Skip to main content
Advertisement
  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Education
    • Genetics
    • Neuroimmunology & Neuroinflammation
  • Online Sections
    • Neurology Video Journal Club
    • Inclusion, Diversity, Equity, Anti-racism, & Social Justice (IDEAS)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • COVID-19
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Neurology Future Forecasting Series
    • Null Hypothesis
    • Patient Pages
    • Topics A-Z
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit a Manuscript
    • Author Center

Advanced Search

Main menu

  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Education
    • Genetics
    • Neuroimmunology & Neuroinflammation
  • Online Sections
    • Neurology Video Journal Club
    • Inclusion, Diversity, Equity, Anti-racism, & Social Justice (IDEAS)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • COVID-19
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Neurology Future Forecasting Series
    • Null Hypothesis
    • Patient Pages
    • Topics A-Z
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit a Manuscript
    • Author Center
  • Home
  • Latest Articles
  • Current Issue
  • Past Issues
  • Neurology Video Journal Club
  • Residents & Fellows

User menu

  • Subscribe
  • My Alerts
  • Log in
  • Log out

Search

  • Advanced search
Neurology
Home
The most widely read and highly cited peer-reviewed neurology journal
  • Subscribe
  • My Alerts
  • Log in
  • Log out
Site Logo
  • Home
  • Latest Articles
  • Current Issue
  • Past Issues
  • Neurology Video Journal Club
  • Residents & Fellows

Share

March 23, 2010; 74 (12) Articles

Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease

LEADe

H. H. Feldman, R. S. Doody, M. Kivipelto, D. L. Sparks, D. D. Waters, R. W. Jones, E. Schwam, R. Schindler, J. Hey-Hadavi, D. A. DeMicco, A. Breazna
First published March 3, 2010, DOI: https://doi.org/10.1212/WNL.0b013e3181d6476a
H. H. Feldman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
R. S. Doody
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
M. Kivipelto
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
D. L. Sparks
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
D. D. Waters
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
R. W. Jones
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
E. Schwam
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
R. Schindler
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
J. Hey-Hadavi
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
D. A. DeMicco
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
A. Breazna
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Full PDF
Citation
Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease
LEADe
H. H. Feldman, R. S. Doody, M. Kivipelto, D. L. Sparks, D. D. Waters, R. W. Jones, E. Schwam, R. Schindler, J. Hey-Hadavi, D. A. DeMicco, A. Breazna
Neurology Mar 2010, 74 (12) 956-964; DOI: 10.1212/WNL.0b013e3181d6476a

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Permissions

Make Comment

See Comments

Downloads
1560

Share

  • Article
  • Figures & Data
  • Info & Disclosures
Loading

Abstract

Background: There is some evidence that statins may have a protective and symptomatic benefit in Alzheimer disease (AD). The LEADe study is a randomized controlled trial (RCT) evaluating the efficacy and safety of atorvastatin in patients with mild to moderate AD.

Methods: This was an international, multicenter, double-blind, randomized, parallel-group study. Subjects had mild to moderate probable AD (Mini-Mental State Examination score 13–25), were aged 50–90 years, and were taking donepezil 10 mg daily for ≥3 months prior to screening. Entry low-density lipoprotein cholesterol levels (LDL-C) were >95 and <195 mg/dL. Patients were randomized to atorvastatin 80 mg/day or placebo for 72 weeks followed by a double-blind, 8-week atorvastatin withdrawal phase. Coprimary endpoints were changes in cognition (Alzheimer's Disease Assessment Scale-Cognitive Subscale [ADAS-Cog]) and global function (Alzheimer's Disease Cooperative Study Clinical Global Impression of Change [ADCS-CGIC]) at 72 weeks.

Results: A total of 640 patients were randomized in the study. There were no significant differences in the coprimary endpoints of ADAS-cog or ADCS-CGIC or the secondary endpoints. Atorvastatin was generally well-tolerated.

Conclusions: In this large-scale randomized controlled trial evaluating statin therapy as a treatment for mild to moderate Alzheimer disease, atorvastatin was not associated with significant clinical benefit over 72 weeks. This treatment was generally well-tolerated without unexpected adverse events.

Classification of evidence: This study provides Class II evidence that intensive lipid lowering with atorvastatin 80 mg/day in patients with mild to moderate probable Alzheimer disease (aged 50–90), taking donepezil, with low-density lipoprotein cholesterol levels between 95 and 195 mg/dL over 72 weeks does not benefit cognition (as measured by Alzheimer's Disease Assessment Scale-Cognitive Subscale) (p = 0.26) or global function (as measured by Alzheimer's Disease Cooperative Study Clinical Global Impression of Change) (p = 0.73) compared with placebo.

Glossary

AchEI=
acetylcholinesterase inhibitor;
AD=
Alzheimer disease;
ADAS-Cog=
Alzheimer's Disease Assessment Scale-Cognitive Subscale;
ADCS-CGIC=
Alzheimer's Disease Cooperative Study Clinical Global Impression of Change;
ADFACS=
Alzheimer's Disease Functional Assessment and Change Scale;
ANCOVA=
analysis of covariance;
APP=
amyloid precursor protein;
BSI=
boundary shift integral;
CDR-SB=
Clinical Dementia Rating-Sum of Boxes;
LDL-C=
low-density lipoprotein cholesterol;
LOCF=
last observation carried forward;
LS=
least-squares;
MITT=
modified intent-to-treat population;
MMSE=
Mini-Mental State Examination;
NPI=
Neuropsychiatric Inventory;
RCT=
randomized controlled trial;
SAE=
serious adverse event.

Alzheimer disease (AD) is a chronic, progressive neurodegenerative disorder that is characterized by a variety of structural and functional neurodegenerative changes. Evidence suggests that β-amyloid (Aβ) and CNS cholesterol play important roles in this multifactorial degenerative process.1,2 Cholesterol modulates the processing of amyloid precursor protein (APP)-related Aβ production in vitro and in animal model studies,3–7 and AD neuropathologic lesions are demonstrable in patients who have coronary artery disease and no dementia.8 Reducing cholesterol through the use of statins (HMG-CoA reductase inhibitors) appears to affect the processing of APP and the production of Aβ.9 However, studies on the effect of statin treatment on serum or CSF Aβ concentrations in human subjects are equivocal.10

Retrospective case control studies have reported a positive relationship between statin use and reduced risk of developing AD,11,12 while prospective cohort studies have yielded mixed results.13,14 Two randomized, controlled cardiovascular prevention trials included cognition as a secondary outcome without significant benefits,15,16 whereas a proof-of-concept randomized controlled trial (RCT) of atorvastatin indicated a trend for symptomatic benefit in mild to moderate AD.17

The LEADe study (Lipitor's Effect in Alzheimer's Dementia) is the first reported large-scale RCT evaluating the efficacy and safety of statin therapy in patients with mild to moderate AD receiving background therapy with the acetylcholinesterase inhibitor (AchEI) donepezil.

METHODS

Standard protocol approvals, registrations, and patient consent.

Before screening, informed written consent or verbal assent was obtained from patients or authorized representatives or legal guardians. Caregivers were asked to participate as collateral informants for the study; they were required to accompany the patient to each visit and to participate in interviews. The study was conducted in accordance with the Declaration of Helsinki.18 Protocol and all amendments were approved by Institutional Review Boards or Ethics committees. Clinical trial registration (clinicaltrials.gov): NCT00053599.

Study design.

The LEADe study was an international, multicenter, double-blind, randomized, placebo-controlled, parallel-group 72-week study followed by a double-blind, 8-week randomized withdrawal phase. A detailed description of the design and methods of the LEADe study has been previously published.19 The primary study objective was to evaluate the efficacy of atorvastatin on the symptoms of mild to moderate AD, with a confirmatory objective of demonstrating its effect on the rate of change of MRI whole brain and hippocampal volumes.

The study was performed in 10 countries and 87 enrolling sites: United States (39), Germany (9), Canada (9), United Kingdom (8), Australia (7), Spain (7), South Africa (5), Sweden (5), Austria (4), and Denmark (4).

There were 3 phases to the study. An initial 2-week screening phase was performed to determine eligibility. Subjects were then randomized to a 72-week double-blind treatment period followed by an 8-week randomized double-blind atorvastatin 80-mg withdrawal period. Subjects took 2 40-mg tablets of atorvastatin or matching placebo (study drug) and 1 tablet of donepezil (10 mg) once daily, at the same time each day. The medication was assembled for each patient based on a randomization code prepared by Clinical Data Operations of Pfizer Inc., with blinding of both the investigator and the subject. The assigned doses of study medication could not be reduced during the trial.

The MRI substudy protocol included a baseline scan and a second scan at month 18 (or early termination if at least 9 months after baseline). Images were acquired at each study site on 1.5-T magnetic resonance units using a T1-weighted 3-dimensional spoiled gradient echo acquisition. All scans were acquired axially with slice thickness of 1.5 mm and 90–128 slices. After acquisition, the MRI scans were submitted for central reading and to determine their suitability for inclusion. Preprocessing steps included a correction for bias in homogeneity.20

Patients.

All patients met criteria for clinically probable AD according to the National Institute of Neurological and Communicative Disorders and Stroke-Alzheimer's Disease and Related Disorders Association21 and DSM-IV criteria for AD (Diagnostic and Statistical Manual of Mental Disorders, American Psychiatric Association, 1994), had screening Mini-Mental State Examination (MMSE)22 scores of 13–25 inclusive, and were aged 50–90 years. Most patients were community dwelling except a few subjects in assisted living situations. The study required caregivers to have contact at least 5 days per week for a minimum of 10 waking hours per week. Modified Hachinski Ischemic Score23 was ≤4. Low-density lipoprotein cholesterol (LDL-C) was ≥95 and ≤195 mg/dL (2.5–5.0 mmol/L) and, in the opinion of the investigator, not otherwise requiring treatment for dyslipidemia with any lipid-lowering agent. A cranial CT or MRI performed within 12 months of entry was consistent with a diagnosis of AD. Subjects experiencing any clinically significant or unstable medical condition were excluded, as were individuals with any cause for dementia other than AD, delirium, depression, or other concurrent diagnosis that might interfere with their participation in the study.

Most concomitant medications were allowed except for other AchEIs or other cholinergically active medications or investigational drugs. Putative nonprescription or prescribed cognitive enhancers (e.g., gingko, high-dose vitamin E, lecithin, estrogen, nonsteroidal antiinflammatory drugs) were not excluded, but dosage had to be stable for at least 3 months prior to randomization with no change anticipated during the course of the study. Selected atypical antipsychotics, antidepressants, and low-dose benzodiazepines were permitted if they had been used for a minimum of 3 months before the baseline visit and with stable frequency and dose.

Outcome measures.

The coprimary outcome measures were the Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-cog 11-item, 70-point scale)24 and the Alzheimer's Disease Cooperative Study Clinical Global Impression of Change (ADCS-CGIC).25 Following randomization, these measures were performed at 3-month intervals through month 18. Secondary outcomes measures which were performed following randomization at 6-month intervals through month 18 included the Neuropsychiatric Inventory (NPI),26 modified ADAS-cog (13-item, 85-point) scale,27 MMSE,22 Clinical Dementia Rating-Sum of Boxes (CDR-SB),28 and Alzheimer's Disease Functional Assessment and Change Scale (ADFACS).29 Exploratory measures including a Caregiver Burden Questionnaire and a Patient Health Resources Utilization were performed following randomization at 6-month intervals through month 18.

The MRI scan volumetric measures of the whole brain and hippocampus for analyses were obtained using semiautomated delineation techniques from the MIDAS image analysis program (Dementia Research Centre, University College of London).30 Differences in structural volumes were computed between baseline and follow-up images. Whole brain volumetric change was determined by measuring the boundary shift integral (BSI) around the whole brain/CSF boundary computed with software implementation of the BSI technique.

Statistical analysis.

The determination of the sample size of 600 subjects has been previously reported.19 It allowed for 80% power on the ADCS-CGIC to detect a 30%–40% difference in change scores between the 2 treatment groups at 18 months with a 2-tailed test (α = 0.05) and allowing for an attrition rate of up to 30%.28 As the sample size for the CGIC exceeded that required for the ADAS-cog, the sample size was deemed sufficient for both outcomes.

MRI substudy.

A sample size of 100 subjects in each treatment arm was estimated, assuming a 30% difference in volumetric change between the treatments and allowing for a 30% attrition rate.

The primary analysis of efficacy was based on a least-squares mean repeated-measures analysis for both the CGIC and the ADAS-cog total score on the modified intent-to-treat population (MITT), where each patient contributed data for each visit until they either completed the study or dropped out. MITT was used for all efficacy analyses (excepting MRI) and included all randomized patients who took at least 1 dose of study medication and who provided both a baseline assessment and at least 1 postbaseline efficacy assessment. The model used for the analyses of efficacy is based on a repeated-measures-mixed-effects analysis including the ADCS-CGIC score or change from baseline score for all other scores as the dependent variable, treatment as the major factor, visit as the repeated factor, and baseline score (if available) as the covariate plus treatment by visit interaction term in the model with subject nested within treatment as the cluster factor.

In addition to running the primary model on all scores, a last observation carried forward (LOCF) approach was employed as a sensitivity analysis (figure 2). The LOCF approach was from an analysis of covariance (ANCOVA) model that included only the last visit data in the analysis with treatment as the major factor and baseline score (if available) as the covariate.

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 2 ADAS-Cog and ADCS-CGIC results

(A) Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) and (B) Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change Scale (ADCS-CGIC) over time (modified intent-to-treat [MITT] population). Repeated-measures analyses were performed (with baseline score as covariate for ADAS-Cog score change from baseline). LS = least-squares; LOCF = last observation carried forward.

The MRI endpoints were evaluated for the patients in the MITT population with available MRI data using the following techniques: least-squares (LS) means and standard error (SE) for each treatment group with LS means differences, 95% confidence interval, and the p values for between-treatment comparison regarding the annualized BSI or whole hippocampus volume are based on ANCOVA model. In the ANCOVA model, the annualized BSI or annualized rate of decrease in whole hippocampus volume is included as the dependent variable with treatment arm as the major factor and baseline whole brain volume or baseline whole hippocampus volume as the covariate.

The safety population consisted of all randomized patients who had taken at least 1 dose of study medication, or for whom it was unknown whether they had taken any study medication but for whom there were postrandomization data. For the safety analysis, all observed AEs were reported, regardless of suspected causal relationship to the study drug. These included adverse drug reactions, illnesses with symptom onset during the study or exacerbations of preexisting illnesses, as well as objective test findings that resulted in a change in the study drug dosage. Rates for adverse events, laboratory abnormalities, and EKG changes were summarized.

All statistical analyses were performed using SAS 8.2.

Classification of evidence.

This study provides Class II evidence on the primary research question of whether intensive lipid lowering with atorvastatin 80 mg/day in patients with mild to moderate probable AD (aged 50–90), taking donepezil, with LDL-C levels between 95 and 195 mg/dL over 72 weeks benefits cognition (as measured by ADAS-Cog) or global function (as measured by ADCS-CGIC) compared with placebo.

RESULTS

Figure 1 presents the flow of patients through the study. Of the 1,008 selected for screening assessment, there were 640 patients randomized to the study, with an MITT population of 297 patients in the atorvastatin group and 317 patients in the placebo group. The overall study completion rate was very close to predicated at 70.6% with a dropout rate of 29.4%. A larger proportion of subjects discontinued the study in the atorvastatin group compared with the placebo group.

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 1 Patient disposition

MITT = modified intent to treat.

There were no significant differences in demographics, mean baseline scores, and mean lipid levels between treatment arms (table 1). Through the 72-week treatment period there were no significant differences on either the ADAS-cog (figure 2A and table e-1 on the Neurology® Web site at www.neurology.org) or the CGIC (figure 2B and table e-2), or among the secondary endpoints (NPI, ADFACS, CDR-SB, MMSE, and modified ADAS-cog [tables e-3 to e-7]). Since the primary endpoints were not met, the withdrawal phase is not reported.

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 1 Baseline patient characteristics (modified intent-to-treat population)

In the MRI substudy, there were 64 subjects evaluable for longitudinal assessment of hippocampal volumes and 58 evaluable for BSI and whole brain volumes. Atorvastatin therapy was associated with a significant smaller annualized decrease in hippocampal volume, but no significant difference in the annualized boundary shift integral between the treatment groups. Post hoc review of this MRI subsample showed differences in baseline demographics between treatment arms and a significant treatment effect on ADCS-CGIC in favor of atorvastatin.

While lipid values at baseline were similar between the 2 treatment groups, by month 3, mean LDL-C had decreased −72.4 mg/dL (−50.2%) in the atorvastatin group and −1.0 mg/dL (−0.2%) in the placebo group (figure 3). These changes remained constant through month 18. Total cholesterol and serum triglyceride levels demonstrated larger decreases in the atorvastatin group compared with the placebo group while there was little change in HDL-C in either treatment group.

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 3 Changes in lipid levels by treatment group over time

(A) Low-density lipoprotein cholesterol, (B) total cholesterol, (C) triglycerides, and (D) high-density lipoprotein cholesterol.

Treatment with atorvastatin was generally well-tolerated (table 2). The incidence of persistent elevations in AST or ALT levels (3 × upper limits of normal on 2 consecutive measures 4 to 10 days apart) was 2.6% in the atorvastatin group 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), with the most common being falls (9 and 13 subjects). Six subjects in the atorvastatin-treated group and 1 subject in the placebo group had SAEs considered treatment related by either the investigator or the sponsor. The SAEs considered treatment related in the atorvastatin group were hepatitis, acute renal failure/rhabdomyolysis/pancreatitis, abdominal pain/nausea/chest discomfort, transaminases elevation, liver disorder, and gastrointestinal hemorrhage. There were 9 deaths (2.9%) in the atorvastatin group and 6 (1.8%) in the placebo group. Causes of death in the atorvastatin group included congestive cardiac failure, pneumonia, acute renal failure/rhabdomyolysis/pancreatitis, gastric perforation, myocardial infarction, sudden cardiac death, complete atrioventricular block, and advancing AD.

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 2 Overview of patient disposition and adverse events

DISCUSSION

The LEADe study was a randomized placebo-controlled trial that was designed to evaluate the long-term symptomatic effects of atorvastatin in mild to moderate AD as well as its potential for disease modification. Significantly, it represents one of the largest and longest RCTs testing both the efficacy and safety of a statin in AD. The results of another large recently completed RCT of simvastatin with a similar study design are anticipated (clinicaltrials.gov, NCT00053599).

There is good evidence that these trial results are valid as most of the a priori assumptions were met. In particular, the placebo rate of decline on the primary outcomes was very close to the estimates in the sample size determination, while the actual dropout rate fell within the anticipated range. The cholesterol-lowering effects of atorvastatin followed expectations, with a sharp reduction seen by week 12 on LDL-C, total cholesterol, and triglycerides and with reversal of the reductions in the atorvastatin group randomized to receive placebo during the 8-week randomized withdrawal period of the trial. Despite the successful design and execution, the treatment intervention of adding atorvastatin on a background of donepezil use was not associated with any significant clinical benefits on either cognition or global functioning. Furthermore, there were no apparent benefits on a broader range of secondary outcome measures including neuropsychiatric symptoms, functional disability, disease staging, caregiver burden, and health economics. These results would indicate that cholesterol lowering in patients with mild to moderate AD without elevated LDL-C and at low risk of ischemic events is not associated with better preservation of cognition or function.

It is important to consider both the limitations as well as the broader implications of this study. The population in the current trial was restricted to patients who did not have an indication for statin treatment. Current practice guidelines for patients with hypercholesterolemia call for treatment with statins when lipid values are substantially out of range or in patients with lower values who have high vascular risk factor burden.31 This precluded the inclusion of patients with AD with significantly elevated LDL-C. It is therefore possible that the AD patient group with higher LDL-C levels with or without cardiovascular risk factors might have responded better to this intervention with atorvastatin. Also unanswered from this study is the question of whether atorvastatin might benefit patients with AD with significant concurrent cardiac, cerebrovascular, or peripheral vascular disease, since these patients were excluded from the current study. Based on the interaction of cerebrovascular lesions with plaque and tangle pathology, one might hypothesize that there would be a more identifiable treatment benefit.32

A key issue emerging from this study is the question of the optimal timing of treatment with a statin to achieve benefit in AD. This trial enrolled patients with well-established AD across a spectrum of mild to moderate severity. This may have been too far down the pathogenic cascade of AD leaving open to speculation whether the optimal period of early intervention might have been missed. Recently proposed revised research criteria for AD allow for diagnosis at the prodromal stage where treatment response might be different.33

This trial provides some important safety data on atorvastatin use within an elderly AD population. The safety profile for atorvastatin at a dosage of 80 mg daily was very similar to that in younger patients as reported in prior RCTs.34,35 The incidence of liver and muscle adverse events were also consistent with the rates reported in its product monograph.36 There were no hemorrhagic strokes encountered in the atorvastatin group and hepatic side effects resolved fully off when treatment was stopped. Contrary to concerns arising from observational studies,37,38 there were no adverse cognitive effects as measured by the ADAS-cog, MMSE, or CDR-SB; in fact, the numerical advantage favored atorvastatin on the primary cognitive measure, the ADAS-cog.

The primary analyses (repeated-measures models) take into account the time of each score evaluation. However, the LOCF uses the last visit of each patient and treats all those last visits the same way (without adjusting for the timing of those visits). It is notable that the LOCF approach presents a more optimistic picture of the treatment effect. In this trial, in which patients tended to show clinical decline over time, the differential withdrawal pattern (in which subjects on atorvastatin were more likely to discontinue prematurely from the study) created a small efficacy advantage for the subjects in the atorvastatin group when analyzed with the LOCF model. The patients who withdrew early due to reasons other than progression of dementia will appear to have higher efficacy in a LOCF model. This supports the use of a repeated-measures model as a superior choice for primary analysis in the setting of a longer-term clinical trial in AD.39

Treatment arm imbalances, especially the difference in function as measured by the ADCS-CGIC, lead us to conclude that the MRI substudy sample is not representative of the original cohort.

COINVESTIGATORS

LEADe Steering Committee— Howard Feldman (Chair), Vancouver, Canada; Rachelle Doody, Houston, TX; Roy Jones, Bath, UK; Miia Kivipelto, Stockholm, Sweden; David Lawrence Sparks, Sun City, AZ; David Waters, San Francisco, CA. LEADe Investigators— Australia: K. Boundy, H. Brodaty, R. Clarnette, D.S. Crimmins, J. Hecker, M.C. Woodward, M.W. Yates; Austria: P. Dal-Bianco, M. Haushofer, D. Poewe, M. Rainer; Canada: H. Bergman, S.E. Black, M.J. Borrie, R. Bouchard, H. Feldman, S. Gauthier, D.B. Hogan, G. Lacombe, C. Macknight, A.A. Robillard; Denmark: H. Braendgaard, M. Fink, S.G. Hasselbalch, S.A. Jakobsen, L.F. Lauritzen; Germany: K. Hager, R. Horn, H.-O. Karnath, A. Kurz, W. Maier, W. Mattern, H.-J. Moeller, J.B. Schulz, E. Steinhagen-Thiessen; South Africa: P.R. Laban, S. Lipschitz, C.M. Maud, F.C. Potocnik, A. Smuts; Spain: J.L.D. Ayuso, M.A. Barbera, A.F. Garcia, C.F. Garcia, J.L. Molinuevo, J. Peña-Casanova, M.B. Rovira; Sweden: N. Andreasen, M. Karlsson, H. Olofsson, J. Marcusson, S. Syversen; United Kingdom: A. Baldwin, A. Bayer, R.A. Bullock, R. Coles, F. Inglis, R.W. Jones, A.P. Passmore, D. Wilkinson; United States: P.G. Antuono, S.M. Asthana, C.B. Bernick, F.F. Chang, G.E. Cooper, S.T. Dekosky, J.E. De la Gandara, G.M. Dempsey, A.N. Dengiz, R.S. Doody, D.A. Drachman, E.A. Duboff, D.G. Folks, J.N. Goldstein, P.M. Green, P.J. Holland, R.F. Holub, M.S. Ismail, M. Kalafer, J.A. Kaye, C.S. Krishnasastry, K.A. Levin, P.D. Londborg, L.O. Macina, D.I. Margolin, J.A. Mattes, J.A. McCarthy, J.M. McKenney, M.C. Nunez, J.M. Pittard, S.H. Preskorn, W.G. Preston, M.A. Raskind, R.W. Richter, C.H. Sadowsky, B.E. Safirstein, S.P. Salloway, F.W. Schaerf, B.P. Seltzer, D.L. Sparks, L.J. Spikol, S.R. Stark, J.A. Stoukides, J.R. Tomlinson, M.A. Usman, M.F. Weiner, K.L. Wilks, J.L. Winston, M.L. Wolff, P.P. Zolnouni.

ACKNOWLEDGMENT

The authors acknowledge IXICO Ltd. (London, UK) for performing the atrophy measurement analysis of the image data collected for this study, including semiautomated brain and hippocampal delineations, as well as the boundary shift integral (BSI) technique. No assistance with content development or writing was provided, but editorial support in preparation for submission was provided by UBC Scientific Solutions and was funded by Pfizer Inc.

STUDY FUNDING

The LEADe study was funded by Pfizer. This manuscript was written by the authors on behalf of the LEADe Investigators. Employees of Pfizer contributed to the design and conduct of the study; the collection, management, analysis, and interpretation of the data; and review of the manuscript.

DISCLOSURE

The LEADe study was funded by Pfizer. Dr. Feldman is an employee of Bristol-Myers Squibb. Since January 1, 2009 Dr Feldman is on leave from University of British Columbia (UBC). The research and writing of this paper were undertaken while he was at UBC. Dr. Feldman served on the scientific advisory boards of Astra Zeneca, Bristol-Myers Squibb, Elan, Glia Scientific, Janssen, Pfizer, Merck Serono, Nestle, Novartis, Wyeth, Glaxo Smith Kline, Neuropharma, Servier, Shionogi/GSK, Targacept, Lundbeck, Pharmagenesis, Sanofi Synthelabo, Eli Lilly Boehringer Ingelheim; received travel expenses and/or honoraria for lectures or educational activities not funded by industry; received reimbursement of travel expenses and/or honoraria for lectures or educational activities sponsored by the Academy of Healthcare Education, Alpha Plus, Astra Zeneca, Cadmus Medea, Eisai, Glia Scientific, Janssen, Lundbeck, Medical Decision Point, Informed Direct PLC, MedPlan Communications, Novartis, and Pfizer. During the other years of the LEADe study (2002–2006), he received reimbursement of travel expenses and/or honoraria for lectures or educational activities sponsored by Academy of Healthcare Education, Alpha Plus Communications, Current Medical Directions, Eisai, Pfizer, Astra Zeneca, Janssen, the Memory Fitness Institute, OCC, Precept Medical, Lundbeck, MedPlan Communications, Excerpta Medica, Impact Communication, Shire, Embryon, Fundacion Maria Wolff Alzheimer, Kenes International, Prescott Medical, Alpha Plus Communications, Six Degrees, Hc3 Communications, Insyght, Ketchum, Novartis, Universal CIT, Center for Health Care Education, Glia Scientific, Health LogiX, and American Academy of Neurology; served as a member of the editorial board or an associate editor of Dementia and Geriatric Cognitive Disorders, Journal of Neurological Sciences, and the Canadian Alzheimer Disease Review; holds patent US Serial Number PCT/2007/070008 “Detecting and Treating Dementia” that may accrue revenue; receives royalties from Atlas of Alzheimer's Disease (Informa Health, London, 2007); served as a paid consultant to Novartis for an FDA Advisory Committee; served as a paid consultant for Migenix, Amorfix, United Biosource, and Ipsen; served on Pfizer Speakers Bureau; served as a member of steering committees of clinical trials sponsored by ONO, Elan and Pfizer; served as a member of data safety monitoring boards for clinical trials of Janssen, Myriad, and Novartis; received research funding from Elan, Lundbeck, Myriad, ONO, Servier, Sanofi Synthelabo, Janssen, and Glaxo Smith Kline; received grant-in-aid funding for the ACCORD Study from Eisai, Pfizer, Lilly and Janssen; and grant funding from the Alzheimer Society of Canada; received or receives research funding from the Canadian Institutes of Health Research (00725-000 [CoInvestigator], 179009 [PI], 74580 [PI], 73376 [PI]), Pacific Alzheimer Research Foundation (C06-01 [Coinvestigator]), NIH (U01AG24904 [Coinvestigator]); received travel reimbursement as PhD opponent Karolinska Institute; and holds stock options in BioMarin. Dr. Doody (during the time span from 2002 to present) served on medical and scientific advisory boards of Astellas, Athenagen, Comentis, Medivation, Myriad, Neurochem, Sonexa, and Zapaq and currently serves on such boards for Medivation and Sonexa; served on steering committees for Sanofi-Synthelabo and Ono; served on a data safety monitoring board for Debiopharm; and currently serves on a data safety monitoring board for GlaxoSmithKline; served on Pfizer Scholars selection committee; received reimbursement for travel to and from advisory board and consulting meetings for those that took place out of town; serves on the editorial boards of Alzheimer's Disease and Associated Disorders, Dementia and Geriatric Cognitive Disorders, and Alzheimer's Research and Therapy (a Biomedical Central journal); receives royalties from Alzheimer's Dementia (Carma Publishing, 2008); received honoraria for academic lectures from Eisai, Lundbeck, Pfizer; Forest (2002–2007 only); attended one or more ad hoc consultation meetings with Abbott, Allon, Amgen, Astellas, AstraZeneca, BristolMeyersSquibb, Dainippon, Eisai, Epix, Forest, Fujisawa, GlaxoSmithKline, Janssen, Lilly, MerckSerono, Merz, Novartis, Noven, Ocera, Pfizer, Saegis, Sanofi-Synthelabo, ScheringPlough, Suven, Takeda, Teva, Transition, Varinel, Voyager; received research funding from Eisai, Elan (now Jannsen Immunotherapy), Forest, GlaxoSmithKline, Myriad, Pfizer, Sanofi- Synthelabo, Teva, and Wyeth (now Pfizer); received research funding from NIH Alzheimer's Disease Cooperative Study Clinical trials (Site PI), NIDDK IR21DK062098 (Coinvestigator), and NIH/NCRR P20 RR020626 (Coinvestigator); received Zenith Award, Alzheimer's Association (PI); holds stock options in Medivation and Sonexa; acted as a consultant to a firm representing Eisai in a patent challenge case and to a firm representing Forest in a patent challenge case. Dr. Kivipelto served on the scientific advisory board for Pfizer and Elan; served as an editor for Journal of Alzheimer's Disease; was reimbursed for meetings organized by Pfizer, Novartis, and Jansen; received honoraria for lectures from Pfizer, Novartis, Jansen, Lundbeck and Merz.; received research support from Academy of Finland 120676 (PI) and 117 458 (PI). Dr. Sparks serves on the scientific advisory board (LEADe steering committee) for Pfizer; received funding from Pfizer to attend LEADe steering committee meetings; serves on the LEADe steering committee, Pfizer; served as a consultant for Resverlogix Pharma; received research support from Resverlogix Pharmaceuticals (0432-03-17607 [ICA]) and Pfizer (140015 [PI]); and receives research support from AZ Biomedical Research (0008-07 [PI]). Dr. Waters served on the scientific advisory board of Pfizer; served on steering committees for Pfizer; served on data safety monitoring boards for Pfizer, Oxigene and Merck Schering-Plough; served on advisory boards of Atherogenics, Merck Schering-Plough, Cortria, Aegerion; served on the endpoint committee of Biosante; served as an associate editor of Journal of the American College of Cardiology; received royalties from publishing Cardiology, Crawford and DiMarco, second edition (Harcourt Publishers, 2004); received honoraria for lectures from Pfizer and Merck; served as a consultant for Anthera; received research funding from Merck Schering-Plough; and held stock options in Anthera. Dr. Jones served on the scientific advisory board of Lundbeck, Pfizer/Medivation, Nutricia, Elan/Wyeth, Lilly, and Glaxo; received funding for travel from Lundbeck, Pfizer/Eisai, Shire, Merz, and Elan; received royalties from publishing Drug Treatment in Dementia, Blackwell (2001-2009); received research funding from Eisai/Pfizer, Lundbeck, Myriad, and Elan; received research funding from the Medical Research Council (grant 2007-001172-36 [Coinvestigator]); received research funding from BRACE (Bristol Research into Alzheimer's and Care of the Elderly [PI]); the Alzheimer's Research Trust (grant ART/NCG2005/1 [Coapplicant]); and served as a witness for the judicial Eisai appeal against NICE (The National Institute for Health and Clinical Excellence) in the UK. Dr. Schwam is an employee of Pfizer; and owns equity/ownership in Pfizer. Dr. Schindler is an employee of Pfizer; and owns equity/ownership in Pfizer. Dr. Hey-Hadavi is an employee of Pfizer; and owns equity/ownership in Pfizer. Dr. DeMicco is an employee of Pfizer; and owns equity/ownership in Pfizer. Dr. Breazna is an employee of Pfizer and he and his wife have equity/ownership interests in Pfizer.

Footnotes

  • Embedded Image

  • Embedded Image

  • Editorial, page 945

    Supplemental data at www.neurology.org

    e-Pub ahead of print on March 3, 2010, at www.neurology.org.

    Disclosure: Author disclosures are provided at the end of the article.

    Received January 27, 2009. Accepted in final form December 1, 2009.

REFERENCES

  1. ↵
    De Strooper B, Annaert W. Proteolytic processing and cell biological functions of the amyloid precursor protein. J Cell Sci 2000;113:1857–1870.
    OpenUrlAbstract/FREE Full Text
  2. Shobab LA, Hsiung GY, Feldman HH. Cholesterol in Alzheimer's disease. Lancet Neurol 2005;4:841–852.
    OpenUrlCrossRefPubMed
  3. ↵
    Wolozin B, Manger J, Bryant R, Cordy J, Green RC, McKee A. Re-assessing the relationship between cholesterol, statins and Alzheimer's disease. Acta Neurol Scand Suppl 2006;185:63–70.
    OpenUrlPubMed
  4. Austen B, Frears E, Davies H. Cholesterol upregulates production of A[beta] 1-40 and 1-42 in transfected cells. Neurobiol Aging 2000;21:254.
    OpenUrl
  5. Bodovitz S, Klein WL. Cholesterol modulates alpha-secretase cleavage of amyloid precursor protein. J Biol Chem 1996;271:4436–4440.
    OpenUrlAbstract/FREE Full Text
  6. Howland DS, Trusko SP, Savage MJ, et al. Modulation of secreted beta-amyloid precursor protein and amyloid beta-peptide in brain by cholesterol. J Biol Chem 1998;273:16576–16582.
    OpenUrlAbstract/FREE Full Text
  7. Simons M, Keller P, De Strooper B, Beyreuther K, Dotti CG, Simons K. Cholesterol depletion inhibits the generation of beta-amyloid in hippocampal neurons. Proc Natl Acad Sci USA 1998;95:6460–6464.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Sparks DL, Hunsaker JC 3rd, Scheff SW, Kryscio RJ, Henson JL, Markesbery WR. Cortical senile plaques in coronary artery disease, aging and Alzheimer's disease. Neurobiol Aging 1990;11:601–607.
    OpenUrlCrossRefPubMed
  9. ↵
    Fassbender K, Simons M, Bergmann C, et al. Simvastatin strongly reduces levels of Alzheimer's disease beta-amyloid peptides Abeta 42 and Abeta 40 in vitro and in vivo. Proc Natl Acad Sci USA 2001;98:5856–5861.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Hoglund K, Wallin A, Blennow K. Effect of statins on beta-amyloid metabolism in humans: potential importance for the development of senile plaques in Alzheimer's disease. Acta Neurol Scand Suppl 2006;185:87–92.
    OpenUrlPubMed
  11. ↵
    Jick H, Zornberg GL, Jick SS, Seshadri S, Drachman DA. Statins and the risk of dementia. Lancet 2000;356:1627–1631.
    OpenUrlCrossRefPubMed
  12. Wolozin B, Wang SW, Li NC, Lee A, Lee TA, Kazis LE. Simvastatin is associated with a reduced incidence of dementia and Parkinson's disease. BMC Med 2007;5:20.
  13. ↵
    Arvanitakis Z, Schneider JA, Wilson RS, et al. Statins, incident Alzheimer disease, change in cognitive function, and neuropathology. Neurology 2008;70:1795–802.
    OpenUrlAbstract/FREE Full Text
  14. Haag MD, Hofman A, Koudstaal PJ, Stricker BH, Breteler MM. Statins are associated with a reduced risk of Alzheimer disease regardless of lipophilicity: The Rotterdam Study. J Neurol Neurosurg Psychiatry 2009;80:13–17.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002;360:7–22.
    OpenUrlCrossRefPubMed
  16. Shepherd J, Blauw GJ, Murphy MB, et al. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002;360:1623–1630.
    OpenUrlCrossRefPubMed
  17. ↵
    Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for the treatment of mild to moderate Alzheimer disease: preliminary results. Arch Neurol 2005;62:753–757.
    OpenUrlCrossRefPubMed
  18. ↵
    World Medical Association. Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA 2000;284:3043–3045.
    OpenUrlCrossRefPubMed
  19. ↵
    Jones RW, Kivipelto M, Feldman H, et al. The Atorvastatin/Donepezil in Alzheimer's Disease Study (LEADe): design and baseline characteristics. Alzheimer Dement 2008;4:145–153.
    OpenUrl
  20. ↵
    Sled JG, Zijdenbos AP, Evans AC. A nonparametric method for automatic correction of intensity nonuniformity in MRI data. IEEE Trans Med Imaging 1998;17:87–97.
    OpenUrlCrossRefPubMed
  21. ↵
    McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease. Neurology 1984;34:939–944.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975;12:189–198.
    OpenUrlCrossRefPubMed
  23. ↵
    Rosen WG, Terry RD, Fuld PA, Katzman R, Peck A. Pathological verification of ischemic score in differentiation of dementias. Ann Neurol 1980;7:486–488.
    OpenUrlCrossRefPubMed
  24. ↵
    Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer's disease. Am J Psychiatry 1984;141:1356–1364.
    OpenUrlPubMed
  25. ↵
    Schneider LS, Olin JT, Doody RS, et al. Validity and reliability of the Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change: The Alzheimer's Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997;11 suppl 2:S22–S32.
    OpenUrlCrossRef
  26. ↵
    Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology 1994;44:2308–2314.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Mohs RC, Knopman D, Petersen RC, et al. Development of cognitive instruments for use in clinical trials of antidementia drugs: additions to the Alzheimer's Disease Assessment Scale that broaden its scope: The Alzheimer's Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997;11 suppl 2:S13–S21.
    OpenUrl
  28. ↵
    Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology 1993;43:2412–2414.
    OpenUrlFREE Full Text
  29. ↵
    Mohs RC, Doody RS, Morris JC, et al. A 1-year, placebo-controlled preservation of function survival study of donepezil in AD patients. Neurology 2001;57:481–488.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Freeborough PA, Fox NC. The boundary shift integral: an accurate and robust measure of cerebral volume changes from registered repeat MRI. IEEE Trans Med Imaging 1997;16:623–629.
    OpenUrlCrossRefPubMed
  31. ↵
    Grundy SM, Cleeman JI, Merz CN, et al. Implications of recent clinical trials for the National Cholesterol Education Program Adult Treatment Panel III guidelines. Circulation 2004;110:227–239.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Snowdon DA, Greiner LH, Mortimer JA, Riley KP, Greiner PA, Markesbery WR. Brain infarction and the clinical expression of Alzheimer disease: The Nun Study. JAMA 1997;277:813–817.
    OpenUrlCrossRefPubMed
  33. ↵
    Dubois B, Feldman HH, Jacova C, et al. Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS-ADRDA criteria. Lancet Neurol 2007;6:734–746.
    OpenUrlCrossRefPubMed
  34. ↵
    LaRosa JC, Grundy SM, Waters DD, et al. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 2005;352:1425–1435.
    OpenUrlCrossRefPubMed
  35. Amarenco P, Bogousslavsky J, Callahan A 3rd, et al. High-dose atorvastatin after stroke or transient ischemic attack. N Engl J Med 2006;355:549–559.
    OpenUrlCrossRefPubMed
  36. ↵
    US Prescribing Information: Lipitor® (atorvastatin calcium) tablets. New York, NY: Pfizer Inc.; November 2007.
  37. ↵
    Galatti L, Polimeni G, Salvo F, Romani M, Sessa A, Spina E. Short-term memory loss associated with rosuvastatin. Pharmacotherapy 2006;26:1190–1192.
    OpenUrlCrossRefPubMed
  38. King DS, Wilburn AJ, Wofford MR, Harrell TK, Lindley BJ, Jones DW. Cognitive impairment associated with atorvastatin and simvastatin. Pharmacotherapy 2003;23:1663–1667.
    OpenUrlCrossRefPubMed
  39. ↵
    Siddiqui O, Hung HMJ, O'Neill R. MMRM vs. LOCF: a comprehensive comparison based on simulation study and 25 NDA datasets. J Biopharm Stats 2009;19:227–246.
    OpenUrl

Letters: Rapid online correspondence

No comments have been published for this article.
Comment

REQUIREMENTS

If you are uploading a letter concerning an article:
You must have updated your disclosures within six months: http://submit.neurology.org

Your co-authors must send a completed Publishing Agreement Form to Neurology Staff (not necessary for the lead/corresponding author as the form below will suffice) before you upload your comment.

If you are responding to a comment that was written about an article you originally authored:
You (and co-authors) do not need to fill out forms or check disclosures as author forms are still valid
and apply to letter.

Submission specifications:

  • Submissions must be < 200 words with < 5 references. Reference 1 must be the article on which you are commenting.
  • Submissions should not have more than 5 authors. (Exception: original author replies can include all original authors of the article)
  • Submit only on articles published within 6 months of issue date.
  • Do not be redundant. Read any comments already posted on the article prior to submission.
  • Submitted comments are subject to editing and editor review prior to posting.

More guidelines and information on Disputes & Debates

Compose Comment

More information about text formats

Plain text

  • No HTML tags allowed.
  • Web page addresses and e-mail addresses turn into links automatically.
  • Lines and paragraphs break automatically.
Author Information
NOTE: The first author must also be the corresponding author of the comment.
First or given name, e.g. 'Peter'.
Your last, or family, name, e.g. 'MacMoody'.
Your email address, e.g. higgs-boson@gmail.com
Your role and/or occupation, e.g. 'Orthopedic Surgeon'.
Your organization or institution (if applicable), e.g. 'Royal Free Hospital'.
Publishing Agreement
NOTE: All authors, besides the first/corresponding author, must complete a separate Publishing Agreement Form and provide via email to the editorial office before comments can be posted.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.

Vertical Tabs

You May Also be Interested in

Back to top
  • Article
    • Abstract
    • Glossary
    • METHODS
    • RESULTS
    • DISCUSSION
    • COINVESTIGATORS
    • ACKNOWLEDGMENT
    • STUDY FUNDING
    • DISCLOSURE
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Info & Disclosures
Advertisement

Hemiplegic Migraine Associated With PRRT2 Variations A Clinical and Genetic Study

Dr. Robert Shapiro and Dr. Amynah Pradhan

► Watch

Related Articles

  • Clinical trial efforts in Alzheimer diseaseWhy test statins?

Topics Discussed

  • Class II
  • Clinical trials Randomized controlled (CONSORT agreement)
  • Alzheimer's disease

Alert Me

  • Alert me when eletters are published
Neurology: 100 (6)

Articles

  • Ahead of Print
  • Current Issue
  • Past Issues
  • Popular Articles
  • Translations

About

  • About the Journals
  • Ethics Policies
  • Editors & Editorial Board
  • Contact Us
  • Advertise

Submit

  • Author Center
  • Submit a Manuscript
  • Information for Reviewers
  • AAN Guidelines
  • Permissions

Subscribers

  • Subscribe
  • Activate a Subscription
  • Sign up for eAlerts
  • RSS Feed
Site Logo
  • Visit neurology Template on Facebook
  • Follow neurology Template on Twitter
  • Visit Neurology on YouTube
  • Neurology
  • Neurology: Clinical Practice
  • Neurology: Education
  • Neurology: Genetics
  • Neurology: Neuroimmunology & Neuroinflammation
  • AAN.com
  • AANnews
  • Continuum
  • Brain & Life
  • Neurology Today

Wolters Kluwer Logo

Neurology | Print ISSN:0028-3878
Online ISSN:1526-632X

© 2023 American Academy of Neurology

  • Privacy Policy
  • Feedback
  • Advertise