Вы находитесь на странице: 1из 10

Vascular Medicine

B Vitamins and the Risk of Total Mortality and


Cardiovascular Disease in End-Stage Renal Disease
Results of a Randomized Controlled Trial
Judith Heinz, MSc; Siegfried Kropf, PhD; Ute Domröse, MD; Sabine Westphal, MD;
Katrin Borucki, MD; Claus Luley, MD; Klaus H. Neumann, MD; Jutta Dierkes, PhD

Background—In observational studies, hyperhomocysteinemia has been found to be a risk factor for total mortality and
cardiovascular events in patients with end-stage renal disease. These patients have grossly elevated homocysteine levels
that can be lowered by supplementation with folic acid and vitamin B12. We conducted a randomized clinical trial with
B vitamins to reduce homocysteine levels and therefore cardiovascular events and total mortality.
Methods and Results—This randomized, double-blind multicenter study was conducted in 33 dialysis centers in north and
east Germany between July 2002 and July 2008. We randomly assigned 650 patients with end-stage renal disease who
were undergoing hemodialysis to 2 postdialysis treatments: 5 mg folic acid, 50 ␮g vitamin B12, and 20 mg vitamin B6
(active treatment) or 0.2 mg folic acid, 4 ␮g vitamin B12, and 1.0 mg vitamin B6 (placebo) given 3 times per week for
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

an average of 2 years. The primary outcome was total mortality; the secondary outcome was fatal and nonfatal
cardiovascular events. The primary outcome occurred in 102 patients (31%) receiving the active treatment and in 92
(28%) receiving placebo (hazard ratio, 1.13; 95% confidence interval, 0.85 to 1.50; P⫽0.51). The secondary outcome
occurred in 83 patients (25%) receiving the active treatment and in 98 (30%) receiving placebo (hazard ratio, 0.80; 95%
confidence interval, 0.60 to 1.07; P⫽0.13).
Conclusions—Increased intake of folic acid, vitamin B12, and vitamin B6 did not reduce total mortality and had no
significant effect on the risk of cardiovascular events in patients with end-stage renal disease.
Clinical Trial Registration—URL: www.anzctr.org.au. Unique identifier: ACTRN12609000911291. URL:
www.cochrane-renal.org. Unique identifier: CRG010600027.
(Circulation. 2010;121:1432-1438.)
Key Words: B vitamins 䡲 cardiovascular diseases 䡲 hemodialysis 䡲 homocysteine 䡲 kidney
䡲 mortality 䡲 prevention

teine levels in patients with ESRD are ⬇25 ␮mol/L in those


A lthough a number of prospective observational studies
have shown that hyperhomocysteinemia is associated
with cardiovascular morbidity and mortality,1–3 clinical stud-
who take folic acid supplementation or fortification and
⬇35 ␮mol/L in those who do not receive additional vita-
ies aimed at lowering homocysteine levels in patients with mins.15 Indeed, similar to the situation in populations without
cardiovascular disease have yielded disappointing results; renal disease, homocysteine is related to cardiovascular
overall, lowering homocysteine by vitamin intervention re- morbidity and mortality in patients with ESRD, as has been
duced neither cardiovascular events nor mortality.4 – 8 shown in a recent meta-analysis.15 Although the homocys-
teine levels are much higher in patients with ESRD than in
Editorial see p 1379 other population groups, vitamin supplementation has been
Clinical Perspective on p 1438 shown to be effective in lowering those levels,16,17 although
As far as cardiovascular disease and homocysteine are so far these studies have not been summarized and no
concerned, patients with end-stage renal disease (ESRD) are consensus has been reached on the amounts necessary or the
of particular interest because they experience high rates of route of administration. In addition, the clinical effect of such
cardiovascular disease and high rates of mortality9 –11 and vitamin intervention on cardiovascular disease and mortality
exhibit the highest homocysteine concentrations, except for has not been demonstrated in randomized clinical trials in
patients with homocystinuria.12–14 On average, the homocys- patients with ESRD without additional vitamin supplementa-

Received August 27, 2009; accepted January 22, 2010.


From the Institute of Clinical Chemistry (J.H., S.W., K.B., C.L., J.D.), Division of Nephrology (U.D., K.H.N.), and Institute of Biometry and Medical
Informatics (S.K.), Magdeburg University Hospital, Magdeburg, Germany.
Correspondence to Judith Heinz, Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistraße
52, D-20246 Hamburg, Germany. E-mail j.heinz@uke.de
© 2010 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.109.904672

1432
Heinz et al B Vitamins and Mortality in Dialysis Patients 1433

tion or fortification. We therefore conducted a randomized Baseline and Follow-Up Investigations


clinical trial to see whether homocysteine-lowering therapy On entry into the study, demographic and clinical data were
with folic acid, vitamin B12, and vitamin B6 could reduce total recorded, as well as the use of medication and lifestyle parameters.
A nonfasting blood sample was collected before the first dialysis
mortality and cardiovascular events in patients with ESRD session after a weekend for the determination of serum and red blood
treated with hemodialysis. cell levels of folate, cobalamin, pyridoxal-5-phosphate (PLP), and
homocysteine. Aliquots of the sample were also used to determine
blood count and clinical chemical, lipid, coagulation, and inflamma-
Methods tion parameters. In a subgroup of patients (n⫽97), homocysteine,
Study Design serum folate, cobalamin, and PLP were also measured after 6 months
of participation in the study.
The objective of this double-blind, placebo-controlled, randomized
Plasma homocysteine was determined in EDTA plasma by high-
multicenter trial was to see whether therapy with homocysteine- performance liquid chromatography with fluorescence detection.20
lowering B vitamins could reduce total mortality and the risk of Serum cobalamin and serum folate were analyzed with commercial
cardiovascular events in patients with ESRD. The trial design and the test kits (Elecsys Module E170, Roche Diagnostics). PLP was
baseline data were reported recently.18 The study was conducted in determined in EDTA plasma by high-performance liquid chroma-
dialysis centers in north and east Germany between July 2002 and tography (ImmuChrom, Bensheim, Germany). All these laboratory
July 2008. It was coordinated by the Institute of Clinical Chemistry analyses were performed in the central laboratory of the Magdeburg
at the Otto-von-Guericke University in Magdeburg, Germany, and University Hospital.
sponsored by the School of Medicine at that university, Roche Follow-up visits were arranged once per year for every patient.
Diagnostics (Mannheim, Germany; laboratory measurements), and The patients’ medical records were checked for death, major cardio-
Fresenius Medical Care (Bad Homburg, Germany; study drug and vascular events, hospitalizations, and compliance with the trial
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

matching placebo). The sponsors were not involved in the design, medication. Dropouts resulting from transplantation, change in
execution, analysis, or reporting of the results of this study. Safety of dialysis center, or any other reasons were documented. In case of any
the intervention and scientific integrity of the study were supervised uncertainty, the physician in charge of the respective patient was
by an independent data and safety monitoring board. The study was consulted.
approved by the ethics committees of the School of Medicine of
Otto-von-Guericke University Magdeburg and the appropriate med- Trial Outcomes
ical associations in the respective federal states of Germany. All The primary outcome was total mortality. The secondary outcome
patients gave informed consent. was the occurrence of the first fatal or nonfatal cardiovascular event
(myocardial infarction, unstable angina pectoris, coronary vascular-
Study Population ization procedures, sudden cardiac death, stroke, peripheral artery
disease, pulmonary embolism, and thromboses). Shunt thromboses
Men and women between 20 and 80 years of age with ESRD treated
were not regarded as an end point.
for at least 1 month by hemodialysis were enrolled, regardless of
Deaths were confirmed by hospital discharge summaries, autopsy
their homocysteine levels. The exclusion criteria were acute coro- reports, or the responsible physicians. Causes of death were catego-
nary events within 6 weeks before randomization, active malignant rized as cardiac (myocardial infarction, sudden cardiac death, pul-
tumor, pregnancy, lactation, and addiction to drugs or alcohol. monary edema), vascular (stroke, pulmonary embolism, thromboses,
Patients who had been taking vitamins before recruitment were mesenterial infarction, rupture of an aortic aneurysm), sepsis and
included after a washout phase of at least 8 weeks. The patients, who infections, tumors, and other causes (consequences of surgery, fluid
came from 33 dialysis centers in Germany, had been put on overload and withdrawal from dialysis, cirrhosis of the liver,
hemodialysis because of diabetic nephropathy (n⫽184), chronic cachexia, diabetic complications, accidents, suicides, shunt compli-
glomerulonephritis (n⫽140), interstitial nephropathy (n⫽84), poly- cations, gastrointestinal bleeding, cerebral hemorrhage, hyperkale-
cystic nephropathy (n⫽75), vascular nephropathy (n⫽63), other mia). In 15 cases, the causes of death could not be established and
reasons (n⫽56), and unknown reasons (n⫽48). Reason for referral to were recorded as unknown.
hemodialysis was not a criterion for inclusion or exclusion. Cardiovascular events were identified by a review of patients’
medical records and by consultation with the responsible physi-
Intervention cians. Myocardial infarction was diagnosed if at least 2 of the
Patients were randomized to 2 treatment groups. Each patient following criteria had been fulfilled according to standard proce-
dures: clinical status, elevated laboratory parameters (myocardi-
received vitamins with an originally assigned code number. The code
um-specific enzymes, myoglobin), and changes in the ECG.
numbers were kept within the central pharmacy of the university
Catheterization was performed in cases of suspected myocardial
hospital. All study investigators, staff, and participants were blinded
infarction or unstable angina pectoris. Surgical revascularization was
to the randomization procedure and treatment assignments. carried out after myocardial infarction, in unstable angina pectoris, or
One tablet contained either 2.5 mg folic acid, 25 ␮g cobalamin, when clinical signs had been detected by catheterization and the
and 10 mg vitamin B6 (active treatment) or 0.1 mg folic acid, 2 ␮g patients’ general conditions permitted surgery. Strokes and ischemic
cobalamin, and 0.5 mg vitamin B6 (placebo). After each dialysis insults were verified by computed tomography. Peripheral artery
session (usually 3 times per week), 2 tablets were taken orally disease was diagnosed according to the Fontaine stages or on the
under the supervision of a nurse. The active treatment and the basis of ⬎50% stenoses detected angiographically or sonographi-
placebo tablets were custom made for this study, were outwardly cally in major arteries and lower limbs. Follow-up and the assigned
indistinguishable, and contained 6 other water-soluble vitamins in treatment were continued in all participants who experienced a
similar concentrations, usually in amounts similar to the recom- secondary outcome event.
mended daily allowance for adults. The exact vitamin content is
given elsewhere.18 The placebo contained vitamins to prevent Statistical Analysis
vitamin deficiencies in patients assigned to the placebo group on The required number of patients was calculated as 350 per treatment
the one hand and to avoid an influence on homocysteine levels on group on the basis of an annual mortality of 16% and a 30%
the other hand. This approach had been tested in a clinical study mortality reduction as a result of the active treatment. A 1-year
with different multivitamin tablets.19 The amounts of vitamins in recruitment phase and a study period of 3 years were assumed, with
the tablets were controlled annually by an independent pharma- an annual dropout rate of 10%, 2-sided type I error of 5% (adjusted
ceutical laboratory. for 1 interim analysis), and a type II error of 20%.21 Because of the
1434 Circulation March 30, 2010

Results
Compliance, Follow-Up, and Adverse Events
Between July 2002 and November 2006, a total of 650
patients were included in the trial from 33 dialysis centers in
Germany and were assigned at random to the 2 treatment
arms. The median duration of follow-up was 2.1 years (5th to
95th percentiles, 0.2 to 5.2 years). Of the 650 patients, 107
received a transplant, 75 withdrew from participation during
the trial, and 18 discontinued treatment because of a change
in dialysis center (Figure 1).23
No serious adverse events relating to the study intervention
were reported. Fifteen patients discontinued the treatment
because of intolerance of the vitamins. The disorders reported
included gastrointestinal discomfort, skin rashes, and head-
aches. Patients who did not continue the therapy until the end
of the study for the reasons mentioned above were included in
the final analysis with data censored at the time of the last
follow-up visit.
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

Baseline Characteristics
The clinical and demographic characteristics of the patients
are presented in Table 1. The baseline characteristics in the
placebo and active treatment groups were generally well
balanced. Significant differences were found only in the
percentage of patients with hypertension, including patients
Figure 1. Trial flow diagram according to Consolidated Stan- receiving antihypertensive therapy. There were no differences
dards of Reporting Trials (CONSORT) Statement 2001.23 in the total blood pressure values between the treatment
groups. With respect to age, sex, distribution, and prevalence
extended recruitment phase and a reduced number of participating of diabetes mellitus, the randomized patients were compara-
patients (n⫽650), the follow-up period was increased to 6 years. An ble to the entire dialysis population in Germany.24
interim analysis was carried out by the data and safety monitoring
board in January 2007 after completion of a 2-year follow-up of 75% Effect of Vitamin Supplementation on Plasma
of the original number of patients, and they gave no reason to end the Homocysteine and B Vitamin Levels
trial before schedule. The effects of vitamin supplementation on plasma concentra-
All analyses were performed according to the intention-to-treat
tions of homocysteine, folate, cobalamin, and PLP are pre-
principle with the exception that patients who left the study could not
be followed up and were considered censored at that time. As shown sented in Table 2. These measurements had been carried out
in Figure 1, the proportion of these patients was similar in both for efficacy control on biochemical parameters after 6 months
treatment arms. The baseline characteristics of the patients in the 2 in a subset of patients (n⫽97). In the active treatment group,
treatment arms are described in absolute terms and as percentages, the median change in homocysteine was ⫺10.4 ␮mol/L
by means with their SDs when the distribution was approximately (P⬍0.001), which corresponds to a 35% decrease in the
normal, or by medians and 5th to 95th percentiles if it was not and baseline level. The median values of folate, cobalamin, and
compared between the 2 arms through the use of the ␹2 test, t test,
and Mann-Whitney U test.
PLP all increased in the active treatment group: folate, 5-fold;
The effect of the study medication on the plasma levels of PLP, 2-fold; and cobalamin, ⬇30% (all P⬍0.001). The
vitamins and homocysteine after 6 months of treatment was checked placebo group had been receiving a low-dose preparation of
in 97 patients by nonparametric tests (Wilcoxon matched-pairs rank the same vitamins, and this was associated with an insignif-
tests and the Mann-Whitney U test). Survival curves were estimated icant increase in PLP (⬍5%) and in both folate and cobal-
by the Kaplan-Meier method, and event rates were compared by the amin by ⬇30% (P⫽0.05 for folate and P⬍0.001 for cobal-
log-rank test. Proportional Cox regression analyses were used to
amin). The median homocysteine concentration in the
calculate risk estimates adjusted for further risk factors.
In addition to the analysis of first cardiovascular events, we placebo arm was lowered by 1.8 ␮mol/L compared with
carried out an analysis of all cardiovascular events that occurred in baseline (P⫽0.07).
the course of the study. Cardiovascular events that occurred after the
first event were counted for this purpose. For this analysis, the Primary Outcome: Total Mortality
method suggested by Andersen and Gill22 was used. Of the 650 randomized patients, 194 died during the study
All reported P values are 2 sided. After adjustment for 1 interim period as a result of all causes, 102 (31%) in the active treatment
analysis according to the O’Brien-Fleming method at an information group, and 92 (28%) in the placebo group (Table 3 and Figure
rate of 75%, in the final analysis, values of Pⱕ0.044 were taken as
significant for the primary and secondary end points. All statistical
2A). The treatment therefore had no effect on total mortality
analyses were done with SPSS version 15.0.1.1 for Windows (SPSS (hazard ratio [HR], 1.13; 95% confidence interval [CI], 0.85 to
Inc, Chicago, Ill) and SAS version 9.2 for Windows (SAS Institute, 1.50; P⫽0.51). Adjustment for prespecified baseline covariates
Inc, Cary, NC; PHREG procedure). did not lead to any substantial change in results. An analysis for
Heinz et al B Vitamins and Mortality in Dialysis Patients 1435

Table 1. Baseline Characteristics of the Study Participants


Total Placebo Active Treatment
Characteristic (n⫽650) (n⫽323) (n⫽327)
Age, y 61⫾13 61⫾13 61⫾13
Male sex, n (%) 379 (58) 189 (59) 190 (58)
Body mass index, kg/m2 27⫾5 27⫾5 27⫾5
Diabetes mellitus, n (%) 262 (40) 123 (38) 139 (43)
HbA1c 5.7 (4.9–7.8) 5.7 (4.7–7.7) 5.8 (4.9–7.9)
Hypertension, n (%) 576 (89) 278 (86)* 298 (91)*
Systolic BP, mm Hg 135 (100–170) 130 (95–180) 136 (102–169)
Diastolic BP, mm Hg 80 (60–90) 80 (60–90) 80 (60–90)
History of CVD, n (%) 312 (48) 149 (46) 163 (50)
Smoking status, n (%) 619 307 312
Never 278 (45) 133 (43) 145 (46)
Former 237 (38) 122 (40) 115 (37)
Current 104 (17) 52 (17) 52 (17)
Dialysis-related parameters
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

Time since dialysis, mo 32 (5–166) 31 (5–152) 32 (5–171)


Time on dialysis, h/wk 13.5 (9.0–16.5) 13.5 (9.0–16.5) 12.5 (9.0–16.5)
Erythropoietin, n (%) 530 (82) 269 (83) 261 (80)
Prestudy vitamins, n (%) 193 (30) 96 (30) 97 (30)
Laboratory measurements
Homocysteine, ␮mol/L 29.0 (14.1–63.3) 28.2 (13.0–62.0) 30.0 (14.9–65.3)
Folate, nmol/L 14.1 (6.6–75.6) 13.8 (6.8–67.5) 14.3 (6.3–84.1)
RBC folate, nmol/L 1758 (707–7399) 1859 (706–7128) 1716 (704–7605)
Cobalamin, pmol/L 344 (154–932) 351 (167–908) 334 (148–994)
PLP, nmol/L 22.9 (6.6–192.8) 23.7 (7.3–187.2) 21.4 (5.5–197.4)
Creatinine, ␮mol/L 809⫾273 795⫾290 823⫾256
Albumin, g/L 38.8 (33.1–43.6) 38.6 (33.1–44.0) 39.0 (33.1–43.4)
CRP, mg/L 5.9 (0.7–42.7) 5.7 (0.6–40.0) 6.2 (0.6–49.3)
CRP ⬎5 mg/L, n (%) 355 (55) 171 (53) 184 (56)
Total cholesterol, mmol/L 4.9 (3.2–7.2) 4.9 (3.1–6.9) 4.9 (3.3–7.3)
HDL cholesterol, mmol/L 0.9 (0.5–1.5) 0.9 (0.5–1.5) 0.9 (0.5–1.6)
LDL cholesterol, mmol/L 2.9 (1.5–4.8) 2.8 (1.4–4.8) 2.9 (1.6–4.8)
Triglycerides, mmol/L 2.3 (0.9–6.1) 2.3 (0.9–5.9) 2.3 (0.9–6.7)
HbA1c indicates hemoglobin A1c; BP, blood pressure; CVD, cardiovascular disease; RBC, red blood cell; PLP,
pyridoxal-5-phosphate; CRP, C-reactive protein; HDL, high-density lipoprotein; and LDL, low-density lipoprotein. Data are
given as mean⫾SD, medians (5th to 95th percentiles), or numbers and percentages as appropriate.
*P⫽0.04 for the comparison with placebo.

specific causes of mortality did not bring to light any significant Additional Analysis: Multiple
effects for particular causes (Table 3). Cardiovascular Events
The total number of cardiovascular events was 234 (181 first
Secondary Outcome: First Fatal and Nonfatal events and 53 subsequent events). The rate per 100 patient-
Cardiovascular Events years was 13.7 in the active treatment group and 17.1 in the
The secondary outcome occurred in 83 patients (25%) placebo group. The HR adjusted for multiple covariates was
in the active treatment and in 98 (30%) in the placebo 0.79 (95% CI, 0.59 to 1.05; P⫽0.10; data not shown).
group. The treatment had no significant effect on fatal and
nonfatal cardiovascular events (HR, 0.80; 95% CI, 0.60 to
1.07; P⫽0.13). After adjustment for the baseline covari- Discussion
ates, the results remained unaltered (Table 3 and Figure The association of high homocysteine levels with the risk of
2B). The numbers of events and HRs specified for indi- mortality and cardiovascular disease is an attractive explana-
vidual events are listed in Table 3. Only symptoms of tion for the elevated risk in hemodialysis patients because
unstable angina pectoris were significantly reduced by the almost every patient exhibits elevated homocysteine levels.
active treatment. Patients with ESRD show the highest homocysteine concen-
1436 Circulation March 30, 2010

Table 2. Plasma Levels of Total Homocysteine, Serum Levels of Folate, Serum Levels of Cobalamin, and Plasma
Levels of PLP at Baseline and After 6 Months in a Subgroup of 97 Patients
Baseline At 6 mo P, Baseline vs 6 mo* Changes During 6 mo
Total homocysteine, ␮mol/L
Placebo (n⫽37) 28.8 (14.1–68.2) 22.3 (9.8–54.1) 0.07 ⫺1.8 (⫺42.3–15.05)
Active treatment (n⫽59) 28.7 (16.5–69.4) 18.8 (7.2–33.6) ⬍0.001 ⫺10.4 (⫺35.8–2.5)
P (placebo vs treatment)† 0.49 0.03 0.001
Folate, nmol/L
Placebo (n⫽37) 11.8 (5.7–61.4) 15.0 (8.2–83.6) 0.05 3.0 (⫺22.9–16.4)
Active treatment (n⫽54) 12.7 (5.7–118.5) 81.8 (34.0–117.4) ⬍0.001 66.4 (⫺2.0–105.8)
P (placebo vs treatment)† 0.35 ⬍0.001 ⬍0.001
Cobalamin, pmol/L
Placebo (n⫽38) 288 (140–690) 399 (227–731) ⬍0.001 125 (⫺158–372)
Active treatment (n⫽58) 279 (72–999) 407 (163–1058) ⬍0.001 100 (⫺225–459)
P (placebo vs treatment)† 0.45 0.87 0.36
PLP, nmol/L
Placebo (n⫽38) 20.6 (9.9–135.5) 22.1 (8.0–284.0) 0.53 0.4 (⫺58.0–218.7)
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

Active treatment (n⫽57) 26.0 (8.8–333.6) 80.5 (14.1–305.7) ⬍0.001 58.4 (⫺238.9–259.3)
P (placebo vs treatment)† 0.35 ⬍0.001 ⬍0.001
Data are given as medians (5th to 95th percentiles).
*Wilcoxon matched-pairs rank test for repeated measurements.
†Mann-Whitney U test.

trations compared with any other patient group except those homocysteine can be lowered by supplementation with folic
with homocystinuria.14 Numerous studies have shown that in acid, vitamin B12, and vitamin B6. It was therefore reasonable
these patients there is an association between homocysteine to embark on a randomized controlled trial with the aim of
on the one hand and cardiovascular events and mortality on reducing homocysteine by vitamin supplementation and thus
the other hand (see the summary by Heinz et al15), and that also reducing cardiovascular risk and mortality.

Table 3. Primary and Secondary Outcomes in the Treatment Groups, Respective Causes of Mortality, Individual Cardiovascular End
Points, and the Corresponding HRs With 95% CIs
Active Treatment Placebo Crude HR Adjusted* HR
(n⫽327), n (%) (n⫽323), n (%) (95% CI) P (95% CI) P
Primary outcome
Total mortality 102 (31) 92 (28) 1.13 (0.85–1.50) 0.51 1.14 (0.85–1.52) 0.37
Secondary outcome
Cardiovascular events 83 (25) 98 (30) 0.80 (0.60–1.07) 0.13 0.79 (0.59–1.07) 0.13
Category of causes of mortality
Cardiac causes 37 (11) 32 (10) 1.18 (0.73–1.89) 0.50 1.26 (0.77–2.06) 0.36
Vascular causes 6 (2) 10 (3) 0.61 (0.22–1.68) 0.34 0.51 (0.18–1.42) 0.20
Sepsis and infections 28 (9) 22 (7) 1.30 (0.74–2.27) 0.36 1.19 (0.67–2.10) 0.55
Tumors 8 (2) 7 (2) 1.17 (0.43–3.23) 0.76 1.44 (0.50–4.17) 0.50
Other causes 14 (4) 15 (5) 0.95 (0.46–1.97) 0.89 1.00 (0.48–2.11) 0.99
Unknown 9 (3) 6 (2) 1.56 (0.56–4.39) 0.40 1.74 (0.61–4.98) 0.31
Individual cardiovascular events
Myocardial infarction (nonfatal and fatal) 20 (6) 19 (6) 1.06 (0.57–1.99) 0.86 1.00 (0.53–1.88) 0.99
Coronary vascularization procedures 8 (2) 18 (6) 0.44 (0.19–1.02) 0.06 0.44 (0.19–1.03) 0.06
Unstable angina pectoris 5 (2) 15 (5) 0.32 (0.12–0.89) 0.03 0.32 (0.12–0.89) 0.03
Sudden cardiac death 22 (7) 24 (7) 0.93 (0.52–1.66) 0.81 1.04 (0.57–1.91) 0.89
Stroke (nonfatal and fatal) 11 (3) 15 (5) 0.74 (0.34–1.62) 0.45 0.73 (0.33–1.60) 0.43
Peripheral artery disease 26 (8) 34 (11) 0.74 (0.45–1.24) 0.26 0.77 (0.46–1.31) 0.34
Pulmonary embolism and thromboses 7 (2) 6 (2) 1.16 (0.39–3.44) 0.79 1.14 (0.37–3.48) 0.82
*Cox regression analysis adjusted for age, sex, diabetes mellitus, hypertension, time on dialysis, C-reactive protein, and albumin.
Heinz et al B Vitamins and Mortality in Dialysis Patients 1437

infarction and 0.90 (95% CI, 0.58 to 1.40) for stroke in 2056
patients with ESRD or chronic kidney disease in HOST. In a
recent meta-analysis, we calculated for a 5-␮mol/L increase
in homocysteine an increase in risk of 9% for cardiovascular
events in patients with ESRD.15 If we apply this result to the
homocysteine levels obtained at 6 months in our study (Table
2), then a small, insignificant reduction in cardiovascular
disease events is plausible.
Concerning mortality, the results of our study are compa-
rable to the results of HOST. We did not observe any effect
on specific causes of death. Regarding cardiovascular events,
analysis of specific events showed a significant reduction in
unstable angina pectoris and fewer vascularization proce-
dures (Table 3). Because this is a post hoc analysis, the results
must be considered carefully.
The results of the present study and of other controlled
randomized trials in patients with (end-stage) renal disease
suggest that B vitamins do not have an effect on total
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

mortality in this population.25,26 However, there is a consis-


tent between-trial reduction in relative risk of cardiovascular
disease, on the order of 10%. The trials conducted so far,
including ours, have been too small to establish a significant
effect of this magnitude. Doing so would require a trial with
⬎2700 patients in each treatment group. It is not likely that an
investigation on this scale will ever be conducted to prove
this statement. The traditional approaches that have been
successful in cardiovascular disease risk reduction in patients
without renal disease are, however, ineffective in patients
with ESRD.27
A major criticism of vitamin supplementation trials in
Figure 2. Primary end point of total mortality (A) and secondary patients without renal disease is that homocysteine is normal
end point of fatal and nonfatal cardiovascular events (B). or nearly normal in these patients. The median baseline
Kaplan-Meier estimates of survival in the active treatment group homocysteine level in our study was 28 ␮mol/L, about twice
vs placebo group. Solid line indicates active treatment; dashed
line, placebo. the value of the homocysteine levels in these trials. Whether
a similar effect would be observed in patients without renal
The results of our study, however, do not support the idea disease but with homocysteine levels of this magnitude
that vitamin supplementation with folic acid, vitamin B12, and remains unknown.
vitamin B6 can generally reduce cardiovascular events and
mortality in patients with ESRD. Although this result is rather Conclusions
disappointing at first glance, some important points must be Active treatment with folic acid, vitamin B12, and vitamin B6
noted. First, the patients in the placebo group received a did not significantly reduce total mortality and cardiovascular
low-dose vitamin supplement with the aim of avoiding risk in patients with ESRD. Our findings do not support the
vitamin deficiencies in this group, which would then have administration of high-dose vitamin supplements in this total
been treated by the physicians in charge. Preliminary studies population. However, in accordance with other studies with B
had suggested that this low-dose supplement would not have vitamins in patients with chronic kidney failure, a smaller
any influence on the homocysteine levels and vitamin lev- protective effect of the vitamins on cardiovascular events is
els.19 However, we did not observe a significant difference in possible.
the cobalamin levels between the treatment groups.
Second, we could have expected this result if we had Acknowledgments
known the results from other trials that have been published We are indebted to all participating dialysis patients, physicians, and
since the beginning of our study in 2002. It should be dialysis staff. None of these people received any compensation for
their contribution.
mentioned that the results of the present trial are in line with the
results of other trials in this population group, the Atherosclero-
Sources of Funding
sis and Folic Acid Supplementation Trial (ASFAST) and the
This work was funded by the School of Medicine of Otto-von-Guericke
Homocysteine Trial (HOST).25,26 The HRs for cardiovascular University Magdeburg; Roche Diagnostics, Mannheim, Germany (lab-
events were 0.87 (95% CI, 0.58 to 1.32) in 315 patients in the oratory measurements); and Fresenius Medical Care, Bad Homburg,
ASFAST trial and 0.86 (95% CI, 0.67 to 1.08) for myocardial Germany (study drug and matching placebo).
1438 Circulation March 30, 2010

Disclosures 14. McCully KS. Homocysteine and vascular disease. Nat Med. 1996;2:
386 –389.
None.
15. Heinz J, Kropf S, Luley C, Dierkes J. Homocysteine as a risk factor for
cardiovascular disease in patients treated by dialysis: a meta-analysis.
References Am J Kidney Dis. 2009;54:478 – 489.
1. Vollset SE, Refsum H, Tverdal A, Nygård O, Nordrehaug JE, Tell GS, 16. Sunder-Plassmann G, Födinger M, Buchmayer H, Papagiannopoulos M,
Ueland PM. Plasma total homocysteine and cardiovascular and noncar- Wojcik J, Kletzmayr J, Enzenberger B, Janata O, Winkelmayer WC, Paul
diovascular mortality: the Hordaland Homocysteine Study. Am J Clin G, Auinger M, Barnas U, Hörl WH. Effect of high dose folic acid therapy
Nutr. 2001;74:130 –136. on hyperhomocysteinemia in hemodialysis patients: results of the Vienna
2. Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A quantitative multicenter study. J Am Soc Nephrol. 2000;11:1106 –1116.
assessment of plasma homocysteine as a risk factor for vascular disease. 17. Dierkes J, Domröse U, Ambrosch A, Bosselmann HP, Neumann KH,
JAMA. 1995;274:1049 –1057. Luley C. Response of hyperhomocysteinemia to folic acid supplemen-
3. Homocysteine Studies Collaboration. Homocysteine and risk of ischemic tation in patients with end-stage renal disease. Clin Nephrol. 1999;51:
heart disease and stroke: a meta-analysis. JAMA. 2002;288:2015–2022. 108 –115.
4. Baker F, Picton D, Blackwood S, Hunt J, Erskine M, Dyas M. Blinded 18. Heinz J, Domröse U, Luley C, Westphal S, Kropf S, Neumann KH,
comparison of folic acid and placebo in patients with ischemic heart Dierkes J. Influence of a supplementation with vitamins on cardiovascular
disease: an outcome trial. Circulation. 2002;106:3642. Abstract. morbidity and mortality in patients with end-stage renal disease: design
5. Toole JF, Malinow MR, Chambless LE, Spence JD, Pettigrew LC, and baseline data of a randomized clinical trial. Clin Nephrol. 2009;71:
Howard VJ, Sides EG, Wang CH, Stampfer M. Lowering homocysteine 363–365.
in patients with ischemic stroke to prevent recurrent stroke, myocardial 19. Dierkes J, Domröse U, Bosselmann KP, Neumann KH, Luley C. Homo-
infarction, and death: the Vitamin Intervention for Stroke Prevention cysteine lowering effect of different multivitamin preparations in patients
(VISP) randomized controlled trial. JAMA. 2004;291:565–575. with end-stage renal disease. J Ren Nutr. 2001;11:67–72.
20. Vester B, Rasmussen K. High performance liquid chromatography
6. Bønaa KH, Njølstad I, Ueland PM, Schirmer H, Tverdal A, Steigen T,
method for rapid and accurate determination of homocysteine in plasma
Wang H, Nordrehaug JE, Arnesen E, Rasmussen K, for the NORVIT
Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

and serum. Eur J Clin Chem Clin Biochem. 1991;29:549 –554.


Trial Investigators. Homocysteine lowering and cardiovascular events
21. O’Brien PC, Fleming TR. A multiple testing procedure for clinical trials.
after acute myocardial infarction. N Engl J Med. 2006;354:1578 –1588.
Biometrics. 1979;35:549 –556.
7. Ebbing M, Bleie Ø, Ueland PM, Nordrehaug JE, Nilsen DW, Vollset SE,
22. Andersen PK, Gill RD. Cox’s regression model counting process: a large
Refsum H, Pedersen EK, Nygård O. Mortality and cardiovascular events
sample study. Ann Stat. 1982;10:1100 –1120.
in patients treated with homocysteine-lowering B vitamins after coronary
23. Moher D, Schulz KF, Altman DG. The CONSORT statement: revised
angiography: a randomized controlled trial. JAMA. 2008;300:795– 804. recommendations for improving the quality of reports of parallel-group
8. Lonn E, Yusuf S, Arnold MJ, Sheridan P, Pogue J, Micks M, McQueen randomised trials. Lancet. 2001;357:1191–1194.
MJ, Probstfield J, Fodor G, Held C, Genest J Jr, for the Heart Outcomes 24. Frei U, Schober-Halstenberg. Nierenersatztherapie in Deutschland:
Prevention Evaluation (HOPE) 2 Investigators. Homocysteine lowering Bericht über Dialysebehandlung und Nierentransplantation in Deut-
with folic acid and B vitamins in vascular disease. N Engl J Med. schland 2006/2007. QuaSi-Niere gGmbH 2008.
2006;354:1567–1577. 25. Zoungas S, McGrath BP, Branley P, Kerr PG, Muske C, Wolfe R,
9. Foley RN, Parfrey PS, Sarnak MJ. Epidemiology of cardiovascular Atkins RC, Nicholls K, Fraenkel M, Hutchison BG, Walker R, McNeil
disease in chronic renal disease. J Am Soc Nephrol. 1998;9:S16 –S23. JJ. Cardiovascular morbidity and mortality in the Atherosclerosis and
10. Ikizler TA. Epidemiology of vascular disease in renal failure. Blood Purif. Folic Acid Supplementation Trial (ASFAST) in chronic renal failure:
2002;20:6 –10. a multicenter, randomized, controlled trial. J Am Coll Cardiol. 2006;
11. Kundhal K, Lok CE. Clinical epidemiology of cardiovascular disease in 47:1108 –1116.
chronic kidney disease. Nephron Clin Pract. 2005;101:c47– c52. 26. Jamison RL, Hartigan P, Kaufman JS, Goldfarb DS, Warren SR, Guarino
12. Dierkes J, Domröse U, Westphal S, Ambrosch A, Bosselmann HP, PD, Gaziano JM, for the Veterans Affairs Site Investigators. Effect of
Neumann KH, Luley C. Cardiac troponin T predicts mortality in patients homocysteine lowering on mortality and vascular disease in advanced
with end-stage renal disease. Circulation. 2000;102:1964 –1969. chronic kidney disease and end-stage renal disease: a randomized con-
13. Bostom AG, Shemin D, Lapane KL, Miller JW, Sutherland P, Nadeau M, trolled trial. JAMA. 2007;298:1163–1170.
Seyoum E, Hartman W, Prior R, Wilson PW, Selhub J. Hyperhomocys- 27. Wanner C, Krane V, März W, Olschewski M, Mann JF, Ruf G, Ritz E,
teinemia and traditional cardiovascular disease risk factors in end-stage for the German Diabetes and Dialysis Study Investigators. Atorvastatin in
renal disease patients on dialysis: a case-control study. Atherosclerosis. patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl
1995;114:93–103. J Med. 2005;353:238 –248.

CLINICAL PERSPECTIVE
We studied the effect of homocysteine-lowering treatment with B vitamins in patients with end-stage renal disease in a
randomized controlled trial. After each dialysis session, patients received either a high-dose vitamin supplement (active
treatment: 5 mg folic acid, 50 ␮g vitamin B12, and 20 mg vitamin B6) or a low-dose vitamin supplement to avoid vitamin
deficiencies (control: 0.2 mg folic acid, 4 ␮g vitamin B12, and 1 mg vitamin B6). Homocysteine concentrations were
significantly lowered in the group receiving high amounts of vitamins. Patients were followed up for a median period of
25 months. The vitamin treatment did not affect total mortality (hazard ratio, 1.13; 95% confidence interval, 0.85 to 1.50;
P⫽0.51) or cardiovascular morbidity (hazard ratio, 0.80; 95% confidence interval, 0.60 to 1.07; P⫽0.13). Adjustment for
other risk factors did not change these results substantially. In this randomized clinical trial, a significant clinical benefit
of additional amounts of B vitamins for lowering of homocysteine concentrations was not shown. The results are in
accordance with other trials that used B vitamins in patients with renal disease.
B Vitamins and the Risk of Total Mortality and Cardiovascular Disease in End-Stage
Renal Disease: Results of a Randomized Controlled Trial
Judith Heinz, Siegfried Kropf, Ute Domröse, Sabine Westphal, Katrin Borucki, Claus Luley,
Klaus H. Neumann and Jutta Dierkes

Circulation. 2010;121:1432-1438; originally published online March 15, 2010;


Downloaded from http://circ.ahajournals.org/ by guest on June 6, 2018

doi: 10.1161/CIRCULATIONAHA.109.904672
Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2010 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/121/12/1432

Data Supplement (unedited) at:


http://circ.ahajournals.org/content/suppl/2013/10/17/CIRCULATIONAHA.109.904672.DC1

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


http://circ.ahajournals.org//subscriptions/
Page 72

Résumés d’articles

Antécédents parentaux d’accident vasculaire cérébral


et risque d’AVC encouru par les enfants
L’Etude de Framingham
Sudha Seshadri, MD ; Alexa Beiser, PhD ; Aleksandra Pikula, MD ; Jayandra J. Himali, MSc ;
Margaret Kelly-Hayes, DEd, RN ; Stephanie Debette, MD, PhD ; Anita L. DeStefano, PhD ;
Jose R. Romero, MD ; Carlos S. Kase, MD ; Philip A. Wolf, MD

Contexte—Les données sur le lien existant entre les antécédents parentaux d’accident vasculaire cérébral (AVC) et le risque de
survenue d’un tel événement chez les enfants sont étonnamment divergentes, cela tenant en grande partie à l’hétérogénéité des
protocoles d’études et à l’absence d’informations validées, contrairement au statut des ascendants en matière d’AVC pour lequel
on dispose de données historiques.
Méthodes et résultats—Nous avons mené une étude en milieu communautaire pour savoir si la survenue d’un AVC chez un parent
(documentée sur un mode prospectif) avait eu ou non pour effet d’accroître le risque encouru par ses enfants de présenter un tel
accident ; pour ce faire, parmi les participants à l’étude de Framingham sur la descendance, nous avons constitué une cohorte de
3 443 individus initialement sans antécédent d’AVC (53 % de femmes ; âge moyen : 48 ± 14 ans) mais dont un parent avait été
victime d’un tel événement (avant l’âge de 65 ans) et qui s’étaient prêtés aux premier, troisième, cinquième et/ou septième
examens prévus, ces sujets ayant été suivis pendant 8 ans après le bilan initial. Sur plus de 11 029 périodes d’observation
individualisée (soit 77 534 années-patients), nous avons recensé 106 AVC parentaux survenus avant l’âge de 65 ans et 128 ayant
frappé la descendance (respectivement 74 et 106 de ces accidents ayant été de type ischémique). Par l’emploi de modèles de Cox
multivariés ajustés pour l’âge, le sexe, la place dans la fratrie et les facteurs de risque d’AVC initialement présents, nous avons
établi que la survenue, chez un ascendant, d’un quelconque type d’AVC et, notamment, d’un accident d’origine ischémique avait
concouru à augmenter le risque de survenue du même type d’AVC chez les enfants (rapport de risques : 2,79 ; intervalle de
confiance [IC] à 95 % : 1,68 à 4,66 ; p <0,001 pour l’analyse portant sur les AVC de tous types ; rapport de risques : 3,15 ; IC à
95 % : 1,69 à 5,88 ; p <0,001 pour l’analyse portant sur les AVC de type ischémique). La corrélation s’est montrée valide, que
l’antécédent d’AVC ait été enregistré chez la mère ou chez le père.
Conclusions—La survenue documentée d’un AVC chez l’un des parents avant l’âge de 65 ans a concouru à tripler le risque
d’accident du même type chez les enfants. Cette augmentation du risque a persisté après ajustement tenant compte des facteurs
conventionnels de risque d’AVC. Les antécédents parentaux validés d’AVC pourraient dès lors se révéler cliniquement utiles
pour évaluer le degré d’exposition d’un individu donné au risque d’AVC. (Traduit de l’anglais : Parental Occurrence of Stroke
and Risk of Stroke in Their Children The Framingham Study. Circulation. 2010;121:1304–1312.)
Mots clés : épidémiologie 䊏 hérédité 䊏 ischémie 䊏 accident vasculaire cérébral

Influence de la supplémentation en vitamines B sur la mortalité


globale et le risque d’événements cardiovasculaires dans
l’insuffisance rénale terminale
Résultats d’un essai randomisé et contrôlé
Judith Heinz, MSc ; Siegfried Kropf, PhD ; Ute Domröse, MD ; Sabine Westphal, MD ;
Katrin Borucki, MD ; Claus Luley, MD ; Klaus H. Neumann, MD ; Jutta Dierkes, PhD

Contexte—Des études observationnelles ont montré que l’hyperhomocystéinémie contribue à augmenter la mortalité globale et le
risque d’événements cardiovasculaires chez les patients atteints d’insuffisance rénale terminale. Ces individus présentent des
taux d’homocystéine extrêmement élevés, qu’il est possible de réduire par une supplémentation en acide folique et en vitamine
B12. Nous avons donc entrepris un essai clinique randomisé visant à abaisser l’homocystéinémie par l’apport de vitamines B afin
de vérifier si cela avait pour effet de diminuer l’incidence des événements cardiovasculaires et la mortalité globale.

© 2011 Lippincott Williams & Wilkins Circulation est disponible sur http://circ.ahajournals.org

72

16:33:15:11:10 Page 72
Page 73

Résumés d’articles 73

Méthodes et résultats—Cette étude multicentrique randomisée et en double aveugle a été menée dans 33 centres d’hémodialyse
d’Allemagne du Nord et de l’Est entre juillet 2002 et juillet 2008. Nous avons randomisé 650 patients hémodialysés au stade
d’insuffisance rénale terminale de manière à constituer deux groupes de traitement qui ont respectivement reçu, 3 fois par
semaine pendant une durée moyenne de 2 ans, 5 mg d’acide folique, 50 µg de vitamine B12 et 20 mg de vitamine B6 (groupe de
traitement actif) ou 0,2 mg d’acide folique, 4 µg de vitamine B12 et 1,0 mg de vitamine B6 (groupe placebo). Le critère de
jugement principal a été la mortalité globale, le critère secondaire ayant été le taux d’événements cardiovasculaires fatals et non
fatals. L’événement cible principal est survenu chez 102 (31 %) des patients qui avaient reçu le traitement actif et chez 92 (28 %)
de ceux auxquels le placebo avait été assigné (rapport de risques : 1,13 ; intervalle de confiance [IC] à 95 % : 0,85 à 1,50 ;
p = 0,51). L’événement cible secondaire s’est produit chez 83 (25 %) des patients du groupe de traitement actif et chez 98 (30 %)
des sujets du groupe placebo (rapport de risques : 0,80 ; IC à 95 % : 0,60 à 1,07 ; p = 0,13).
Conclusions—L’augmentation des apports en acide folique et en vitamines B12 et B6 n’a pas diminué la mortalité globale et n’a pas
eu d’influence significative sur le risque d’événements cardiovasculaires chez les patients atteints d’insuffisance rénale terminale.
Registre des Essais cliniques—URL : www.anzctr.org.au. Identifiant unique : ACTRN12609000911291. URL : www.cochrane-
renal.org. Identifiant unique : CRG010600027.
(Traduit de l’anglais : B Vitamins and the Risk of Total Mortality and Cardiovascular Disease in End-Stage Renal Disease Results
of a Randomized Controlled Trial. Circulation. 2010;121:1432–1438.)
Mots clés : vitamines B 䊏 maladies cardiovasculaires 䊏 hémodialyse 䊏
homocystéine 䊏 rein 䊏 mortalité 䊏 prévention

Incidence, facteurs prédictifs et implications pronostiques


de l’hospitalisation pour saignement différé après
intervention coronaire percutanée chez les
patients âgés de plus de 65 ans

Dennis T. Ko, MD, MSc ; Lingsong Yun, MSc ; Harindra C. Wijeysundera, MD ;


Cynthia A. Jackevicius, PharmD, MSc ; Sunil V. Rao, MD ; Peter C. Austin, PhD ;
Jean-François Marquis, MD ; Jack V. Tu, MD, PhD

Contexte—Les données recueillies sur les complications hémorragiques des interventions coronaires percutanées (ICP) émanent
essentiellement d’essais randomisés qui étaient axés sur les saignements survenus pendant l’hospitalisation. De ce fait, on ne
connaît pas avec précision l’incidence des hémorragies postopératoires différées, les facteurs indépendants qui les favorisent ni
leur importance pronostique en pratique clinique.
Méthodes et résultats—Notre étude a porté sur 22 798 patients âgés de plus de 65 ans qui avaient fait l’objet d’une ICP entre le 1er
décembre 2003 et le 31 mars 2007 en Ontario (Canada). Nous avons eu recours à des modèles de risques proportionnels de Cox
pour établir les facteurs corrélés avec la survenue d’un saignement différé (défini comme ayant motivé l’hospitalisation du
patient postérieurement à celle au cours de laquelle l’ICP avait été pratiquée) et estimer le risque de décès ou d’infarctus du
myocarde associé à un tel saignement. Nous avons observé que 2,5 % des patients avaient été hospitalisés pour un événement
hémorragique dans l’année ayant suivi l’ICP, 56 % des épisodes ayant eu pour siège la sphère digestive. Le facteur ayant le plus
significativement majoré le risque de saignement différé a été l’administration postopératoire de warfarine (rapport de risques
[RR] : 3,12). Les autres facteurs de risque significatifs ont été l’âge (RR : 1,41 par tranche de 10 ans), le sexe masculin
(RR : 1,24), le cancer (RR : 1,80), les antécédents hémorragiques (RR : 2,42), l’insuffisance rénale chronique (RR : 1,93)
et la prise d’anti-inflammatoires non stéroïdiens (RR : 1,73). Après ajustement en fonction des covariables initiales, il est
apparu que l’hospitalisation pour épisode hémorragique avait concouru à augmenter significativement le risque de décès
ou d’infarctus du myocarde à un an (RR : 2,39 ; IC à 95 % : 1,93 à 2,97) ainsi que la mortalité globale (RR : 3,38 ;
IC à 95 % : 2,60 à 4,40).
Conclusions—L’hospitalisation motivée par la survenue d’un saignement différé après ICP contribue à accroître fortement le
risque de décès ou d’infarctus du myocarde. C’est la prescription d’une trithérapie anticoagulante (aspirine, thiénopyridine
et warfarine) qui est à l’origine du risque d’événement hémorragique différé le plus élevé. (Traduit de l’anglais : Incidence,
Predictors, and Prognostic Implications of Hospitalization for Late Bleeding After Percutaneous Coronary Intervention for
Patients Older Than 65 Years. Circ Cardiovase Interv. 2010;3:140–147.)
Mots clés : saignement 䊏 intervention coronaire percutanée 䊏 mortalité 䊏 anticoagulants oraux

16:33:15:11:10 Page 73

Вам также может понравиться