You are on page 1of 4

BRIEF REPORTS

A Pilot Study of Vitamins to E levated plasma homocysteine, which is associated


with increased risk of vascular disease, may also be
Lower Plasma associated with risk1,2 and radiologically demonstrated
progression1 of Alzheimer disease (AD). In vitro and in
Homocysteine Levels in vivo studies suggest that homocysteine contributes to
Alzheimer Disease excitotoxic damage3 and augments amyloid toxicity,4
and thus may play an important role in AD-related neu-
rodegeneration. Reduction of homocysteine levels with
Paul S. Aisen, M.D. vitamin supplements thus represents a plausible thera-
Susan Egelko, Ph.D. peutic strategy.
Howard Andrews, Ph.D. Previous studies in persons not diagnosed with AD
suggest that high-dose vitamin supplementation can re-
Ramon Diaz-Arrastia, M.D.
duce plasma levels of homocysteine by 25%.5 In the
Myron Weiner, M.D. United States, grain products have been fortified with
Charles DeCarli, M.D. folic acid since 1998, an intervention aimed at reducing
William Jagust, M.D. neonatal neural tube defects known to be linked to folic
Joshua W. Miller, Ph.D. acid deficiency. This fortification has reduced mean ho-
mocysteine levels by 7%6 and thus may have reduced
Ralph Green, M.D. the potential impact of high-dose vitamin supplemen-
Karen Bell, M.D. tation.
Mary Sano, Ph.D. In preparation for a large multicenter trial to deter-
mine whether homocysteine reduction slows progres-
sion in persons with AD, we conducted the present
Objective: Authors determined the impact of high-
study to determine the impact of high-dose vitamin sup-
dose vitamin supplements on plasma homocysteine
plementation on fasting and post–methionine-loading
levels in patients with Alzheimer disease (AD). Meth-
homocysteine in American subjects with AD. In partic-
ods: Authors used an open-label trial of folic acid, vi-
ular, we sought to determine whether high-dose vitamin
tamin B12, and vitamin B6, in combination for 8
supplements reduce plasma homocysteine despite folic
weeks, with measurement of plasma homocysteine
acid fortification of grain products in the United States
levels in the fasting state and after methionine-
and whether high-dose supplements reduce homocys-
loading. A total of 69 subjects with AD were enrolled,
teine levels in users of daily multivitamins.
including 33 who were taking standard multivitamin
supplements; 66 were available at 8-week follow-up.
Results: The high-dose vitamin regimen was associ-
ated with a significant reduction in fasting and post– METHODS
methionine-loading homocysteine. Reductions were
greater in the subgroup not using multivitamins, but Study Design
were also significant in the multivitamin users. Con-
clusion: High-dose vitamin supplementation reduces The 8-week study used an open-label design to de-
homocysteine levels in patients with AD. The effect of termine the impact of high-dose vitamin supplementa-
supplementation on rate of cognitive decline will be tion on homocysteine levels. The treatment regimen
assessed later in a randomized, double-blind study. consisted of folic acid 5 mg (given as five 1-mg tablets
(Am J Geriatr Psychiatry 2003; 11:246–249) taken simultaneously), plus vitamin B12 1 mg, plus vi-
tamin B6 50 mg, administered together each morning;

Received November 18, 2002; revised December 23, 2002, January 3, 2003; accepted January 3, 2003. From the Department of Neurology,
Georgetown University Medical Center, Washington, DC. Address correspondence to Paul S. Aisen, M.D., Department of Neurology, Georgetown
University Medical Center, Building D, Suite 207, 4000 Reservoir Road NW, Washington, DC 20007.
Copyright 䉷 2003 American Association for Geriatric Psychiatry

246 Am J Geriatr Psychiatry 11:2, March-April 2003


Aisen et al.

this regimen was selected to provide optimal reduction correlation coefficients among these variables as well as
in homocysteine.5 conducting a simultaneous multiple-regression analysis
Medically stable subjects with probable AD7 were predicting change in fasting levels of homocysteine.
eligible for enrollment regardless of level of cognitive
impairment if a caregiver was available to supervise ad-
ministration of the vitamin regimen. Subjects were ex-
cluded if there was evidence of significant renal insuf- RESULTS
ficiency (creatinine ⬎1.5 mg/dL), a history of B12 or
folate deficiency, use of vitamin supplements containing Participant Flow and Follow-Up
greater than 400 lg of folic acid, regular use of vitamin
B12 injections, or use of medications known to influence A total of 69 subjects (44 women) were enrolled at
homocysteine metabolism (e.g., methotrexate, azathio- the four sites; 54 were taking cholinesterase inhibitors
prine, phenytoin). Subjects who were taking multivita- at the time of enrollment, and 33 were taking multivi-
mins at the time of enrollment continued these multi- tamins. The typical multivitamin used by study partici-
vitamins in addition to the study regimen during the pants contained 400 lg folic acid, 25 lg vitamin B12,
trial. and 3 mg–25 mg vitamin B6. The mean age was 70.8
The cognitive/behavioral assessments included the years (standard deviation [SD]: 9.4), and the mean du-
Folstein Mini-Mental State Exam (MMSE),8 the Alzhei- ration of AD was 3.3 (SD: 2.5) years. At enrollment, the
mer’s Disease Assessment Scale9 cognitive subscale mean MMSE score was 19.3 (SD: 7.0); mean ADAScog
(ADAScog), and the Geriatric Depression Scale (GDS).10 score was 34.9 (SD: 9.5); and mean GDS score was 6.8
Blood specimens were obtained for measurement of ho- (SD: 5.5). There were no significant differences in base-
mocysteine in the fasting state and 2 hours after admin- line characteristics between multivitamin users and
istration of L-methionine (0.1 g/kg body weight). non-users.
Pill counts at the Week 8 visit indicated that com-
Laboratory Measures pliance with each component of the high-dose supple-
ment regimen exceeded 80%.
Plasma homocysteine was determined by high- Table 1 shows the effect of treatment on fasting and
pressure liquid chromatography with post-column fluo- post–methionine-loading homocysteine levels. Overall,
rescence detection.11 Normal quality control (mean 7.5 there was a decline in both fasting and post–methionine-
lmol/L): intra-assay coefficient of variation (CV): 1.3%, loading homocysteine. As expected, change was greater
inter-assay CV: 4.2%; high quality control (mean 27.8 among non-users of multivitamins than users in fasting
lmol/L): intra-assay CV: 1.1%, inter-assay CV: 3.0%. homocysteine level (t[45.6]⳱2.80; p⳱0.008, using
Vitamin B12 was measured by automated chemilu- a separate variance estimate), as well as in post–
minescence assay (ACS 180, Bayer Diagnostics, Tarry- methionine-loading homocysteine level (t[46.9]⳱2.31;
town, NY); red blood cell folate, by radioassay (Quan- p⳱0.03, also using a separate variance estimate). The
taphase II, BioRad Diagnostics, Hercules, CA); and change in fasting homocysteine level was shown to be
plasma pyridoxal-5’-phosphate (an indicator of tissue related to change in red blood cell folate (Pearson
stores of vitamin B6) by radioenzymatic assay (ALPCO, r[64]⳱0.29; p⳱0.02), but not with change in B12 or
Windham, NH). MTHFR genotype was determined by pyridoxal-5’-phosphate.
the method of Frosst.12 A paired t-test comparing baseline and 8-week
MMSE scores showed no treatment effect on MMSE:
Statistical Analysis mean baseline MMSE was19.2 (SD: 7.0), versus 8-week
MMSE of 19.3 (SD: 7.7); (t[62]⳱0.25; p⳱0.80).
The primary analysis was a comparison by paired t- In zero-order correlations conducted among study
tests of the mean change in fasting and post–methionine- variables, there was a correlation between baseline fast-
loading homocysteine levels from baseline to the 8-week ing homocysteine level and age (r[66]⳱0.23; p⳱0.06);
study visit. Further analyses examined the relationship there was no correlation between fasting homocysteine
among demographic, clinical, and genetic factors and levels and MMSE, ADAScog, or GDS. In a simultaneous
homocysteine levels, by calculating zero-order Pearson multiple-regression equation in which fasting homocys-

Am J Geriatr Psychiatry 11:2, March-April 2003 247


Vitamins and Homocysteine in AD

teine level, vitamin use, age, and gender were entered with both the CC (9.0 [SD: 2.9]) and CT (9.4 [SD: 2.8])
as predictors, the most significant predictor of decline groups. MTHFR genotype was not a significant predic-
in fasting homocysteine at 8 weeks was baseline fasting tor of change in fasting homocysteine level with treat-
homocysteine (partial correlation: r[60]⳱0.73; ment in an ANCOVA, with baseline homocysteine level
p⳱0.001). In this equation, after controlling for base- as a covariate: F[2,62]⳱1.71; p⳱0.189.
line fasting homocysteine, multivitamin use was not re-
lated to change in fasting homocysteine (r⳱ –0.15; Adverse Events
p⳱0.25), but age (r⳱0.28; p⳱0.03) and gender (r⳱
–0.174; p⳱0.05) each showed predictive value. There were no serious adverse events, and in no
Subjects were divided into two groups by median instance was the vitamin regimen discontinued as a re-
baseline fasting homocysteine level (cutpoint of 8.575 sult of adverse events. In no instance was an adverse
lmol/L). There was a significant decline in mean fasting symptom judged likely to be related to the study inter-
homocysteine levels with treatment in both the high vention.
group (baseline: 11.7 [SD: 3.2] lmol/L; supplemented:
9.0 [SD: 2.4] lmol/L; t[32]⳱5.18; p⳱0.001; change:
23%) and the low group (baseline: 7.4 [SD: 1.1] lmol/ DISCUSSION
L; supplemented: 6.7 [SD: 1.3] lmol/L; t[32]⳱3.28;
p⳱0.003; change: 10%) in paired t-tests. Treatment of patients with AD, living in the United
MTHFR analysis revealed that 29 (42%) of subjects States, with a regimen of high-dose folic acid, B6, and
carried the CC genotype; 34 (49%) were heterozygous B12 for 8 weeks reduced plasma homocysteine. Fasting
CT, and 6 (9%) were homozygous TT. There was a sig- and post–methionine-loading levels declined with this
nificant association between MTHFR genotype and treatment. The extent of decline was related to baseline
baseline fasting homocysteine levels in the overall one- homocysteine levels. Subjects who were using multivi-
way ANOVA across the three genotype groups tamins at the time of enrollment showed decline in
(F[2,66]⳱5.67; p⳱0.005). In the post-hoc Tukey HSD plasma homocysteine with the treatment regimen, al-
test to identify pairwise group differences, as expected, though the decline was greater in non-users of multi-
the TT genotype group had a significantly higher mean vitamins. Similarly, subjects with baseline plasma ho-
fasting homocysteine level (13.6 [SD: 4.8]), compared mocysteine levels below and above the median showed

TABLE 1. Homocysteine and vitamin levelsa before and after supplementation in multivitamin users and non-users (Nⴔ66)
Baseline Supplemented Mean Change p
Fasting homocysteine 9.6 (3.2) 7.8 (2.3) 1.7 (2.5) 0.001
Post–methionine-loading homocysteine (n⳱62) 27.0 (10.2) 21.4 (5.9) 5.6 (9.3) 0.001
Red blood cell folate 434 (166) 819 (259) 385 (255) 0.001
Plasma B12 560 (343) 1,158 (514) 598 (408) 0.001
Plasma pyridoxal-5⬘-phosphate 107 (120) 314 (171) 207 (157) 0.001
Multivitamin non-users (n⳱35)
Fasting homocysteine 10.4 (3.8)b 7.9 (2.6) 2.5 (3.1) 0.001
Post–methionine-loading homocysteine (n⳱33) 29.9 (12.2)c 21.9 (5.7) 8.0 (11.2) 0.001
Red blood cell folate 361 (127) 834 (289) 473 (287) 0.001
Plasma B12 514 (345) 1,174 (567) 660 (434) 0.001
Plasma pyridoxal-5⬘-phosphate 55 (57) 284 (169) 229 (174) 0.001
Multivitamin users (n⳱31)
Fasting homocysteine 8.7 (2.1)b 7.8 (1.9) 0.9 (1.2) 0.005
Post–methionine-loading homocysteine (n⳱29) 23.7 (6.0)c 20.8 (6.2) 2.9 (5.3) 0.005
Red blood cell folate 516 (168) 802 (225) 286 (168) 0.001
Plasma B12 613 (339) 1,140 (456) 527 (363) 0.001
Plasma pyridoxal-5⬘-phosphate 166 (143) 349 (168) 183 (135) 0.001
Note: Values are mean (standard deviation).
a
Units: homocysteine: lmol/L; red blood cell folate, ng/ml; B12, pg/ml; pyridoxal-5⬘-phosphate, nmol/L; p values for paired t-tests.
b
Multivitamin non-users had higher fasting homocysteine levels at baseline than multivitamin users: t[62.1]⳱1.98; p⳱0.05.
c
Multivitamin non-users had higher methionine-loaded homocysteine levels at baseline than multivitamin users: t[52.0]⳱2.33; p⳱0.02.

248 Am J Geriatr Psychiatry 11:2, March-April 2003


Aisen et al.

decline in levels with the treatment regimen. Thus, de- spective trial. A multicenter trial of this type, funded by
spite fortification of grain products with folic acid and the National Institute on Aging, will begin early in 2003.
use of multivitamins containing the recommended daily
The following individuals helped conduct the
allowances of folic acid, B6, and B12, there was a reduc-
study: Carolyn Ward, MSPH; Mary Ann Cachola; Kath-
tion in plasma homocysteine in this population with the
leen Redington, M.D., Ph.D. (Georgetown University);
use of high-dose supplements. Baseline homocysteine Nicholas Scarmeas, M.D.; Gina Garcia-Camilo, M.D.;
levels were higher in C677T homozygotes (TT); how- Karen Andrews (Columbia University); Carol Moore,
ever, significant decline in fasting and post–methionine- M.A. (University of Texas, Southwestern); Rebecca F.
loading homocysteine levels occurred in subjects with Cotterman; Marisa I. Ramos (University of California,
each of the three genotypes. Davis).
The present study confirms that homocysteine re- This study was supported by a grant from the In-
duction is feasible in the American AD population. Max- stitute for the Study of Aging, as well as a grant (5
imal reduction occurred with supplementation in sub- M01-RR13297) from the General Clinical Research
jects with relatively high baseline levels. However, Center Program of the National Center for Research
reduction occurred in unselected AD patients, includ- Resources, and grants (R01-AG17861 and U01-
ing those taking multivitamin supplements. Whether AG10483) from the National Institute on Aging. Vi-
homocysteine reduction will slow AD progression must tamin B12 and vitamin B6 tablets were kindly supplied
be tested in a long-term, randomized, controlled, pro- by Rexall Sundown, Inc.

References
1. Clarke R, Smith AD, Jobst KA, et al: Folate, vitamin B12, and serum fortification on plasma folate and total homocysteine concentra-
total homocysteine levels in confirmed Alzheimer disease. Arch tions. N Engl J Med 1999; 340:1449–1454
Neurol 1998; 55:1449–1455 7. McKhann G, Drachman D, Folstein M, et al: Clinical diagnosis of
2. Seshadri S, Beiser A, Selhub J, et al: Plasma homocysteine as a Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group.
risk factor for dementia and Alzheimer’s disease. N Engl J Med Neurology 1984; 34:939–944
2002; 346:476–483 8. Folstein M, Folstein S, McHugh P: The Mini-Mental State Exam. J
3. Lipton SA, Kim WK, Choi YB, et al: Neurotoxicity associated with Psychiatr Res 1975; 12:189–198
dual actions of homocysteine at the N-methyl-D-aspartate recep- 9. Rosen WG, Mohs RC, Davis KL: A new rating scale for Alzhei-
tor. Proc Natl Acad Sci U S A 1997; 94:5923–5928 mer’s disease. Am J Psychiatry 1984; 141:1356–1364
4. Kruman II, Kumaravel TS, Lohani A, et al: Folic acid deficiency 10. Yesavage JA: Geriatric Depression Scale. Psychopharmacol Bull
and homocysteine impair DNA repair in hippocampal neurons 1988; 24:709–711
and sensitize them to amyloid toxicity in experimental models 11. Gilfix BM, Blank DW, Rosenblatt DS: Novel reductant for deter-
of Alzheimer’s disease. J Neurosci 2002; 22:1752–1762 mination of total plasma homocysteine. Clin Chem 1997;
5. Homocysteine Lowering Trialists’ Collaboration: Lowering blood 43:687–688
homocysteine with folic acid-based supplements: metaanalysis of 12. Frosst P, Blom HJ, Milos R, et al: A candidate genetic risk factor
randomised trials. BMJ 1998; 316:894–898 for vascular disease: a common mutation in methylenetetrahy-
6. Jacques PF, Selhub J, Bostom AG, et al: The effect of folic acid drofolate reductase. Nat Genet 1995; 10:111–113

Am J Geriatr Psychiatry 11:2, March-April 2003 249

You might also like