Scolaris Content Display Scolaris Content Display

Estatinas para el tratamiento de la demencia

Collapse all Expand all

Resumen

Antecedentes

Ha sido poco explorado el uso de estatinas para el tratamiento de la enfermedad de Alzheimer (EA) establecida o la demencia vascular (DV). En la EA, la proteína amiloide β (Aβ) se deposita en forma de placas extracelulares y estudios anteriores han determinado que la generación Aβ es dependiente del colesterol. La hipercolesterolemia también ha sido implicada en la patogenia de la demencia vascular. Debido a la función de las estatinas en la reducción del colesterol es biológicamente posible que puedan ser efectivas en el tratamiento de la EA y la DV.

Objetivos

Evaluar la eficacia clínica y tolerabilidad de las estatinas en el tratamiento de la EA y la DV. Evaluar si la eficacia de las estatinas en el tratamiento de la EA y la DV depende del nivel de colesterol, el genotipo de la ApoE o el nivel cognitivo.

Métodos de búsqueda

Se realizaron búsquedas en ALOIS, el Registro Especializado del Grupo Cochrane de Demencia y Trastornos Cognitivos (Cochrane Dementia and Cognitive Improvement Group), The Cochrane Library, MEDLINE, EMBASE, PsycINFO, CINAHL y LILACS, así como en muchos registros de ensayos y fuentes de literatura gris (20 de enero de 2014).

Criterios de selección

Ensayos controlados con asignación aleatoria y doble ciego de las estatinas administradas durante al menos seis meses en pacientes con diagnóstico de demencia.

Obtención y análisis de los datos

Dos autores independientes extrajeron y evaluaron los datos sobre la base de los criterios de inclusión. Los datos se agruparon cuando fue apropiado y se ingresaron en un metanálisis. Se utilizaron los procedimientos metodológicos estándar previstos por la Colaboración Cochrane.

Resultados principales

Se identificaron cuatro estudios (1154 participantes, con edades comprendidas entre los 50 y los 90 años). Todos los participantes tenían un diagnóstico de probable o posible EA de acuerdo con criterios estándar y la mayoría de los participantes se establecieron con un inhibidor de colinesterasa. El resultado principal en todos los estudios fue el cambio en la Escala de Evaluación de la Enfermedad de Alzheimer ‐ subescala cognitiva (ADAS‐Cog) de la línea de base. Cuando se agruparon los datos, no hubo un beneficio significativo de la estatina (diferencia de medias ‐0,26, intervalo de confianza (IC) del 95% ‐1,05 a 0,52, valor de P = 0,51). Todos los estudios proporcionaron un cambio en la Mini Mental State Examination (Mini Examen del Estado Mental) (MMSE) a partir del valor inicial. No hubo un beneficio significativo de las estatinas en el MMSE cuando se agruparon los datos (diferencia de medias ‐0,32, IC del 95%: ‐0,71 a 0,06, valor de P = 0,10). Tres estudios informaron sobre los efectos adversos relacionados con el tratamiento. Cuando se agruparon los datos, no hubo diferencias significativas entre las estatinas y el placebo (odds‐ratio 1,09, IC del 95%: 0,58 a 2,06, valor P = 0,78). No hubo diferencias significativas en el comportamiento, la función global o las actividades de la vida diaria en los grupos de estatinas y de placebo. Se evaluó el riesgo de sesgo como bajo para todos los estudios. No se encontraron estudios que evaluaran el papel de las estatinas en el tratamiento de la DV.

Conclusiones de los autores

Los análisis de los estudios disponibles, incluidos dos grandes ensayos controlados aleatorizados, indican que las estatinas no tienen ningún beneficio en las medidas de resultado primarias de la ADAS‐Cog o el MMSE.

Resumen en términos sencillos

Estatinas para el tratamiento de la enfermedad de Alzheimer

Antecedentes

Se cree que los altos niveles de colesterol (una sustancia grasa conocida como lípido) en la sangre contribuyen a la causa de la enfermedad de Alzheimer y la demencia vascular. La familia de medicamentos de la estatina (lovastatina, pravastatina, simvastatina, y otros) son medicaciones potentes para la disminución del colesterol y son tratamientos de primera línea para la reducción del colesterol en pacientes con, o en riesgo de enfermedades cardiovasculares. Ha habido mucho interés en el posible papel de las estatinas en el tratamiento de la demencia.

Características de los estudios

Dos autores independientes buscaron en las bases de datos científicas estudios en los que se administró una estatina o un placebo (un tratamiento simulado) durante al menos seis meses. Se incluyeron a personas con un probable o posible diagnóstico de la enfermedad de Alzheimer según los criterios clínicos estándar. Los hallazgos estuvieron vigentes hasta enero de 2014.

Resultados clave

Se identificaron cuatro estudios con 1.154 participantes (de edades comprendidas entre los 50 y los 90 años). Los estudios utilizaron pruebas estándar para evaluar la gravedad de la enfermedad de Alzheimer. De estos ensayos, incluidos dos grandes ensayos, no se encontró evidencia de que las estatinas ayuden en el tratamiento del deterioro cognitivo en la demencia.

Calidad de la evidencia

Se considera que la calidad de la evidencia es alta, ya que se han incluido dos grandes ensayos controlados aleatorizados junto con otros dos más pequeños.

Authors' conclusions

Implications for practice

There is no evidence to recommend statins for the treatment of Alzheimer's disease (AD) or dementia. In LEADe 2010, the first large‐scale randomised controlled trial (RCT) evaluating statins as a treatment for mild‐to‐moderate AD, the regimen of atorvastatin plus donepezil was not associated with significant benefit on clinical outcome measures over 72 weeks. Sano 2011 was a further well‐designed large RCT incorporating the regimen of simvastatin versus placebo. There again was no benefit from the statin in terms of cognition despite a lowering of cholesterol in the active treatment group. When data from these trials were pooled with two smaller‐scale studies (ADCLT 2005; Simons 2002), there was no benefit from statins seen with the primary outcome measure Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog) or in Mini Mental State Examination (MMSE).

From ADCLT 2005, there was some evidence that atorvastatin treatment was more beneficial at six months in patients with AD with higher MMSE at baseline, patients with an apolipoprotein E4 allele and higher cholesterol levels at baseline. This was not replicated in Sano 2011, where there was no effect on MMSE score or ApoE genotype at baseline on the primary outcome measure.

Implications for research

Two large‐scale RCTs and two smaller studies have assessed the effect of statins on the treatment of dementia. Combined data from the four studies and individual results from the large‐scale RCTs were negative. If further RCTs are to be carried out, lipophilic statins should be used due to their blood‐brain barrier permeability and patients with only mild cognitive impairment should be recruited.

The question of whether statins are of benefit in the treatment of vascular dementia remains unanswered and would be an avenue of research worth pursuing.

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Statins compared with placebo for the treatment of dementia

Patients: patients with dementia

Setting: community

Intervention: statin medication

Comparison: placebo

Outcomes

Absolute difference in means

(95% CI)

Number of participants

(studies)

GRADE Quality of evidence

Comments

Change in ADAS‐Cog

‐0.26

(‐1.05 to 0.52)

1110

(4)

High

Change in MMSE

‐0.32

(‐0.71 to 0.06)

1127

(4)

High

Change in CGIC

‐0.02

(‐0.14 to 0.10)

660

(2)

High

Change in NPI

‐1.11

(‐2.10 to ‐0.12)

983

(3)

High

ADAS‐Cog: Alzheimer's Disease Assessment Scale ‐ cognitive subscale; CGIC: Clinical Global Impression of Change; CI: confidence interval; MMSE: Mini Mental State Examination; NPI: Neuropsychiatric Inventory.

GRADE Working Group grades of evidence:

Hign quality: Further research is very unlikely to change our confidence in the estimate of effect

Moderate quality: Further evidence is likely to have an important impact on our confidence in the estimate of effect and may change the estimate

Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate

Very low quality: We are very uncertain about the estimate

Background

Alzheimer's disease (AD) is the most common cause of dementia, accounting for 50% to 60% of all cases. Cerebrovascular disease is the second most common cause, responsible for 25% to 30% of cases and resulting in vascular dementia (VaD). AD and VaD also frequently co‐occur leading to mixed dementia. Dementia is thought to affect approximately 7% of the population older than 65 years of age and 30% of people older than 80 years of age. Other studies have quoted that 10% of people aged over 65 years are affected, rising to nearly 50% of all people aged over 85 years (Evans 1989; von Strauss 1999). Dementia is already a major public health problem and is set to become even more so due to the anticipated increase in life expectancy. As of 2013, there were an estimated 44.4 million people with dementia worldwide. This number will increase to an estimated 75.6 million in 2030 and 135.5 million in 2050 (Alzheimers Disease International 2013). Prevalence estimates from the English Cognitive Function and Ageing Study Ⅰ and Ⅱ in people aged over 65 years were 8.3% and 6.5%, respectively (Matthews 2013).

There is accumulating evidence that cholesterol may be implicated in the pathogenesis of dementia (AD and VaD) and this has led investigators to assess the possible role of lipid‐lowering agents (LLA) in the treatment of dementia. However, many questions remain unanswered. This review aimed to collate the best evidence available regarding use of statins in the treatment of dementia.

Cholesterol and Alzheimer's disease

A possible role for cholesterol in AD was based on observations of AD neuropathology among relatively young individuals with no history of dementia but with coronary heart disease (Sparks 1990). A central event in the development of AD is thought to be the abnormal processing of the cell membrane‐associated amyloid precursor protein (APP) followed by deposition of toxic β‐amyloid (Aβ) protein in the form of amyloid plaques in the extracellular space of the neocortex (Selkoe 2001). APP is a protein containing 770 amino acids. Aβ peptide is generated by the sequential cleavage of APP by beta and gamma secretase in the amyloidogenic pathway. Aβ genesis may be precluded if APP is instead cleaved first by alpha secretase within the Aβ domain, and then by gamma secretase, forming a non‐amyloidogenic fragment (Cole 2007). The non‐amyloidogenic pathway appears to be neuroprotective compared to the neurodegenerative, amyloidogenic pathway (Vetrivel 2006). Aβ occurs in two different forms, Aβ40 and Aβ42, varying in the length at the C terminus. It is the longer Aβ42 that aggregates more avidly. Early work discovered that elevated cholesterol levels led to greatly reduced levels of non‐amyloidogenic APP alpha in vitro (Bodovitz 1996). Perhaps more importantly, subsequent studies suggested that cerebral Aβ generation in vitro (Mizuno 1999; Simons 1998), and in vivo (Burns 2003; Refolo 2000; Sparks 1994), is cholesterol dependent. Cell biology investigations indicated that specialised cellular membrane microdomains rich in cholesterol and sphingolipids, termed lipid rafts, might be the link between cholesterol and amyloidogenic processing of APP. Both beta and gamma secretases are active in lipid rafts and it appears that APP processing within these lipid rafts by secretases determines the levels of Aβ production (Ehehalt 2003; Vetrivel 2004).

The ApoE ϵ4 allele is associated with sporadic AD. Meta‐analysis has shown that the ApoE ϵ4 allele increases the risk of the disease by three times in heterozygotes and by 15 times in homozygotes (Farrer 1997). It acts mainly by modifying age of onset, with each copy of the allele lowering the age at onset by almost 10 years (Corder 1993). This is significant in the context of cholesterol metabolism as ApoE acts as a cholesterol transporter in the brain. It binds directly to the Aβ peptide and influences its fibrillogenesis and clearance in vitro (Strittmatter 1993), and in vivo (Naslund 1995; Wisniewski 1995). ApoE is critically important for the formation of fibrillar Aβ in brain parenchyma in vivo (Holtzman 2000). Two genome‐wide association studies reported a significant association of AD with a locus within the clusterin (CLU) gene (Harold 2009; Lambert 2009). Functionally, clusterin has similarities to ApoE as both are major brain apolipoproteins and act as cholesterol transporters in the central nervous system. Both are also present in amyloid plaques and interact with Aβ, and regulating the conversion of Aβ into insoluble forms, co‐operating to suppress Aβ deposition and modifying Aβ clearance at the blood‐brain barrier (BBB) (van Es 2009). The amyloid cascade hypothesis states that an imbalance between production of and clearance of Aβ in the brain is the initiating event in the pathogenesis of AD, ultimately leading to neuronal degeneration and dementia (Hardy 2002), and thus theoretically links cholesterol metabolism to the development of AD.

There is also emerging evidence that genetics may play a role in responsiveness to statins. See Sabbagh 2012 for a review of findings in which new polymorphisms have been discovered that influence statin responsiveness. These have not been explored in AD and may aid in a priori identifying those most likely to benefit from treatment.

Central and peripheral cholesterol pools are separate, however, and almost all cholesterol in the brain is synthesised locally and is not transferred into plasma because of the BBB (Dietschy 2001). How serum cholesterol affects brain cholesterol has been a major question to date. Brain cholesterol content does not seem to be affected by high serum low‐density lipoprotein cholesterol (LDL‐C) or low serum high‐density lipoprotein cholesterol (HDL‐C) levels, perhaps as a result of the stability of cholesterol in myelin; however, it has not been established whether intramembranous lipid domains or intracellular cholesterol content are affected (Lane 2005). Studies in animal models have shown that diet‐induced hypercholesterolaemia increases Aβ and ApoE concentrations in the temporal and frontal cortices, but not in the cerebellum, and that these regional increases parallel the amyloid pathology observed in the AD brain (Wu 2003). Side chain‐oxidised cholesterol metabolites, such as hydroxy‐cholesterols, do cross the BBB. In the steady state in the adult brain, cholesterol clearance is facilitated by the formation and excretion of 24‐hydroxycholesterol (Lutjohann 2000). This is the major pathway for efflux of brain cholesterol and is crucial for maintenance of brain cholesterol homeostasis (Reiss 2004). 27‐Hydroxycholesterol is also found in the brain and may also provide a link between hypercholesterolaemia and AD (Heverin 2005); the contribution of the 27‐hydroxylase pathway to AD is an area in need of further exploration.

Several epidemiological studies have shown an association between high serum cholesterol levels and an increased susceptibility to AD (Jarvik 1995; Kivipelto 2002; Notkola 1998). The Notkola study was a long‐term prospective study that found elevated total serum cholesterol level was a risk factor for AD, independent of the ApoE ϵ4 allele; however, the association between AD and the ApoE ϵ4 allele became weaker after adjustment for serum total cholesterol (Notkola 1998). The authors concluded that some of the effect of the ApoE ϵ4 allele on the risk of AD might be mediated through elevated levels of total serum cholesterol. The Kivipelto study was again a prospective population‐based study that showed elevated mid‐life total cholesterol level was a risk factor for AD and was independent from risk from the ApoE ϵ4 allele and high mid‐life systolic blood pressure (Kivipelto 2002). The Jarvik study was a case‐control study, which again showed a positive association between serum cholesterol and risk of AD (Jarvik 1995). More recently, the Norwegian Counties Study, a long‐term observational study, showed dementia death was associated with increased total cholesterol levels in mid‐life (Strand 2013).

There may also be converging pathogenic mechanisms between cerebrovascular and Aβ plaque pathology ‐ cerebrovascular pathology with ischaemia resulting in upregulation of APP expression followed by Aβ deposition (Jendroska 1995). However, co‐existing pathology may occur independently of the disease process and increase the probability of exhibiting dementia in otherwise asymptomatic people (Riekse 2004).

Cholesterol and vascular dementia

VaD is the second most common form of dementia. It is characterised by both large and small vessel lesions. Subcortical ischaemic vascular disease caused by damage to tiny blood vessels that lie deep in the brain is now thought to be more prevalent than multi‐infarct dementia caused by large vessel lesions and stroke (Ballard 2000; Esiri 1997).

Sclerosis of small cerebral arteries and arterioles is considered to be responsible for the diffuse periventricular white matter abnormalities involved in the pathogenesis of subcortical VaD (Ryglewicz 2002). Risk factors for VaD are similar to risk factors for all types of vascular disease, namely hypertension, diabetes, smoking and hypercholesterolaemia (Ott 1998; Posner 2002; Stewart 1999). These factors are also important in the pathogenesis of AD (Decarli 2004); furthermore, the effects of vascular and AD pathologies are additive and in most population samples these disorders appear together (Snowdon 1997).

Plasma lipids could be associated with the risk of VaD through several mechanisms. High levels of LDL‐C and low levels of HDL‐C are established risk factors for coronary heart disease (Moroney 1999) and carotid artery atherosclerosis (Sharrett 1994). These may lead to cognitive impairment through cerebral hypoperfusion or embolism (Breteler 1994). LDL‐C may interact with ApoE to cause small vessel disease, and low levels of antioxidants known to occur in brains of people with VaD may lead to a higher susceptibility to oxidative stress and a higher grade of LDL‐C oxidation (Dantoine 2002; Paragh 2002).

One previous cross‐sectional analysis showed that the prevalence of VaD decreased with higher levels of HDL‐C and increased with higher levels of non‐HDL‐C. However, treatment with LLAs was not associated with the risk of prevalent VaD. Incidence of VaD was also calculated, again risk of VaD increased with increasing non‐HDL level but treatment with LLAs did not lower the risk of incident VaD (Reitz 2004). Other studies have found an association of VaD with decreased levels of HDL‐C (Kuriyama 1994; Muckle 1985; Zuliani 2001). The role of LDL‐C remains controversial, with some studies finding an association between increased LDL‐C and risk of VaD (Klich‐Raczka 2002; Moroney 1999; Paragh 2002), and other studies reporting a negative association (van Exel 2002; Yoshitake 1995).

Stroke is also a major risk factor for VaD. Debate continues as to whether increased cholesterol levels are a risk factor for stroke. Clinical trials indicated that statins significantly decreased stroke risk in vascular patients including patients with stroke (CTTC 2005; SPARCL 2006). The meta‐analysis carried out by the Cholesterol Treatment Trialists' Collaborators including 90,056 participants found that the use of statins caused a significant 17% proportional reduction in the incidence of first‐ever stroke of any type per 1 mmol/L LDL‐C reduction (CTTC 2005). In the secondary prevention of stroke, the Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) study showed that treatment with atorvastatin reduced the risk of recurrent cerebrovascular events in people with recent stroke or transient ischaemic attack but no history of heart disease (SPARCL 2006). By reducing the risk of stroke, statins may also act to reduce the incidence of post‐stroke dementia.

Statins

Statins are a class of drugs that inhibit 3‐hydroxy‐3‐methylglutaryl coenzyme A (HMG‐CoA) reductase. HMG‐CoA reductase is the rate‐limiting enzyme in the cascade of cellular cholesterol biosynthesis. Statins thereby reduce the formation and entry of LDL‐C into the circulation and upregulate LDL receptor activity, lowering LDL‐C and triglycerides and increasing HDL‐C. Several studies in cell culture and animals have demonstrated that treatment with cholesterol‐lowering drugs reduces the production of Aβ (Fassbender 2001; Refolo 2001; Simons 1998). It was therefore hypothesised that reduction of Aβ levels by statins may have neuroprotective effects in patients with AD (Simons 2001; Wolozin 2001). However, one study of transgenic mice found that levels of Aβ in the brains of simvastatin‐treated mice did not differ from those of untreated mice. Simvastatin treatment did lead to the reversal of learning and memory deficits and the authors hypothesised that the benefit of simvastatin may have been due to modulation of signalling pathways in memory formation (Li 2006). However, further work then demonstrated an association between antecedent statin use and neurofibrillary tangle burden at autopsy with risk for typical AD pathology reduced in statin users (Li 2007). The effects of statins on AD neuropathology are therefore not totally understood. Their possible role in the treatment of VaD includes secondary prevention of stroke and other pleiotropic effects as detailed below.

Statins are classified according to their solubility in lipids (lipophilic) or water (hydrophilic). Lipophilic statins (lovastatin, simvastatin, atorvastatin, fluvastatin) cross the BBB and penetrate cell membranes more effectively than hydrophilic statins and may be more efficient theoretically in the treatment of dementia than the hydrophilic statins (pravastatin). However, in contrast, reducing cholesterol synthesis below a critical level can induce neuronal death (Michikawa 1998), and may paradoxically make treatment with hydrophilic statins more appropriate (Sparks 2006).

Statins also have pleiotropic effects. They can improve the endothelial function of atherosclerotic vessels by decreasing endothelial 1 and angiotensin II type 1 receptors and increasing nitric oxide (Wassmann 2001). Low nitric oxide levels lead to impaired endothelial function, platelet aggregation and enhanced leukocyte adhesion to the endothelium. Statins also have antithrombotic effects as they decrease plasminogen activator levels and have anti‐inflammatory effects as they decrease adhesion molecules (Reitz 2004). They may also have the ability to reduce apoptosis and cellular death (Ruocco 2002). Many of these cholesterol‐independent effects reflect the ability of statins to block the synthesis of important isoprenoid intermediates, which serve as lipid attachments for a variety of intracellular signalling molecules (Liao 2002).

It is also possible that reduced cholesterol synthesis and concentration in the CNS caused by treatment with statins may cause neurocognitive deficits. Several investigators have therefore questioned the potential detrimental effects of lowering cholesterol on cognition (King 2003; Muldoon 2000; Wagstaff 2003; Zhang 2004). It has been shown that large doses of statins can produce substantial neurotoxicity in dogs (Berry 1988; Walsh 1996). Statins lower circulating levels of vitamin E and ubiquinone (Coenzyme Q10) and may affect the synthesis of polyunsaturated fatty acids that are integral to neuronal membranes (Palomaki 1997; Rise 1997). Researchers have speculated that low concentrations of one or more components of lipoprotein particles circulating in the bloodstream may produce subtle but measurable impairments of mental processes by influencing the supply of fat‐soluble micronutrients, specifically, vitamin E, β‐carotene and vitamin A (Muldoon 1997). However, on balance, potential benefits from statins appear to outweigh potential detrimental effects and we will assess adverse effects from statins in this review.

Statins are widely available and prescribed for treatment of dyslipidaemia and secondary prevention of cardiovascular and cerebrovascular disease. Their cost is relatively low and some have come off patent so are prescribed generically.

Statin treatment in dementia

The use of statin therapy in established AD or VaD is a relatively unexplored area. There have been a number of studies on the role of statins in the prevention of dementia but these are the focus of another Cochrane review (McGuinness 2009). Further trials have followed patients with AD and dementia; these are the focus of this review and are presented in the results section.

One post‐hoc analysis on data pooled from three double‐blind placebo‐controlled clinical trials of galantamine in AD showed no significant change in cognitive status in association with the use of statins (Winblad 2007).

One observational study in patients with AD followed for 34.8 months showed that patients treated with LLAs had a slower decline on the Mini Mental State Examination (MMSE) than patients with untreated dyslipidaemia or normolipidaemic patients. The study concluded that LLAs (including fibrates and statins) may slow cognitive decline in patients with AD and may have a neuroprotective effect, but this finding needs to be confirmed by randomised placebo‐controlled trials (Masse 2005).

Patients with mild cognitive impairment (MCI) at baseline in the observational Ginkgo Evaluation of Memory Study showed no significant cognitive benefit from treatment with LLAs including statins. This was in contrast to people without MCI at baseline who showed a reduced risk of all‐cause dementia and AD (Betterman 2012).

Other LLAs (fibrates, niacin/nicotinic acid, anion‐exchange resins) have been assessed with statins in several dementia studies. Fibrates are the main class in use other than statins. Rockwood et al. published a population‐based survey from the Canadian Study of Health and Aging (CSHA) demonstrating use of statins and other LLAs on reduced risk of AD in people younger than 80 years old (Rockwood 2002). In contrast, the UK General Practice Research Database study showed that only statins reduced the risk of dementia, other LLAs did not (Jick 2000). In a further study in patients with AD, all LLAs including statins were associated with a slower annual cognitive decline but there was no significant difference between statins and other LLAs and there was lack of statistical power to compare statins to fibrates (Masse 2005).

Fibrates do not inhibit cholesterol biosynthesis, they stimulate β‐oxidation of fatty acids and act mainly by decreasing serum triglycerides. They are only used first line in people with hypertriglyceridaemia and can be used in combination with statins in people not responding to single therapy. Fibrates also have anti‐inflammatory effects as they inhibit the production of different pro‐inflammatory molecules (Pahan 2006). However, in this review, we were primarily interested in role of statins in the treatment of dementia.

This review aimed to collate the best available evidence regarding use of statins in AD and VaD.

Statins decrease coronary events in the primary and secondary prevention of coronary heart disease significantly. The question is whether they have a significant therapeutic effect in dementia. Any intervention shown to slow the progression of dementia would have huge worldwide economic benefit.

Objectives

Primary objective

To evaluate the efficacy and safety of statins in the treatment of AD and VaD.

Secondary objective

To evaluate if the efficacy of statins in the treatment of AD and VaD depends on cholesterol level, ApoE genotype or cognitive level.

Methods

Criteria for considering studies for this review

Types of studies

Randomised, double‐blind, placebo‐controlled trials in which a statin was given for at least six months. We chose six months as we considered this to be the minimum length of time required to be on treatment to allow a disease‐modifying effect and before any cognitive benefit could be attained.

We excluded trials comparing two different statins without a placebo.

Types of participants

Patients with a diagnosis of probable or possible AD according to National Institute of Neurological and Communicative Disorders and Stroke ‐ the Alzheimer's Disease and Related Disorders Association (NINCDS‐ADRDA) criteria or acceptable equivalent.

Patients with a diagnosis of probable or possible VaD according to National Institute of Neurological Disorders and Stroke/Association International pour le Recherché at l'Enseignement en Neurosciences (NINDS/AIREN) criteria or acceptable equivalent.

We included trials with Diagnostic and Statistical Manual (DSM) III, IIIR or IV dementia, but analysed these separately from those with causal diagnoses for dementia.

Types of interventions

Any type of statin (hydrophilic and lipophilic) given in appropriate dose compared with placebo.

Types of outcome measures

Primary outcomes

Change in MMSE, Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐cog) or other accepted cognitive measure.

Secondary outcomes

  • Incidence and severity of adverse effects.

  • Change in cognitive status accounting for prior cholesterol level, ApoE genotype and cognitive level.

  • Patient perceived quality of life.

  • Change in activities of daily living (ADL).

  • Change in behaviour.

Search methods for identification of studies

We searched ALOIS (www.medicine.ox.ac.uk/alois) ‐ the Cochrane Dementia and Cognitive Improvement Group's Specialized Register on 20 January 2014. We used the search terms: statin OR statins OR "Hydroxymethylglutaryl‐CoA Reductase Inhibitors".

ALOIS is maintained by the Trials Search Co‐ordinator of the Cochrane Dementia and Cognitive Improvement Group and contains studies in the areas of dementia prevention, dementia treatment and cognitive enhancement in healthy people. The studies are identified from:  

  1. monthly searches of a number of major healthcare databases: MEDLINE, EMBASE, CINAHL, PsycINFO and LILACS;

  2. monthly searches of a number of trial registers: ISRCTN; UMIN (Japan's Trial Register); the World Health Organization (WHO) portal (which covers ClinicalTrials.gov; ISRCTN; the Chinese Clinical Trials Register; the German Clinical Trials Register; the Iranian Registry of Clinical Trials and the Netherlands National Trials Register, plus others);

  3. quarterly search of the Cochrane Central Register of Controlled Trials (CENTRAL);

  4. six‐monthly searches of several grey literature sources: ISI Web of Knowledge Conference Proceedings; Index to Theses; Australasian Digital Theses.

To view a list of all sources searched for ALOIS see About ALOIS on the ALOIS website.

Details of the search strategies used for the retrieval of reports of trials from the healthcare databases, CENTRAL and conference proceedings can be viewed in the 'methods used in reviews' section within the editorial information about the Dementia and Cognitive Improvement Group.

We performed additional searches in many of the sources listed above to cover the timeframe from the last searches performed for ALOIS to ensure that the search for the review was as up‐to‐date and as comprehensive as possible. The search strategies used can be seen in Appendix 1.

Searches carried out in the previous version(s) of the review can be viewed in Appendix 2.

Data collection and analysis

Selection of studies

Two authors undertook the search and screening of publications (BMcG, supported by PP). Both authors agreed on and tested the MeSH terms and search strategy. Three other authors (DC, RM and RB) acted as adjudicators and reviewed the process. Authors independently selected trials for relevance against the defined inclusion criteria.

Data extraction and management

We extracted data from the published reports using a data collection form. One author (BMcG) extracted data and was by supported by a second author (RM).

Assessment of risk of bias in included studies

We assessed methodological quality of the included trials using The Cochrane Collaboration's tool for assessing risk of bias. We assessed the following domains: selection bias, performance bias, detection bias, attrition bias and reporting bias. We included trials with low risk of bias. We planned sensitivity analyses to determine inclusion/exclusion of low‐quality studies.

Measures of treatment effect

Continuous data

We extracted the mean change from baseline, the standard error of the mean change and the number of patients for each treatment group at each assessment for continuous data. Where changes from baseline were not reported, we extracted the mean, standard deviation (SD) and the number of patients for each treatment group at each time point. We also extracted available data on demographics of patients (age, gender, lipid values at baseline), statin regimen (type of statin, daily dosage, starting time, duration) and follow‐up duration. We reported 95% confidence intervals (CI).

Dichotomous data

For binary data (adverse events or not), we sought the numbers in each treatment group and the numbers experiencing the outcome of interest. We reported results as an odds ratio (OR) with 95% CI.

Dealing with missing data

For each outcome measure, we sought data on every patient assessed. To allow an intention‐to‐treat analysis, we sought the data irrespective of compliance, whether or not the patient was subsequently deemed ineligible, or otherwise excluded from treatment or follow‐up. If intention‐to‐treat data were not available in the publications, we sought "on‐treatment" or the data of those who completed the trial and indicated as such. We did not use data from titration phases prior to the randomised phase to assess safety or efficacy because patients are usually not randomised and treatments are not usually concealed.

Assessment of heterogeneity

In all cases, we presented the overall estimate from a fixed‐effect model and performed a standard Chi2 test to check for heterogeneity. We also assessed the impact of heterogeneity on the meta‐analysis using the I2 statistic. If we found significant heterogeneity, we presented a random‐effects model and reported this as the main result.

Sensitivity analysis

If heterogeneity still existed with any model, then we carried out a sensitivity analysis (excluding studies with conflicting results from the rest) thereby assessing the robustness of the results of fixed‐effect versus random‐effects models.

Results

Description of studies

Results of the search

The initial electronic searches retrieved 152 references. We included three studies in the initial meta‐analysis (ADCLT 2005;LEADe 2010;Simons 2002) (Figure 1).


Study flow diagram initial review.

Study flow diagram initial review.

We performed two top‐up searches for this update: one in February 2013 (which yielded 105 references) and one in January 2014 (which yielded 27 references). We included one new study from the search in 2013 (Sano 2011, 406 participants) (Figure 2). We included no studies from the 2014 search.


Study flow diagram update 2013 search.

Study flow diagram update 2013 search.

Included studies

The search identified four RCTs with 1154 participants (ADCLT 2005;LEADe 2010;Sano 2011; Simons 2002). For full details, see the Characteristics of included studies table. Ages for participation ranged from 50 to 90 years with mean ages in studies of 68 to 78 years representing older adults with dementia.

ADCLT 2005 included 63 patients with a diagnosis of probable or possible AD as outlined by NINCDS‐ADRDA and Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM‐IV) criteria; people 51 years or older with mild‐to‐moderate impairment (MMSE score 12 to 28) were eligible. All but six individuals were taking cholinesterase inhibitors, three people in the atorvastatin group and three people in the placebo group. The mean age (± SD) was 78.9 ± 1.2 years in placebo group and 78.15 ± 1.3 years in atorvastatin group.

LEADe 2010 included 614 patients with a diagnosis of probable AD according to DSM‐IV and NINCDS‐ADRDA criteria and of mild‐to‐moderate severity, defined as a MMSE score of 13 to 25 at screening. Participants were 53% female, age range 50 to 90 years, mean age (± SD) 74 ± 8 years. Patients were receiving donepezil 10 mg for at least three months before randomisation and LDL‐C was 2.5 to 3.5 mmol/L for inclusion.

Sano 2011 included 406 patients with a diagnosis of mild‐to‐moderate AD according to NINCDS‐ADRDA criteria. Patients were older than 50 years, with an MMSE score of 12 to 26. Participants were 59.9% female, mean age (± SD) 75.1 ± 9.0 years in the placebo group and 58.8% female, mean age 74.0 ± 9.6 years in the intervention group. Stable use (for at least three months) of cholinesterase inhibitors and memantine was allowed. Patients were excluded if they were taking LLAs or if they had conditions requiring cholesterol‐lowering treatment according to guidelines at the time. Participants were also excluded if they had LDL‐C below 80 mg/dL (2.1 mmol/L) or triglycerides greater than 500 mg/dL (5.6 mmol/L).

Simons 2002 was primarily a study investigating whether statins alter cholesterol metabolites and reduce Aβ levels in the cerebrospinal fluid (CSF) of AD patients. Cognition was assessed as a secondary outcome. Forty‐four patients with probable AD as defined by NINCDS‐ADRDA criteria and mild‐to‐moderate severity (MMSE scores 12 to 26) were recruited. Patients were allowed to take donepezil or rivastigmine if the dose had been unchanged for three months prior to study entry and remained stable during the 26‐week study period. Mean age (± SD) was 68.5 ± 8 years in the placebo group and 68.0 ± 9 years in the simvastatin group. Participants were recruited primarily from the community.

ADCLT 2005 provided data on change in ADAS‐Cog at three‐monthly intervals for up to one year. Data were also provided on change in total cholesterol level, Clinical Global Impression of Change (CGIC) score, MMSE score, Neuropsychiatric Inventory (NPI) total score and Geriatric Depression Scale (GDS) total score between placebo and atorvastatin groups.

LEADe 2010 provided data on change in ADAS‐Cog and Alzheimer's Disease Cooperative Study‐Clinical Global Impression of Change (ADCS‐CGIC) scores between atorvastatin and placebo groups. Following randomisation, these measures were performed at three‐month intervals through month 18. Secondary outcome measures were change in NPI, Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Clinical Dementia Rating‐Sum of Boxes (CDR‐SB), MMSE and modified ADAS‐Cog. Change in total, LDL‐C and HDL‐C, and triglycerides was provided.

Sano 2011 provided data on change in ADAS‐Cog score between simvastatin and placebo groups at three, six, 12 and 18 months from baseline. Secondary outcome measures included change in ADCS‐CGIC, MMSE, dependence scale, ADCS‐ADL, NPI and three supplemental cognitive tests from the ADCS instrument protocol.

Simons 2002 provided data on change in MMSE and ADAS‐Cog score between simvastatin and placebo groups at 26 weeks.

Treatment in ADCLT 2005 consisted of atorvastatin 80 mg daily or matching placebo. Sixty‐three individuals were considered evaluable by completing the three‐month visit, 32 people received atorvastatin and 31 people received placebo. Forty‐six people completed the one‐year study, 25 received atorvastatin and 21 received placebo. Reasons for drop‐out were not provided. Atorvastatin treatment produced significant decreases in total cholesterol (40%), LDL‐C (54%), and very‐low‐density‐cholesterol (VLDL‐C) (30%) relative to placebo. ApoE genotyping was carried out on study participants. Sixty per cent of the placebo group and 62.5% of the atorvastatin group had one or more E4 allele. Change in performance among participants with screening cholesterol levels 200 mg/dL (5.2 mmol/L) or greater was compared with performance change in participants with levels less than 200 mg/dL (5.2 mmol/L). Mean change in ADAS‐Cog performance at six months was established for atorvastatin‐ and placebo‐treated participants grouped according to their ApoE genotype. Within‐group and between‐group comparisons according to presence of apolipoprotein E4 allele were performed, followed by comparisons based on dose of the apolipoprotein E4 allele.

Treatment in LEADe 2010 consisted of atorvastatin 80 mg daily or matching placebo for 72 weeks. A total of 640 patients were randomised with a modified intention‐to‐treat population of 297 in the atorvastatin group and 317 in the placebo group. Mean (± SD) prior donepezil treatment was 409 ± 407 days. Results concerning ApoE genotype were available for 511 patients, observed ApoE4 frequency was 60%. Atorvastatin treatment produced significant decreases in total cholesterol, LDL‐C (50.2%) and triglycerides but no significant change in HDL‐C. Reasons for drop‐outs were given.

Treatment in Sano 2011 consisted of simvastatin 20 mg for six weeks and 40 mg daily thereafter for the remainder of the 18‐month study. A total of 406 participants were randomised with 204 in the treatment group and 202 in the placebo group. Discontinuation reasons were given. Total cholesterol and LDL‐C levels were significantly reduced by the treatment compared with placebo; the reduction was 23% in total cholesterol and 37% in LDL‐C. HDL‐C levels were also increased with treatment by 2%. There was no difference in the use of approved anti‐dementia medications between the treatment groups during the study. Presence of the ApoE4 allele was not different in the two groups.

In Simons 2002, treatment consisted of simvastatin 40 mg daily for four weeks and 80 mg daily for the following 22 weeks. Disease duration was a mean (± SD) 2.8 ± 1.3 years in the placebo group and 2.6 ± 1.4 years in the simvastatin group. Serum LDL‐C showed few changes in the placebo group but was reduced by 52% on average in the simvastatin group. Reasons for drop‐outs were given. ApoE genotyping was not carried out.

Total adverse events were reported by LEADe 2010, Sano 2011, and Simons 2002.

Excluded studies

We excluded two studies from the analysis (Gutterman 2002;Winblad 2007). For full details, see Characteristics of excluded studies table.

Gutterman 2002 used data pooled from trials of patients treated with galantamine 24 mg daily or placebo for five to six months in randomised, double‐blind, placebo‐controlled trials. This was a post‐hoc analysis and so did not fulfil criteria for inclusion neither did it have adequate power to examine the effects of statins.

Winblad 2007 was also a post‐hoc analysis conducted on data pooled from three double‐blind, placebo‐controlled clinical trials of galantamine in patients with AD. There were four treatment groups: statin plus galantamine (42 participants), statin alone (50 participants), galantamine alone (614 participants) and neither galantamine nor statin (619 participants). As stated by the authors, it did not have sufficient power to examine the effects of statins.

Risk of bias in included studies

For full details, see 'Risk of bias' tables and Figure 3; Figure 4.


Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.


Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Allocation

In ADCLT 2005, randomisation was performed in blocks of 10 using the Excel spreadsheet random‐number generator, this appeared adequate.

In LEADe 2010, 1:1 randomisation was carried out, the medication was assembled for each patient based on a randomisation code prepared by Clinical Data Operations of Pfizer Inc. This appeared satisfactory.

In Sano 2011, the randomisation sequence was generated with equal probability of assignment to drug and placebo using a random permuted block treatment assignment stratified by site. The randomisation sequence was generated by the ADCS, a consortium of US centres funded by the National Institute on Aging. This appeared satisfactory.

In Simons 2002, a randomisation list was computer generated, two copies were prepared: one was used by the packaging department of the study medication or placebo and the other was kept in a locked location until the study was completed. This appeared satisfactory.

Blinding

In ADCLT 2005, all investigators were blinded to both treatment group and cholesterol profiles after randomisation as active treatment was expected to reduce circulating cholesterol levels. Medications were supplied in bulk by the pharmaceutical company and were coded at the pharmacy. This appeared adequate.

In LEADe 2010, it is stated in the published article there was "blinding of both the investigator and the subject" and in conference proceedings "trial data remained blinded, and the authors, steering committee, and the sponsor had no information relating to study outcomes". This appeared adequate.

In Sano 2011, it is stated "scratch‐off" code‐breakers were used so that instances of unblinding would be documented; all code breakers were collected at the end of the trial. Adequacy of the blind was assessed by questionnaires completed by participants, carers, psychometrists and site investigators.

In Simons 2002, adequate blinding appears to have been carried out. "All personnel directly involved in the conduct of the study remained unaware of the treatment groups until all patients had completed the trial and all data had been retrieved". Blood results were monitored by a physician not involved in the study, this guaranteed that all investigators were kept blinded.

Incomplete outcome data

In ADCLT 2005, flow of participants through the one‐year‐long study was provided, from 63 evaluable participants, 46 attended for final assessment. Reasons for not attending were not given.

In LEADe 2010 and Sano 2011, incomplete data were addressed.

In Simons 2002, incomplete data were addressed comprehensively.

Selective reporting

There was no evidence of selective reporting.

Other potential sources of bias

We identified no other potential sources of bias.

Effects of interventions

See: Summary of findings for the main comparison Statins compared with placebo for the treatment of dementia

Primary outcomes

The four studies assessed change in ADAS‐Cog from baseline. We calculated the mean change and SD from the available data and entered them into a meta‐analysis.

When we combined the four studies, there was no significant difference in ADAS‐Cog between the statin group and placebo group (mean difference ‐0.26, 95% CI ‐1.05 to 0.52, P value = 0.51) (Analysis 1.1) (Figure 5). There was also considerable heterogeneity when the studies were combined (Chi2 = 7.98, degrees of freedom (df) = 3, P value = 0.05, I2 = 62%). The random‐effects model, which usually gives more weight to small studies, was therefore used to re‐pool the data; the combined results were not significant (mean difference ‐0.33, 95% CI ‐2.10 to 1.44, P value = 0.71) (Analysis 1.2) (Figure 6). We carried out a sensitivity analysis and removal of ADCLT 2005 (the study with conflicting results) resulted in a reduction of I2 to 41% with the fixed‐effect model. Again, however, there was no significant difference in ADAS‐Cog between the statin and placebo groups (P value = 0.85). As the Simons 2002 study ran for 26 weeks, we combined data from ADCLT 2005 at 24 weeks and from LEADe 2010 and Sano 2011 at 26 weeks and again there was no significant difference in ADAS‐Cog between the statin and placebo groups (mean difference ‐0.08, 95% CI ‐0.85 to 0.70). Change in ADAS‐Cog from LEADe 2010 and Sano 2011 has been provided in the tables from the various time points (Analysis 1.3; Analysis 1.4; Analysis 1.5; Analysis 1.6; Analysis 1.7). Change in modified ADAS‐Cog (13‐item, 85‐point scale, Mohs 1997) has been provided from LEADe 2010. At no time was a beneficial effect on ADAS‐Cog or modified ADAS‐Cog seen with statin treatment (Analysis 1.8).


Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.1 ADAS‐Cog change from baseline.

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.1 ADAS‐Cog change from baseline.


Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.2 ADAS‐Cog change from baseline (using random‐effects model).

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.2 ADAS‐Cog change from baseline (using random‐effects model).

Change in MMSE was available from the four studies. When we combined data in a meta‐analysis there was no significant difference between the statin and placebo groups (mean difference ‐0.32, 95% CI ‐0.71 to 0.06, P value = 0.10) (Analysis 2.1) (Figure 7). Again there was significant heterogeneity when the studies were combined Chi2 = 15.46, df = 3, P value < 0.01, I2 = 81%) so we used the random‐effects model. There was no significant difference between statins and placebo (mean difference ‐0.88, 95% CI ‐2.04 to 0.28, P value = 0.09) (Analysis 2.2). A sensitivity analysis with Simons 2002 removed (the greatest outlier) reduced the heterogeneity (I2 statistic) to 67% with the fixed‐effect model with no effect on the outcome. Data were also compared at 24 weeks for ADCLT 2005, LEADe 2010, and Sano 2011; there was no significant difference between the statin and placebo groups (mean difference ‐0.30, 95% CI ‐0.66 to 0.05, P value = 0.09) (Analysis 2.4). Change in MMSE at different time points has been provided from LEADe 2010 and Sano 2011; at no time was there a beneficial effect from statin therapy.


Forest plot of comparison: 2 Mini Mental State Examination (MMSE) change from baseline, outcome: 2.1 MMSE change from baseline.

Forest plot of comparison: 2 Mini Mental State Examination (MMSE) change from baseline, outcome: 2.1 MMSE change from baseline.

Change in ADCS‐CGIC assessing CGIC was given in two studies: ADCLT 2005 and LEADe 2010. When data from these two trials were combined in a meta‐analysis using generic inverse variance there was no significant difference between the statin and placebo groups (mean difference ‐0.02, 95% CI ‐0.14 to 0.10, P value = 0.74) (Analysis 3.1) (Figure 8). We contacted the study author for the Sano 2011 trial as ADCS‐CGIC was an outcome but we received no response.


Forest plot of comparison: 3 Clinical Global Impression of Change (CGIC), outcome: 3.1 CGIC change.

Forest plot of comparison: 3 Clinical Global Impression of Change (CGIC), outcome: 3.1 CGIC change.

Secondary outcomes

Side effects

Side effects were elicited from blood tests and from speaking to patients and carers. LEADe 2010 stated incidence of persistent elevated liver enzymes (three times upper limit of normal on two consecutive measures four to 10 days apart) in the atorvastatin group was low at 2.6% and 0% in the placebo group. There were 60 (19.1%) atorvastatin‐treated and 69 (21.2%) placebo‐treated patients who experienced serious adverse events (SAEs), six of which in the atorvastatin group and one in the placebo group were considered to be treatment related by the investigator or sponsor. The SAEs considered to be treatment related in the atorvastatin group were hepatitis, acute renal failure/rhabdomyolysis/pancreatitis, abdominal pain/nausea/chest discomfort, transaminases elevation, liver disorder and gastrointestinal haemorrhage. In Simons 2002, two patients in the simvastatin group experienced adverse events: one patient had muscle pain without elevation of creatine kinase and one patient was withdrawn because creatine kinase was elevated. No adverse effects were reported in the placebo group. In Sano 2011, placebo and treatment groups did not differ in number of participants with one or more adverse events (181/202 with placebo and 189/204 with treatment). The most commonly occurring adverse events were falls, agitation, asthenia and anxiety. There was no significant difference either between the groups in the number of participants with SAEs, number of participants with adverse events requiring hospitalisation and number of deaths. Liver enzyme elevations were noted in 2% of the treatment and 4% of the placebo groups. Data from LEADe 2010, Sano 2011, and Simons 2002 were combined and there was no significant difference between statin and placebo groups (Analysis 4.1) (Figure 9).


Forest plot of comparison: 4 Incidence of adverse effects, outcome: 4.1 Treatment‐related adverse effects requiring discontinuation of treatment.

Forest plot of comparison: 4 Incidence of adverse effects, outcome: 4.1 Treatment‐related adverse effects requiring discontinuation of treatment.

Change in cognitive status accounting for prior cholesterol, ApoE genotype and cognitive level

In ADCLT 2005, among participants treated with atorvastatin, patients who had improved on the ADAS‐Cog at six months had baseline MMSE scores (mean ± SD, 21.93 ± 0.85) two points higher than patients who continued to deteriorate (19.83 ± 1.10) (P value < 0.06). Patients who improved on the ADAS‐Cog also had higher baseline cholesterol levels than patients who deteriorated (mean change (± SD) in ADAS‐Cog ‐2.14 ± 1.20 atorvastatin plus cholesterol greater than 200 mg/dL (5.2 mmol/L); 0.11 ± 0.68 atorvastatin plus cholesterol less than 200 mg/dL (5.2 mmol/L)). A significant difference was seen in ADAS‐Cog performance at six months between the atorvastatin and placebo groups in individuals with an apolipoprotein E‐4 allele (P value = 0.012) but not between the groups comprised of participants without an apolipoprotein E4 allele (P value = 0.967). (Note: there were very small numbers in all groups.)

In Sano 2011, presence of the APOE4 allele was not different in the two groups and was not associated with the rate of change in the ADAS‐Cog score. A median split was used to examine those of low and high baseline MMSE score; there was no difference in rate of ADAS‐Cog change between the simvastatin and placebo groups.

Quality of life

In LEADe 2010, there was no significant difference between the atorvastatin and placebo groups in Caregiver Burden Questionnaire and Patient Health Resources Utilization.

In Sano 2011, it is stated there was no significant difference between groups in quality of life as measured by informant or participant.

Behaviour

ADCLT 2005, LEADe 2010, and Sano 2011 provided data on NPI (information obtained from the carer). We combined data from these three studies at six (Analysis 5.1) and 12 months (Analysis 5.2) (Figure 10). There was a small but significant benefit from statins seen (mean difference at six months ‐0.84, 95% CI ‐1.64 to ‐0.05, P value = 0.04; mean difference at 12 months ‐1.11, 95% CI ‐2.10 to ‐0.12, P value = 0.03). This was no longer evident when we used the random‐effects model due to significant heterogeneity (mean difference at 12 months 1.64, 95% CI ‐3.45 to 0.18, P value = 0.11] (Analysis 5.3). In addition, if we removed ADCLT 2005 (the study with outlying results) from the analysis, the heterogeneity disappeared as did the significant treatment effect (I2 = 0%, P value = 0.13). ADCLT 2005 provided change in GDS (information obtained from the patient). Atorvastatin provided significant benefit on the GDS (P value < 0.04); there was deterioration in the placebo group and improvement in the atorvastatin group.


Forest plot of comparison: 5 Neuropsychiatric Inventory (NPI), outcome: 5.2 NPI change from baseline at 1 year.

Forest plot of comparison: 5 Neuropsychiatric Inventory (NPI), outcome: 5.2 NPI change from baseline at 1 year.

Activities of daily living

ADCLT 2005 and Sano 2011 provided differences in performance on the carer‐rated Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL) Inventory. In ADCLT 2005, the difference between the treatment and placebo groups did not reach significance (P value > 0.23) (no further data provided). Sano 2011 provided data on change from baseline ADCS‐ADL score. At no time was a difference seen between the placebo and treatment groups (Analysis 7.1). In LEADe 2010, there was no benefit from atorvastatin compared with placebo in the ADFACS, a measure of function (Analysis 6.4).

Discussion

Summary of main results

We identified four studies (ADCLT 2005; LEADe 2010; Sano 2011; Simons 2002). Mean change in ADAS‐Cog from baseline was an outcome in the four trials and there was no significant difference between the statin and the placebo groups.

Change in MMSE from baseline was also reported in all of the studies. There was no significant difference between the statin and placebo groups at any time point.

CGIC did not differ between the two groups in the two studies that recorded this measure (ADCLT 2005; LEADe 2010).

The statins were well tolerated and incidence of side effects was low. The statin group did not have a significantly higher rate of adverse effects requiring discontinuation of treatment when the data were combined.

There was some evidence from ADCLT 2005 that greater cognitive effect from atorvastatin was seen in patients with higher cholesterol at baseline, higher MMSE at baseline and in people with an apolipoprotein E4 allele present. The MMSE and ApoE effect was not seen in the larger Sano 2011 study.

There was no difference in ADL or quality of life between the two treatment groups. There was some evidence that statins provided a benefit in behaviour and mood.

The four trials included patients with AD only. We identified no trials assessing the effect of statins in the treatment of VaD.

It was not possible to assess if lipophilic statins or hydrophilic statins were more efficacious as all studies used the lipophilic statins simvastatin (Sano 2011; Simons 2002) or atorvastatin (ADCLT 2005; LEADe 2010). There appears to be a spectrum of lipophilicity within the lipophilic statins with simvastatin and cerivastatin being 2.5 to 4.0 times more lipophilic than lovastatin, fluvastatin and atorvastatin under the same physiological pH conditions (Joshi 1999). There is also debate about atorvastatin's lipophilicity and permeability across the BBB with some evidence that it crosses the BBB effectively (Kandiah 2009), and other evidence to suggest it does not (Knopp 1999). In any case, there was no convincing evidence that either atorvastatin or simvastatin were beneficial to cognition in the treatment of AD. There was no evidence that statins were detrimental to cognition.

One other comment relates to the fact that patients with elevated cholesterol levels were excluded from the large RCTs for safety and ethical reasons so the very group perhaps most likely to benefit was excluded (LEADe 2010; Sano 2011). This may be especially relevant given the ADCLT trial showed people with higher cholesterol level at baseline showed most cognitive benefit.

Overall completeness and applicability of evidence

The LEADe 2010 study was the largest with 640 patients so results from this are likely to be robust. Cognition was a primary outcome (ADAS‐Cog) along with global function (ADCS‐CGIC). Secondary outcomes included NPI, modified ADAS‐Cog, MMSE, CDR‐SB and ADFACS. Exploratory measures included a Caregiver Questionnaire and a Patient Health Resources Utilization questionnaire.

The Sano 2011 study was a large RCT with 406 participants. Cognition was a primary outcome (ADAS‐Cog) with ethnicity, baseline MMSE, ADCS‐ADL and LDL‐C meeting criteria for consideration as confounders in outcome analyses. Secondary outcomes included MMSE, Dependence Scale, ADCS‐ADL, NPI and CGIC (upon request from author). Findings from this study are considered complete and applicable.

In Simons 2002, the primary outcome was effect of statins on cholesterol metabolites and Aβ levels in the CSF of 44 patients with AD. Cognitive performance was a secondary outcome and was assessed at the beginning and end of the 26‐week study. Only 37 patients completed the study, so the impact of this study is likely to be small.

In ADCLT 2005, the primary outcomes were change in cognitive function (ADAS‐Cog) and clinical efficacy (CGIC). Secondary outcomes were change in MMSE, NPI, ADCS‐ADL and GDS, so results were applicable. However, the study was small with data available from 63 participants.

Quality of the evidence

All studies had adequate sequence generation and blinding.

Potential biases in the review process

None identified.

Agreements and disagreements with other studies or reviews

One previous systematic review assessed prevention and treatment of dementia or AD by statins (Zhou 2007). This was published before the LEADe 2010 results were available. Two studies were identified as identified in this review and there was no statistically significant difference in ADAS‐Cog between the statin and placebo groups when the trials were pooled (Simons 2002; ADCLT 2005). This is in agreement with this review.

One further review reported findings similar to this Cochrane review (Shepardson 2011). The review combined results from prevention and treatment of dementia with statins trials. Prevention of dementia with statins is the subject of another Cochrane review (McGuinness 2009). With respect to treatment trials, they identified ADCLT 2005 and LEADe 2010. They also reported negative findings for LEADe 2010 and positive findings for ADCLT 2005, but did not perform a meta‐analysis. The authors recommended future human studies take into account BBB permeabilities of the statins when analysing results, include specific analyses of the effects on LDL‐C and HDL‐C and future trials to be conducted solely in patients with mild AD, who have the best chance for disease modification.

Two more‐recent meta‐analyses found no significant benefit from statins in the primary outcome measure ADAS‐Cog in agreement with this study (Gizachew 2012; Pandey 2013). The Gizachew 2012 study assessed results from observational studies and RCTs. When they combined RCTs in a meta‐analysis, they made similar conclusions; there was no benefit from statins in treatment of dementia. The Pandey 2013 meta‐analysis included five studies and concluded no significant benefit in ADAS‐Cog from statins, they found a significant benefit on MMSE but significant heterogeneity similar to this review.

Study flow diagram initial review.
Figures and Tables -
Figure 1

Study flow diagram initial review.

Study flow diagram update 2013 search.
Figures and Tables -
Figure 2

Study flow diagram update 2013 search.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 4

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.1 ADAS‐Cog change from baseline.
Figures and Tables -
Figure 5

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.1 ADAS‐Cog change from baseline.

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.2 ADAS‐Cog change from baseline (using random‐effects model).
Figures and Tables -
Figure 6

Forest plot of comparison: 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, outcome: 1.2 ADAS‐Cog change from baseline (using random‐effects model).

Forest plot of comparison: 2 Mini Mental State Examination (MMSE) change from baseline, outcome: 2.1 MMSE change from baseline.
Figures and Tables -
Figure 7

Forest plot of comparison: 2 Mini Mental State Examination (MMSE) change from baseline, outcome: 2.1 MMSE change from baseline.

Forest plot of comparison: 3 Clinical Global Impression of Change (CGIC), outcome: 3.1 CGIC change.
Figures and Tables -
Figure 8

Forest plot of comparison: 3 Clinical Global Impression of Change (CGIC), outcome: 3.1 CGIC change.

Forest plot of comparison: 4 Incidence of adverse effects, outcome: 4.1 Treatment‐related adverse effects requiring discontinuation of treatment.
Figures and Tables -
Figure 9

Forest plot of comparison: 4 Incidence of adverse effects, outcome: 4.1 Treatment‐related adverse effects requiring discontinuation of treatment.

Forest plot of comparison: 5 Neuropsychiatric Inventory (NPI), outcome: 5.2 NPI change from baseline at 1 year.
Figures and Tables -
Figure 10

Forest plot of comparison: 5 Neuropsychiatric Inventory (NPI), outcome: 5.2 NPI change from baseline at 1 year.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 1 ADAS‐Cog change from baseline.
Figures and Tables -
Analysis 1.1

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 1 ADAS‐Cog change from baseline.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 2 ADAS‐Cog change from baseline (using random‐effects model).
Figures and Tables -
Analysis 1.2

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 2 ADAS‐Cog change from baseline (using random‐effects model).

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 3 ADAS‐Cog change at 3 months.
Figures and Tables -
Analysis 1.3

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 3 ADAS‐Cog change at 3 months.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 4 ADAS‐Cog change at 6 months.
Figures and Tables -
Analysis 1.4

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 4 ADAS‐Cog change at 6 months.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 5 ADAS‐Cog change at 12 months.
Figures and Tables -
Analysis 1.5

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 5 ADAS‐Cog change at 12 months.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 6 ADAS‐Cog change at 18 months.
Figures and Tables -
Analysis 1.6

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 6 ADAS‐Cog change at 18 months.

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 7 ADAS‐Cog change to 18 months (last observation carried forward).
Figures and Tables -
Analysis 1.7

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 7 ADAS‐Cog change to 18 months (last observation carried forward).

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 8 Modified ADAS‐Cog.
Figures and Tables -
Analysis 1.8

Comparison 1 Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline, Outcome 8 Modified ADAS‐Cog.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 1 MMSE change from baseline.
Figures and Tables -
Analysis 2.1

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 1 MMSE change from baseline.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 2 MMSE change from baseline using random‐effects model.
Figures and Tables -
Analysis 2.2

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 2 MMSE change from baseline using random‐effects model.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 3 MMSE change at 3 months.
Figures and Tables -
Analysis 2.3

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 3 MMSE change at 3 months.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 4 MMSE change at 6 months.
Figures and Tables -
Analysis 2.4

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 4 MMSE change at 6 months.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 5 MMSE change at 12 months.
Figures and Tables -
Analysis 2.5

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 5 MMSE change at 12 months.

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 6 MMSE change at 18 months.
Figures and Tables -
Analysis 2.6

Comparison 2 Mini Mental State Examination (MMSE) change from baseline, Outcome 6 MMSE change at 18 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 1 CGIC change.
Figures and Tables -
Analysis 3.1

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 1 CGIC change.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 2 CGIC change at 3 months.
Figures and Tables -
Analysis 3.2

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 2 CGIC change at 3 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 3 CGIC change at 6 months.
Figures and Tables -
Analysis 3.3

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 3 CGIC change at 6 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 4 CGIC change at 9 months.
Figures and Tables -
Analysis 3.4

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 4 CGIC change at 9 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 5 CGIC change at 12 months.
Figures and Tables -
Analysis 3.5

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 5 CGIC change at 12 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 6 CGIC change at 15 months.
Figures and Tables -
Analysis 3.6

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 6 CGIC change at 15 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 7 CGIC change at 18 months.
Figures and Tables -
Analysis 3.7

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 7 CGIC change at 18 months.

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 8 CGIC (last observation carried forward).
Figures and Tables -
Analysis 3.8

Comparison 3 Clinical Global Impression of Change (CGIC), Outcome 8 CGIC (last observation carried forward).

Comparison 4 Incidence of adverse effects, Outcome 1 Treatment‐related adverse effects requiring discontinuation of treatment.
Figures and Tables -
Analysis 4.1

Comparison 4 Incidence of adverse effects, Outcome 1 Treatment‐related adverse effects requiring discontinuation of treatment.

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 1 NPI change from baseline at 6 months.
Figures and Tables -
Analysis 5.1

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 1 NPI change from baseline at 6 months.

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 2 NPI change from baseline at 1 year.
Figures and Tables -
Analysis 5.2

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 2 NPI change from baseline at 1 year.

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 3 NPI change from baseline at 1 year (using random‐effects model).
Figures and Tables -
Analysis 5.3

Comparison 5 Neuropsychiatric Inventory (NPI), Outcome 3 NPI change from baseline at 1 year (using random‐effects model).

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 1 ADFACS change from baseline at 6 months.
Figures and Tables -
Analysis 6.1

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 1 ADFACS change from baseline at 6 months.

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 2 ADFACS change from baseline at 12 months.
Figures and Tables -
Analysis 6.2

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 2 ADFACS change from baseline at 12 months.

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 3 ADFACS change from baseline at 18 months.
Figures and Tables -
Analysis 6.3

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 3 ADFACS change from baseline at 18 months.

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 4 ADFACS overall (0‐18) months.
Figures and Tables -
Analysis 6.4

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 4 ADFACS overall (0‐18) months.

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 5 ADFACS (last observation carried forward).
Figures and Tables -
Analysis 6.5

Comparison 6 Alzheimer's Disease Functional Assessment and Change Scale (ADFACS), Outcome 5 ADFACS (last observation carried forward).

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 1 ADCS‐ADL change from baseline at 3 months.
Figures and Tables -
Analysis 7.1

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 1 ADCS‐ADL change from baseline at 3 months.

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 2 ADCS‐ADL change from baseline at 6 months.
Figures and Tables -
Analysis 7.2

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 2 ADCS‐ADL change from baseline at 6 months.

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 3 ADCS‐ADL change from baseline at 12 months.
Figures and Tables -
Analysis 7.3

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 3 ADCS‐ADL change from baseline at 12 months.

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 4 ADCS‐ADL change from baseline at 18 months.
Figures and Tables -
Analysis 7.4

Comparison 7 Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL), Outcome 4 ADCS‐ADL change from baseline at 18 months.

Summary of findings for the main comparison. Statins compared with placebo for the treatment of dementia

Patients: patients with dementia

Setting: community

Intervention: statin medication

Comparison: placebo

Outcomes

Absolute difference in means

(95% CI)

Number of participants

(studies)

GRADE Quality of evidence

Comments

Change in ADAS‐Cog

‐0.26

(‐1.05 to 0.52)

1110

(4)

High

Change in MMSE

‐0.32

(‐0.71 to 0.06)

1127

(4)

High

Change in CGIC

‐0.02

(‐0.14 to 0.10)

660

(2)

High

Change in NPI

‐1.11

(‐2.10 to ‐0.12)

983

(3)

High

ADAS‐Cog: Alzheimer's Disease Assessment Scale ‐ cognitive subscale; CGIC: Clinical Global Impression of Change; CI: confidence interval; MMSE: Mini Mental State Examination; NPI: Neuropsychiatric Inventory.

GRADE Working Group grades of evidence:

Hign quality: Further research is very unlikely to change our confidence in the estimate of effect

Moderate quality: Further evidence is likely to have an important impact on our confidence in the estimate of effect and may change the estimate

Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate

Very low quality: We are very uncertain about the estimate

Figures and Tables -
Summary of findings for the main comparison. Statins compared with placebo for the treatment of dementia
Comparison 1. Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ADAS‐Cog change from baseline Show forest plot

4

1110

Mean Difference (IV, Fixed, 95% CI)

‐0.26 [‐1.05, 0.52]

2 ADAS‐Cog change from baseline (using random‐effects model) Show forest plot

4

1110

Mean Difference (IV, Random, 95% CI)

‐0.33 [‐2.10, 1.44]

3 ADAS‐Cog change at 3 months Show forest plot

2

1019

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐0.46, 0.74]

4 ADAS‐Cog change at 6 months Show forest plot

4

1067

Mean Difference (IV, Fixed, 95% CI)

‐0.08 [‐0.85, 0.70]

5 ADAS‐Cog change at 12 months Show forest plot

2

920

Mean Difference (IV, Fixed, 95% CI)

‐0.12 [‐1.04, 0.80]

6 ADAS‐Cog change at 18 months Show forest plot

2

1020

Mean Difference (IV, Fixed, 95% CI)

‐0.68 [‐1.49, 0.14]

7 ADAS‐Cog change to 18 months (last observation carried forward) Show forest plot

1

Mean Difference (Fixed, 95% CI)

‐1.11 [‐2.37, 0.14]

8 Modified ADAS‐Cog Show forest plot

1

Mean Difference (Fixed, 95% CI)

Subtotals only

8.1 Modified ADAS‐Cog change from baseline at 3 months

1

612

Mean Difference (Fixed, 95% CI)

‐0.15 [‐0.99, 0.68]

8.2 Modified ADAS‐Cog change from baseline at 6 months

1

571

Mean Difference (Fixed, 95% CI)

‐0.23 [‐1.27, 0.81]

8.3 Modified ADAS‐Cog change from baseline at 9 months

1

533

Mean Difference (Fixed, 95% CI)

‐0.66 [‐1.73, 0.41]

8.4 Modified ADAS‐Cog change from baseline at 12 months

1

514

Mean Difference (Fixed, 95% CI)

‐0.41 [‐1.76, 0.95]

8.5 Modified ADAS‐Cog change from baseline at 18 months

1

437

Mean Difference (Fixed, 95% CI)

‐0.76 [‐2.40, 0.89]

8.6 Modified ADAS‐Cog overall (0‐18) months

1

614

Mean Difference (Fixed, 95% CI)

0.41 [‐0.62, 1.44]

8.7 Modified ADAS‐Cog (last observation carried forward)

1

614

Mean Difference (Fixed, 95% CI)

‐1.04 [‐2.44, 0.37]

Figures and Tables -
Comparison 1. Cognitive change (Alzheimer's Disease Assessment Scale ‐ cognitive subscale (ADAS‐Cog)) from baseline
Comparison 2. Mini Mental State Examination (MMSE) change from baseline

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 MMSE change from baseline Show forest plot

4

1127

Mean Difference (IV, Fixed, 95% CI)

‐0.32 [‐0.71, 0.06]

2 MMSE change from baseline using random‐effects model Show forest plot

4

1127

Mean Difference (IV, Random, 95% CI)

‐0.88 [‐2.04, 0.28]

3 MMSE change at 3 months Show forest plot

2

1017

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.31, 0.38]

4 MMSE change at 6 months Show forest plot

4

1084

Mean Difference (IV, Fixed, 95% CI)

‐0.30 [‐0.66, 0.05]

5 MMSE change at 12 months Show forest plot

4

1027

Mean Difference (IV, Fixed, 95% CI)

‐0.46 [‐0.90, ‐0.03]

6 MMSE change at 18 months Show forest plot

2

845

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.68, 0.41]

Figures and Tables -
Comparison 2. Mini Mental State Examination (MMSE) change from baseline
Comparison 3. Clinical Global Impression of Change (CGIC)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 CGIC change Show forest plot

2

660

Mean Difference (Fixed, 95% CI)

‐0.02 [‐0.14, 0.10]

2 CGIC change at 3 months Show forest plot

1

603

Mean Difference (Fixed, 95% CI)

0.01 [‐0.10, 0.12]

3 CGIC change at 6 months Show forest plot

1

564

Mean Difference (Fixed, 95% CI)

0.03 [‐0.11, 0.18]

4 CGIC change at 9 months Show forest plot

1

527

Mean Difference (Fixed, 95% CI)

‐0.02 [‐0.18, 0.14]

5 CGIC change at 12 months Show forest plot

1

505

Mean Difference (Fixed, 95% CI)

‐0.03 [‐0.20, 0.13]

6 CGIC change at 15 months Show forest plot

1

486

Mean Difference (Fixed, 95% CI)

0.02 [‐0.16, 0.20]

7 CGIC change at 18 months Show forest plot

1

435

Mean Difference (Fixed, 95% CI)

0.12 [‐0.07, 0.31]

8 CGIC (last observation carried forward) Show forest plot

1

614

Mean Difference (Fixed, 95% CI)

0.16 [‐0.01, 0.33]

Figures and Tables -
Comparison 3. Clinical Global Impression of Change (CGIC)
Comparison 4. Incidence of adverse effects

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Treatment‐related adverse effects requiring discontinuation of treatment Show forest plot

3

1089

Odds Ratio (M‐H, Fixed, 95% CI)

1.09 [0.58, 2.06]

Figures and Tables -
Comparison 4. Incidence of adverse effects
Comparison 5. Neuropsychiatric Inventory (NPI)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 NPI change from baseline at 6 months Show forest plot

3

1039

Mean Difference (IV, Fixed, 95% CI)

‐0.84 [‐1.64, ‐0.05]

2 NPI change from baseline at 1 year Show forest plot

3

983

Mean Difference (IV, Fixed, 95% CI)

‐1.11 [‐2.10, ‐0.12]

3 NPI change from baseline at 1 year (using random‐effects model) Show forest plot

3

983

Mean Difference (IV, Random, 95% CI)

‐1.64 [‐3.45, 0.18]

Figures and Tables -
Comparison 5. Neuropsychiatric Inventory (NPI)
Comparison 6. Alzheimer's Disease Functional Assessment and Change Scale (ADFACS)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ADFACS change from baseline at 6 months Show forest plot

1

596

Mean Difference (Fixed, 95% CI)

0.45 [‐0.36, 1.26]

2 ADFACS change from baseline at 12 months Show forest plot

1

512

Mean Difference (Fixed, 95% CI)

0.4 [‐0.74, 1.54]

3 ADFACS change from baseline at 18 months Show forest plot

1

470

Mean Difference (Fixed, 95% CI)

0.45 [‐0.99, 1.89]

4 ADFACS overall (0‐18) months Show forest plot

1

614

Mean Difference (Fixed, 95% CI)

0.43 [‐0.55, 1.42]

5 ADFACS (last observation carried forward) Show forest plot

1

614

Mean Difference (Fixed, 95% CI)

0.04 [‐1.24, 1.32]

Figures and Tables -
Comparison 6. Alzheimer's Disease Functional Assessment and Change Scale (ADFACS)
Comparison 7. Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ADCS‐ADL change from baseline at 3 months Show forest plot

1

406

Mean Difference (IV, Fixed, 95% CI)

‐0.33 [‐1.65, 0.99]

2 ADCS‐ADL change from baseline at 6 months Show forest plot

1

406

Mean Difference (IV, Fixed, 95% CI)

0.29 [‐1.33, 1.91]

3 ADCS‐ADL change from baseline at 12 months Show forest plot

1

406

Mean Difference (IV, Fixed, 95% CI)

‐1.24 [‐3.30, 0.82]

4 ADCS‐ADL change from baseline at 18 months Show forest plot

1

406

Mean Difference (IV, Fixed, 95% CI)

‐0.85 [‐3.50, 1.80]

Figures and Tables -
Comparison 7. Alzheimer's Disease Cooperative Study‐Activities of Daily Living (ADCS‐ADL)