change on the MMSE between baseline and the second assessment  Not specified  74 
Donepezil and rivastigmine1, 3, 9, and 15 months  MMSE  Not specified  Not specified  25 
2 years  SIB, ADCS-ADL, MMSE, NPI, GDS  Responders: equal or better scores on the SIB, NPI, GDS, or ADCS-ADL after 2 years’ treatment  Identified: vomiting, diarrhoea, nausea, anorexia, abdominal pain, falls, anxiety, agitation, dizziness, headache, urinary tract infection  29 
Rivastigmine and memantine16 weeks  ADAS-Cog, MMSE, FAB, CGA-NPI, ADCS-ADL  Responders: equal or better scores vs baseline  Not specified  30 
16 weeks  MMSE  Responders: cognitive improvement or absence of deterioration according to MMSE score  Not specified  75 

ADAS-Cog: Alzheimer's Disease Assessment Scale-Cognitive section; ADCS-ADL: Alzheimer's Disease Cooperative Study Activities of Daily Living; ADL: Activities of Daily Living; CDR-SB: Clinical Dementia Rating – Sum of Boxes; CDR: Clinical Dementia Rating; CDT: Clock Drawing Test; CGA-NPI: Caregiver-Administered Neuropsychiatric Inventory; CIBIC-Plus: Clinician's Interview-Based Impression of Change Plus caregiver input; FAB: Frontal Assessment Battery; FDA: United States Food and Drug Administration; GDS: Global Deterioration Scale; IADL: Instrumental Activities of Daily Living; K-MMSE: Korean-language version of the MMSE; MMSE: Mini-Mental State Examination; NICE: UK National Institute for Health and Care Excellence; NPI: Neuropsychiatric Inventory; ADR: adverse drug reaction; SIB: Severe Impairment Battery.

Numerous clinical tests are available for evaluating cognition and functional status2,31; Table 4 shows the main tests used in the pharmacogenetic studies reviewed. To avoid biased results, it is important when testing cognitive function to consider the following issues32:

  • -

    When using scales to determine AD severity, healthcare professionals should account for any learning difficulties, whether sensory or physical.

  • -

    Cognitive status assessment should not be based exclusively on patients’ scores on these scales if patients present any disability or linguistic/communication difficulty, are native speakers of a different language, or have a low level of schooling.

  • -

    MMSE scores are not sufficient for assessing disease severity and treatment response; complementary tests should be performed to assess multiple domains.

Table 4.

Main tests used to assess cognitive impairment and functional status in pharmacogenetic studies into Alzheimer disease.

Test  Description  Ref. 
Cognition
Mini–Mental State Examination (MMSE)  The most widely used brief cognitive assessment instrument, which includes tests of orientation, attention, calculation, recall, language, and visual construction. Its use is limited in illiterate populations. Age and level of schooling also influence global score.  77 
Alzheimer's Disease Assessment Scale-Cognitive section (ADAS-Cog)  70-point scale that measures alterations to memory, language, praxis, attention, and other cognitive skills affected in patients with AD  78,79 
Neurobehavioral Cognitive Status Examination (COGNISTAT)  Better detection of cognitive alterations than with the MMSE. With 5 tests, it assesses language, constructional ability, memory, calculation skills, and reasoning.  80 
Global changes
Global Deterioration Scale (GDS)  Clinical scale measuring behavioural, neuroanatomical, and neurophysiological changes. Describes the 7 characteristic phases of AD progression  81 
Clinical Dementia Rating (CDR)  5-point scale assessing 6 domains of cognitive and functional performance in patients with dementia: memory, orientation, judgement and problem solving, community affairs, home and hobbies, and personal care  82 
Functional status
Lawton-Brody Instrumental Activities of Daily Living (IADL) scale  Assesses activities that enable an individual to adapt to their surroundings, maintaining their independence  2,83 
Katz Activities of Daily Living (ADL) index  Assesses 6 basic activities of daily living; the results enable classification of patients as dependent or independent.  84 
Behaviour
Neuropsychiatric Inventory (NPI)  114-point scale in which caregivers are interviewed to assess the frequency and severity of such symptoms as hallucination, dysphoria, anxiety, agitation, aggressiveness, euphoria, disinhibition, irritability, and apathy  84 

AD: Alzheimer disease.

Plasma concentrations of drugs

Eight of the 33 pharmacogenetic studies identified determined plasma concentrations of disease-modifying drugs for AD in order to study the association of these values with the gene variants of interest. These studies include both acetylcholinesterase inhibitors and memantine. Table 5 summarises the methodologies followed by these articles.

Table 5.

Specifications of the methods used to determine plasma concentrations of disease-modifying drugs for Alzheimer disease in the pharmacogenetic studies reviewed.

Drug  Time between last administration of drug and sample collection  Type of sample  Analytical method used  Reference 
Donepezil12-15Blood  HPLC-UV  42,54 
Mean 14h (range, 1-28)  Blood  HPLC-MS  53 
Not specified  Blood  HPLC-ESI-MS/MS  59 
Galantamine  1-7Blood  HPLC-MS  62 
Rivastigmine  Not specified  Blood  MEKC-UV  67 
Memantine  0-24Blood  HPLC-MS  76 
Rivastigmine and memantine  Not specified  Blood  LC-MS/MS  75 

HPLC–ESI-MS/MS: high-performance liquid chromatography/electrospray ionisation tandem mass spectrometry; HPLC–MS: high-performance liquid chromatography/mass spectrometry; HPLC–UV: high-performance liquid chromatography and ultraviolet detection; LC–MS/MS: liquid chromatography/tandem mass spectrometry; MEKC-UV: micellar electrokinetic capillary chromatography with ultraviolet detection.

Plasma concentrations of disease-modifying drugs for AD are determined at steady state, which occurs at 4-5 half-lives.33 The reported half-life of donepezil (70-80hours) is significantly longer than those of other acetylcholinesterase inhibitors (0.3-12hours).34–36 Memantine also has a long half-life, at 60-80hours.37 Given these half-life values, we can expect a steady state to be reached by a maximum of 17 days after the administration of these drugs.

Liquid chromatography–mass spectrometry is the technique of choice in pharmacogenetic studies quantifying plasma levels of disease-modifying drugs for dementia. The advantages of this method are its sensitivity, specificity, the speed of testing, the small plasma volumes needed (250-500μL), and the ability to simultaneously quantify multiple drugs and some of their metabolites, in some cases.38–41

Important considerations in pharmacogenetic studies into Alzheimer disease

On account of the complexity of AD, pharmacogenetic studies also assess patients’ physical health, the presence of other diseases and concomitant treatments,42 and treatment adherence.26,27 These factors clearly have an important impact on treatment response and on plasma drug concentrations; therefore, they may interfere in the analysis of associations between these parameters and pharmacogenetic variants.

Studies evaluating treatment adherence in patients with AD report compliance ranging from 34% to 94%; the large disparities reported may be explained by the use of techniques supporting treatment adherence, such as a primary caregiver taking responsibility for administering the drugs.43 This variability may reflect a bias in the evaluation of treatment response. Numerous methods are available for evaluating treatment adherence,44 and using such measures would contribute to the reliability of the results.

Several articles report that approximately 50% of the elderly population studied present one or more chronic diseases.45,46 This may influence the evaluation of treatment response, as comorbidity is reported to complicate the clinical course of AD, for example by accelerating cognitive impairment and functional loss. Conditions affecting cognitive function in patients with dementia include heart failure, coronary artery disease, hypertension, diabetes, and chronic obstructive pulmonary disease.45,47

According to one study, primary care patients with dementia present an average of 2.4 chronic diseases and receive 5.1 different medications.48 Polymedication may strongly influence treatment response and concentrations of disease-modifying drugs for AD, particularly those metabolised by cytochromes, which are easily inhibited or induced by other drugs. However, acetylcholinesterase antagonists and the NMDA receptor antagonist memantine are thought to have little potential for drug-drug interactions.49 Despite this, recording concomitant treatments may be relevant in studies into AD drugs, as they may explain some variation in treatment response and the presence of adverse reactions.45

Immunotherapy with monoclonal antibodies

Novel approaches to AD treatment are based on the amyloid hypothesis, and involve the use of monoclonal antibodies that recognise different epitopes of Aβ peptide and display selective binding.50 Clinical trials with bapineuzumab, ponezumab, solanezumab, and gantenerumab have been suspended, while aducanumab is currently in phase III. This human antibody selectively targets Aβ aggregates (including soluble oligomers and insoluble fibrils), reducing Aβ plaques and slowing clinical progression in patients with mild or prodromal AD. In the phase Ib trial of aducanumab, which included 196 patients with AD, an important consideration was patient selection: presence of Aβ deposits had to be confirmed by positron emission tomography molecular imaging. The safety and tolerability of the antibody were acceptable, but an adverse reaction associated with elimination of Aβ was observed; this reaction was dose-dependent and occurred more frequently in carriers of the ɛ4 allele of APOE than in non-carriers. However, as this trial did not find sufficient efficacy to meet the exploratory clinical criteria, cognitive findings should be interpreted with caution.51

Conclusions

Few pharmacogenetic studies have been performed on drugs for AD. However, some studies report an association between variants of genes including CYP2D6, ABCB1, and BCHE and treatment response or plasma concentration of disease-modifying drugs for AD.

Performing this type of research in other populations may contribute further data to assist regulators in establishing more precise pharmacogenetic recommendations for the treatment of AD. For example, CYP2D6 is a pharmacogenetic biomarker recommended for multiple drugs; in AD, it may assist in the safe, effective use of acetylcholinesterase inhibitors, despite the fact that its use in this context remains under study. In this context, pharmacogenetics is a promising tool for identifying novel, safer, more efficacious treatments for AD, as well as supporting the development of drugs currently being researched.

None of the drugs developed for stopping the production or aggregation of Aβ or for promoting its elimination have been shown to be efficacious in the phase III clinical trials performed to date. Therefore, research into new therapeutic targets is needed, particularly in the preclinical stage of the disease.

Identifying pharmacogenetic biomarkers that may predict patient response to acetylcholinesterase inhibitors and memantine may contribute to therapeutic decision-making for these patients. In a context as complex as ageing, characterised by polymedication and comorbidity, the presence of pleiotropic effects may play a decisive role.

Funding

This project was funded by the Mexican National Council for Science and Technology (CONACYT). This article is part of CONACYT's Scientific Development Project to Address National Issues (proposal no. 3099; 2016), Dr Tirso Zúñiga Santamaría's postdoctorate fellowship (funded by CONACYT), and the grant for doctoral studies awarded to Ingrid Fricke-Galindo (grant no. CONACYT#369708).

Conflicts of interest

The authors have no conflicts of interest to declare.

References
[1]
A. Martin Prince, A. Wimo, M. Guerchet, M. Gemma-Claire Ali, Y.-T. Wu, M. Prina, et al.
World Alzheimer Report 2015. The global impact of dementia an analysis of prevalence, incidence, cost and trends.
Alzheimer's Disease International, (2015),
[2]
T. Zúniga, Z. Trujillo, G. Cortés, I. Acosta, A.L. Sosa.
Impacto de los programas de estimulación en adultos mayores con demencia que asisten a un centro de día.
Arch Neurocien, 19 (2014), pp. 192-198
[3]
P. Scheltens, K. Blennow, M.M. Breteler, B. de Strooper, G.B. Frisoni, S. Salloway, et al.
Alzheimer's disease.
[4]
WHO.
Dementia: a public health priority.
World Health Organization, (2012),
[5]
S. López Pousa.
Definición, prevalencia, incidencia y factores de riesgo de la enfermedad de Alzheimer.
Enfermedad de Alzheimer y otras Demencias, pp. 143-150
[6]
J. Cummings, P.S. Aisen, B. DuBois, L. Frölich, C.R. Jack, R.W. Jones, et al.
Drug development in Alzheimer's disease: the path to 2025.
Alzheimers Res Ther, 8 (2016), pp. 39
[7]
B. Dubois, H. Hampel, H.H. Feldman, P. Scheltens, P. Aisen, S. Andrieu, et al.
Preclinical Alzheimer's disease: definition, natural history, and diagnostic criteria.
Alzheimer's Dement, 12 (2016), pp. 292-323
[8]
J.C. Morris, K. Blennow, L. Froelich, A. Nordberg, H. Soininen, G. Waldemar, et al.
Harmonized diagnostic criteria for Alzheimer's disease: recommendations.
J Intern Med, 275 (2014), pp. 204-213
[9]
C.-C. Liu, C.-C. Liu, T. Kanekiyo, H. Xu, G. Bu.
Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy.
Nat Rev Neurol, 9 (2013), pp. 106-118
[10]
G. McKhann, D. Drachman, M. Folstein, R. Katzman, D. Price, E.M. Stadlan.
Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease.
Neurology, 34 (1984), pp. 939-944
[11]
B. Dubois, H.H. Feldman, C. Jacova, S.T. DeKosky, P. Barberger-Gateau, J. Cummings, et al.
Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS–ADRDA criteria.
Lancet Neurol, 6 (2007), pp. 734-746
[12]
G.M. McKhann, D.S. Knopman, H. Chertkow, B.T. Hyman, C.R. Jack, C.H. Kawas, et al.
The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease.
Alzheimer's Dement, 7 (2011), pp. 263-269
[13]
M.E. Alonso Vilatela, M. López-López, P. Yescas-Gómez.
Genetics of Alzheimer's disease.
Arch Med Res, 43 (2012), pp. 622-631
[14]
C.T. Loy, P.R. Schofield, A.M. Turner, J.B.J. Kwok.
Genetics of dementia.
[15]
C. Ballard, Z. Khan, H. Clack, A. Corbett.
Nonpharmacological treatment of Alzheimer disease.
Can J Psychiatry, 56 (2011), pp. 589-595
[16]
J. Olazarán, B. Reisberg, L. Clare, I. Cruz, J. Peña-Casanova, T. del Ser, et al.
Nonpharmacological therapies in Alzheimer's disease: a systematic review of efficacy.
Dement Geriatr Cogn Disord, 30 (2010), pp. 161-178
[17]
L.F. Miranda, K.B. Gomes, J.N. Silveira, G.A. Pianetti, R.M. Byrro, P.R. Peles, et al.
Predictive factors of clinical response to cholinesterase inhibitors in mild and moderate Alzheimer's disease and mixed dementia: a one-year naturalistic study.
J Alzheimers Dis, 45 (2015), pp. 609-620
[18]
M. Bond, G. Rogers, J. Peters, R. Anderson, M. Hoyle, A. Miners, et al.
The effectiveness and cost-effectiveness of donepezil, galantamine, rivastigmine and memantine for the treatment of Alzheimer's disease (review of Technology Appraisal No 111): a systematic review and economic model.
Health Technol Assess (Rockv), 16 (2012), pp. 1-470
[19]
A. Clegg, J. Bryant, T. Nicholson, L. McIntyre, S. de Broe, K. Gerard, et al.
Clinical and cost-effectiveness of donepezil, rivastigmine and galantamine for Alzheimer's disease: a rapid and systematic review.
Health Technol Assess, 5 (2001), pp. 1-137
[20]
R.A. Hansen, G. Gartlehner, A.P. Webb, L.C. Morgan, C.G. Moore, D.E. Jonas.
Efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer's disease: a systematic review and meta-analysis.
Clin Interv Aging, 3 (2008), pp. 211-225
[21]
R. Cacabelos.
Donepezil in Alzheimer's disease: from conventional trials to pharmacogenetics.
Neuropsychiatr Dis Treat, 3 (2007), pp. 303-333
[22]
C. Campos, N.B. Rocha, R.T. Vieira, S.A. Rocha, D. Telles-Correia, F. Paes, et al.
Treatment of cognitive deficits in Alzheimer's disease: a psychopharmacological review.
Psychiatr Danub, 28 (2016), pp. 2-12
[23]
J. Patocka, K. Kuca, D. Jun.
Acetylcholinesterase and butyrylcholinesterase – important enzymes of human body.
Acta Medica (Hradec Kralove), 47 (2004), pp. 215-228
[24]
K. Ishiwata, K. Kawamura, K. Yanai, N.H. Hendrikse.
In vivo evaluation of P-glycoprotein modulation of 8 PET radioligands used clinically.
J Nucl Med, 48 (2007), pp. 81-87
[25]
R. Scacchi, G. Gambina, G. Moretto, R.M. Corbo.
Variability of AChE, BChE, and ChAT genes in the late-onset form of Alzheimer's disease and relationships with response to treatment with donepezil and rivastigmine.
Am J Med Genet B Neuropsychiatr Genet, 150B (2009), pp. 502-507
[26]
A. Pilotto, M. Franceschi, G. D’Onofrio, A. Bizzarro, F. Mangialasche, L. Cascavilla, et al.
Effect of a CYP2D6 polymorphism on the efficacy of donepezil in patients with Alzheimer disease.
Neurology, 73 (2009), pp. 761-767
[27]
D. Seripa, A. Bizzarro, A. Pilotto, G. D’onofrio, G. Vecchione, A.P. Gallo, et al.
Role of cytochrome P4502D6 functional polymorphisms in the efficacy of donepezil in patients with Alzheimer's disease.
Pharmacogenet Genomics, 21 (2011), pp. 225-230
[28]
C.E. Patterson, S.A. Todd, A.P. Passmore.
Effect of apolipoprotein E and butyrylcholinesterase genotypes on cognitive response to cholinesterase inhibitor treatment at different stages of Alzheimer's disease.
Pharmacogenomics J, 11 (2011), pp. 444-450
[29]
R. Blesa, R. Bullock, Y. He, H. Bergman, G. Gambina, J. Meyer, et al.
Effect of butyrylcholinesterase genotype on the response to rivastigmine or donepezil in younger patients with Alzheimer's disease.
Pharmacogenet Genomics, 16 (2006), pp. 771-774
[30]
H.J. Han, J.C. Kwon, J.E. Kim, S.G. Kim, J.-M. Park, K.W. Park, et al.
Effect of rivastigmine or memantine add-on therapy is affected by butyrylcholinesterase genotype in patients with probable Alzheimer's disease.
Eur Neurol, 73 (2015), pp. 23-28
[31]
R.A. Hansen, G. Gartlehner, D.J. Kaufer, K.N. Lohr, T. Carey.
Drug class review: Alzheimer's drugs.
Oregon Health & Science University, (2006),
[32]
Donepezil, galantamine, rivastigmine and memantine for the treatment of Alzheimer's disease: 1-Guidance. Guidance and guidelines. NICE. Available from: https://www.nice.org.uk/guidance/ta217/chapter/1-Guidance [accessed 06.09.16].
[33]
S. Ito, M. Frcpc.
Pharmacokinetics 101.
Paediatr Child Heal, 16 (2011), pp. 535-536
[34]
A. Nordberg, A.L. Svensson.
Cholinesterase inhibitors in the treatment of Alzheimer's disease: a comparison of tolerability and pharmacology.
[35]
M. Shigeta, A. Homma.
Donepezil for Alzheimer's disease: pharmacodynamic, pharmacokinetic, and clinical profiles.
[36]
J.J. Sramek, E.J. Frackiewicz, N.R. Cutler.
Review of the acetylcholinesterase inhibitor galanthamine.
Expert Opin Investig Drugs, 9 (2000), pp. 2393-2402
[37]
R. Boinpally, L. Chen, S.R. Zukin, N. McClure, R.K. Hofbauer, A. Periclou.
A novel once-daily fixed-dose combination of memantine extended release and donepezil for the treatment of moderate to severe Alzheimer's disease: two phase I studies in healthy volunteers.
Clin Drug Investig, 35 (2015), pp. 427-435
[38]
M. Noetzli, N. Ansermot, M. Dobrinas, C.B. Eap.
Simultaneous determination of antidementia drugs in human plasma: procedure transfer from HPLC–MS to UPLC–MS/MS.
J Pharm Biomed Anal, 64–65 (2012), pp. 16-25
[39]
E.J. Park, H.W. Lee, H.Y. Ji, H.Y. Kim, M.H. Lee, E.-S. Park, et al.
Hydrophilic interaction chromatography-tandem mass spectrometry of donepezil in human plasma: application to a pharmacokinetic study of donepezil in volunteers.
Arch Pharm Res, 31 (2008), pp. 1205-1211
[40]
B.N. Patel, N. Sharma, M. Sanyal, P.S. Shrivastav.
Quantitation of donepezil and its active metabolite 6-O-desmethyl donepezil in human plasma by a selective and sensitive liquid chromatography–tandem mass spectrometric method.
Anal Chim Acta, 629 (2008), pp. 145-157
[41]
N.R. Pilli, J.K. Inamadugu, N. Kondreddy, V.K. Karra, R. Damaramadugu, JVLN.S. Rao.
A rapid and sensitive LC–MS/MS method for quantification of donepezil and its active metabolite, 6-o-desmethyl donepezil in human plasma and its pharmacokinetic application.
Biomed Chromatogr, 25 (2011), pp. 943-951
[42]
F. Varsaldi, G. Miglio, M.G. Scordo, M.-L. Dahl, L.M. Villa, A. Biolcati, 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, 62 (2006), pp. 721-726
[43]
C.J. Maxwell, K. Stock, D. Seitz, N. Herrmann.
Persistence and adherence with dementia pharmacotherapy: relevance of patient, provider, and system factors.
Can J Psychiatry, 5959 (2014), pp. 624-631
[44]
L. Osterberg, T. Blaschke.
Adherence to medication.
N Engl J Med, 353 (2005), pp. 487-497
[45]
A. Clodomiro, P. Gareri, G. Puccio, F. Frangipane, R. Lacava, A. Castagna, et al.
Somatic comorbidities and Alzheimer's disease treatment.
Neurol Sci, 34 (2013), pp. 1581-1589
[46]
A. Marengoni, B. Winblad, A. Karp, L. Fratiglioni.
Prevalence of chronic diseases and multimorbidity among the elderly population in Sweden.
Am J Public Health, 98 (2008), pp. 1198-1200
[47]
A. Solomon, L. Dobranici, I. Kåreholt, C. Tudose, M. Lăzărescu.
Comorbidity and the rate of cognitive decline in patients with Alzheimer dementia.
Int J Geriatr Psychiatry, 26 (2011), pp. 1244-1251
[48]
C.C. Schubert, M. Boustani, C.M. Callahan, A.J. Perkins, C.P. Carney, C. Fox, et al.
Comorbidity profile of dementia patients in primary care: are they sicker?.
J Am Geriatr Soc, 54 (2006), pp. 104-109
[49]
R.H. Levy, C. Collins.
Risk and predictability of drug interactions in the elderly.
Int Rev Neurobiol, 81 (2007), pp. 235-251
[50]
J. Sevigny, P. Chiao, T. Bussière, P.H. Weinreb, L. Williams, M. Maier, et al.
The antibody aducanumab reduces Aβ plaques in Alzheimer's disease.
Nature, 537 (2016), pp. 50-56
[51]
S. Budd Haeberlein, J. O’Gorman, P. Chiao, T. Bussière, P. von Rosenstiel, Y. Tian, et al.
Clinical development of aducanumab, an anti-Aβ human monoclonal antibody being investigated for the treatment of early Alzheimer's disease.
J Prev Alzheimer's Dis, 4 (2017), pp. 255-263
[52]
A.-S. Rigaud, L. Traykov, F. Latour, R. Couderc, F. Moulin, F. Forette.
Presence or absence of at least one epsilon 4 allele and gender are not predictive for the response to donepezil treatment in Alzheimer's disease.
Pharmacogenetics, 12 (2002), pp. 415-420
[53]
M. Noetzli, M. Guidi, K. Ebbing, S. Eyer, L. Wilhelm, A. Michon, et al.
Population pharmacokinetic approach to evaluate the effect of CYP2D6, CYP3A, ABCB1, POR and NR1I2 genotypes on donepezil clearance.
Br J Clin Pharmacol, 78 (2014), pp. 135-144
[54]
L. Magliulo, M.-L. Dahl, G. Lombardi, S. Fallarini, L.M. Villa, A. Biolcati, et al.
Do CYP3A and ABCB1 genotypes influence the plasma concentration and clinical outcome of donepezil treatment?.
Eur J Clin Pharmacol, 67 (2011), pp. 47-54
[55]
A. Bizzarro, C. Marra, A. Acciarri, A. Valenza, F.D. Tiziano, C. Brahe, et al.
Apolipoprotein E ɛ4 allele differentiates the clinical response to donepezil in Alzheimer's disease.
Dement Geriatr Cogn Disord, 20 (2005), pp. 254-261
[56]
L. De Beaumont, S. Pelleieux, L. Lamarre-Théroux, D. Dea, J. Poirier, Alzheimer's Disease Cooperative Study.
Butyrylcholinesterase K and apolipoprotein E-ɛ4 reduce the age of onset of Alzheimer's disease, accelerate cognitive decline, and modulate donepezil response in mild cognitively impaired subjects.
J Alzheimer's Dis, 54 (2016), pp. 913-922
[57]
J.F. Waring, Q. Tang, W.Z. Robieson, D.P. King, U. Das, J. Dubow, et al.
APOE-ɛ4 carrier status and donepezil response in patients with Alzheimer's disease.
J Alzheimer's Dis, 47 (2015), pp. 137-148
[58]
S.H. Choi, S.Y. Kim, H.R. Na, B.-K. Kim, D.W. Yang, J.C. Kwon, et al.
Effect of ApoE genotype on response to donepezil in patients with Alzheimer's disease.
Dement Geriatr Cogn Disord, 25 (2008), pp. 445-450
[59]
L.F. Miranda, K.B. Gomes, P.A. Tito, J.N. Silveira, G.A. Pianetti, R.M. Byrro, et al.
Clinical response to donepezil in mild and moderate dementia: relationship to drug plasma concentration and CYP2D6 and APOE genetic polymorphisms.
J Alzheimer's Dis, 55 (2017), pp. 539-549
[60]
A. Klimkowicz-Mrowiec, A. Slowik, P. Wolkow, A. Malgorzata Sado, J. Pera, T. Dziedzic, et al.
Influence of rs1080985 single nucleotide polymorphism of the CYP2D6 gene on response to treatment with donepezil in patients with Alzheimer's disease.
Neuropsychiatr Dis Treat, 9 (2013), pp. 1029
[61]
S. Sokolow, X. Li, L. Chen, K.D. Taylor, J.I. Rotter, R.A. Rissman, et al.
Deleterious effect of butyrylcholinesterase K-variant in donepezil treatment of mild cognitive impairment.
J Alzheimers Dis, 56 (2017), pp. 229-237
[62]
M. Noetzli, M. Guidi, K. Ebbing, S. Eyer, S. Zumbach, P. Giannakopoulos, et al.
Relationship of CYP2D6, CYP3A, POR, and ABCB1 genotypes with galantamine plasma concentrations.
Ther Drug Monit, 35 (2013), pp. 270-275
[63]
S.H. MacGowan, G.K. Wilcock, M. Scott.
Effect of gender and apolipoprotein E genotype on response to anticholinesterase therapy in Alzheimer's disease.
[64]
J. Aerssens, P. Raeymaekers, S. Lilienfeld, H. Geerts, F. Konings, W. Parys.
APOE genotype: no influence on galantamine treatment efficacy nor on rate of decline in Alzheimer's disease.
Dement Geriatr Cogn Disord, 12 (2001), pp. 69-77
[65]
S. Ferris, A. Nordberg, H. Soininen, T. Darreh-Shori, R. Lane.
Progression from mild cognitive impairment to Alzheimer's disease: effects of sex, butyrylcholinesterase genotype, and rivastigmine treatment.
Pharmacogenet Genomics, 19 (2009), pp. 635-646
[66]
M. Farlow, R. Lane, S. Kudaravalli, Y. He.
Differential qualitative responses to rivastigmine in APOE epsilon 4 carriers and noncarriers.
Pharmacogenomics J, 4 (2004), pp. 332-335
[67]
T.-H. Chen, M.-C. Chou, C.-L. Lai, S.-J. Wu, C.-L. Hsu, Y.-H. Yang.
Factors affecting therapeutic response to rivastigmine in Alzheimer's disease patients in Taiwan.
Kaohsiung J Med Sci, 33 (2017), pp. 277-283
[68]
C. Chianella, D. Gragnaniello, P. Maisano Delser, M.F. Visentini, E. Sette, M.R. Tola, et al.
BCHE and CYP2D6 genetic variation in Alzheimer's disease patients treated with cholinesterase inhibitors.
Eur J Clin Pharmacol, 67 (2011), pp. 1147-1157
[69]
H. Yoon, W. Myung, S.-W.W. Lim, H.S. Kang, S. Kim, H.-H. Won, et al.
Association of the choline acetyltransferase gene with responsiveness to acetylcholinesterase inhibitors in Alzheimer's disease.
Pharmacopsychiatry, 48 (2015), pp. 111-117
[70]
I.L.S. Braga, P.N. Silva, T.K. Furuya, L.C. Santos, B.C. Pires, D.R. Mazzotti, et al.
Effect of APOE and CHRNA7 genotypes on the cognitive response to cholinesterase inhibitor treatment at different stages of Alzheimer's disease.
Am J Alzheimers Dis Other Demen, 30 (2015), pp. 139-144
[71]
F. Martinelli-Boneschi, G. Giacalone, G. Magnani, G. Biella, E. Coppi, R. Santangelo, et al.
Pharmacogenomics in Alzheimer's disease: a genome-wide association study of response to cholinesterase inhibitors.
[72]
D. Harold, S. Macgregor, C.E. Patterson, P. Hollingworth, P. Moore, M.J. Owen, et al.
A single nucleotide polymorphism in CHAT influences response to acetylcholinesterase inhibitors in Alzheimer's disease.
Pharmacogenet Genomics, 16 (2006), pp. 75-77
[73]
P.-H. Weng, J.-H. Chen, T.-F. Chen, Y. Sun, L.-L. Wen, P.-K. Yip, et al.
CHRNA7 polymorphisms and response to cholinesterase inhibitors in Alzheimer's disease.
[74]
F. Clarelli, E. Mascia, R. Santangelo, S. Mazzeo, G. Giacalone, D. Galimberti, et al.
CHRNA7 gene and response to cholinesterase inhibitors in an Italian cohort of Alzheimer's disease patients.
J Alzheimer's Dis, 52 (2016), pp. 1203-1208
[75]
N. Sonali, M. Tripathi, R. Sagar, T. Velpandian, V. Subbiah.
Clinical effectiveness of rivastigmine monotherapy and combination therapy in Alzheimer's patients.
CNS Neurosci Ther, 19 (2013), pp. 91-97
[76]
M. Noetzli, M. Guidi, K. Ebbing, S. Eyer, L. Wilhelm, A. Michon, et al.
Population pharmacokinetic study of memantine: effects of clinical and genetic factors.
Clin Pharmacokinet, 52 (2013), pp. 211-223
[77]
M.F. Folstein, S.E. Folstein, P.R. McHugh.
“Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician.
J Psychiatr Res, 12 (1975), pp. 189-198
[78]
W.G. Rosen, R.C. Mohs, K.L. Davis.
A new rating scale for Alzheimer's disease.
Am J Psychiatry, 141 (1984), pp. 1356-1364
[79]
E. Kolibas, V. Korinkova, V. Novotny, K. Vajdickova, D. Hunakova.
ADAS-cog (Alzheimer's Disease Assessment Scale-cognitive subscale) – validation of the Slovak version.
Bratisl Lekárske List, 101 (2000), pp. 598-602
[80]
R.J. Kiernan, J. Mueller, J.W. Langston, C. van Dyke.
The neurobehavioral cognitive status examination: a brief but quantitative approach to cognitive assessment.
Ann Intern Med, 107 (1987), pp. 481-485
[81]
B. Reisberg, S.H. Ferris, M.J. de Leon, T. Crook.
The Global Deterioration Scale for assessment of primary degenerative dementia.
Am J Psychiatry, 139 (1982), pp. 1136-1139
[82]
C.P. Hughes, L. Berg, W.L. Danziger, L.A. Coben, R.L. Martin.
A new clinical scale for the staging of dementia.
Br J Psychiatry, 140 (1982), pp. 566-572
[83]
M.P. Lawton, E.M. Brody.
Assessment of older people: self-maintaining and instrumental activities of daily living.
Gerontologist, 9 (1969), pp. 179-186
[84]
S. Katz, T.D. Downs, H.R. Cash, R.C. Grotz.
Progress in development of the index of ADL.
Gerontologist, 10 (1970), pp. 20-30
[85]
J.L. Cummings, M. Mega, K. Gray, S. Rosenberg-Thompson, D.A. Carusi, J. Gornbein.
The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia.
Neurology, 44 (1994), pp. 2308-2314

Please cite this article as: Zúñiga Santamaría T, Yescas Gómez P, Fricke Galindo I, González González M, Ortega Vázquez A, López López M. Estudios farmacogenéticos en la enfermedad de Alzheimer. Neurología. 2022;37:287–303.

Copyright © 2018. Sociedad Española de Neurología
Article options
Tools
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.nrleng.2019.11.006
No mostrar más