Raloxifene and Cardiovascular Events in Osteoporotic Postmenopausal Women

Four-Year Results From the MORE (Multiple Outcomes of Raloxifene Evaluation)
Randomized Trial

JAMA. 2002;287:847-857

Author Information <http://jama.ama-assn.org/issues/v287n7/rfull/#aainfo>
Elizabeth Barrett-Connor, MD; Deborah Grady, MD; Andreas Sashegyi, PhD;
Pamela W. Anderson, MD; David A. Cox, PhD; Krzysztof Hoszowski, MD; Pentti
Rautaharju, MD; Kristine D. Harper, MD; for the MORE Investigators
Context  Raloxifene, a selective estrogen receptor modulator, improves
cardiovascular risk factors, but its effect on cardiovascular events is
unknown.
Objective  To determine the effect of raloxifene on cardiovascular events in
osteoporotic postmenopausal women.
Design  Secondary analysis of data from the Multiple Outcomes of Raloxifene
Evaluation trial, a randomized, double-blind, placebo-controlled trial
conducted between November 1994 and September 1999.
Setting  Outpatient and community settings at 180 sites in 25 countries.
Participants  A total of 7705 osteoporotic postmenopausal women (mean age,
67 years).
Intervention  Patients were randomly assigned to receive raloxifene, 60 mg/d
(n = 2557), or 120 mg/d (n = 2572), or placebo (n = 2576) for 4 years.
Main Outcome Measures  Cardiovascular events, including coronary events
(myocardial infarction, unstable angina, or coronary ischemia) and
cerebrovascular events (stroke or transient ischemic attack), collected as
safety end points and subsequently adjudicated by a cardiologist blinded to
therapy. Cardiovascular risk at study entry was determined by the presence
of multiple cardiovascular risk factors or prior coronary events or
revascularization procedure.
Results  In the overall cohort, there were no significant differences
between treatment groups in the number of combined coronary and
cerebrovascular events: 96 (3.7%) with placebo, 82 (3.2%) with 60 mg/d of
raloxifene, and 94 (3.7%) with 120 mg/d of raloxifene. Relative risks (RRs)
were 0.86 (95% confidence interval [CI], 0.64-1.15) and 0.98 (95% CI,
0.74-1.30) for 60 mg/d and 120 mg/d of raloxifene, respectively. Similar
results were obtained when coronary and cerebrovascular events were analyzed
separately. Among the subset of 1035 women with increased cardiovascular
risk at baseline, those assigned to raloxifene had a significantly lower
risk of cardiovascular events compared with placebo (RR, 0.60; 95% CI,
0.38-0.95 for both raloxifene groups). The number of cardiovascular events
during the first year was not significantly different across groups in the
overall cohort (P = .94), or among women at increased cardiovascular risk (P
= .86) or with evidence of established coronary heart disease (P = .60).
Conclusions  Raloxifene therapy for 4 years did not significantly affect the
risk of cardiovascular events in the overall cohort but did significantly
reduce the risk of cardiovascular events in the subset of women with
increased cardiovascular risk. There was no evidence that raloxifene caused
an early increase in risk of cardiovascular events. Before raloxifene is
used for prevention of cardiovascular events, these findings require
confirmation in trials with evaluation of cardiovascular outcomes as the
primary objective.
JAMA. 2002;287:847-857
JOC11015
Coronary heart disease (CHD) is the leading cause of death among
postmenopausal women in the United States and most of the Western world. 1
<http://jama.ama-assn.org/issues/v287n7/rfull/#r1>  In observational
studies, postmenopausal women who take hormone replacement therapy (HRT),
alone or with a progestin, have approximately 30% less risk of CHD compared
with untreated postmenopausal women. 2
<http://jama.ama-assn.org/issues/v287n7/rfull/#r2>  Randomized clinical
trials have not confirmed a beneficial effect of HRT on either the
progression of coronary atherosclerosis or the risk of cardiovascular (CV)
events in postmenopausal women with existing CHD. 3
<http://jama.ama-assn.org/issues/v287n7/rfull/#r3> , 4
<http://jama.ama-assn.org/issues/v287n7/rfull/#r4>  In the Heart and
Estrogen/progestin Replacement Study (HERS), 3
<http://jama.ama-assn.org/issues/v287n7/rfull/#r3>  and in preliminary
information from the Women's Health Initiative (WHI), 5
<http://jama.ama-assn.org/issues/v287n7/rfull/#r5> , 6
<http://jama.ama-assn.org/issues/v287n7/rfull/#r6>  women assigned to
receive HRT experienced an early increased risk of acute coronary events. In
the Women's Estrogen for Stroke Trial (WEST), 7
<http://jama.ama-assn.org/issues/v287n7/rfull/#r7>  women assigned to
estradiol had an increased risk of stroke compared with placebo during the
first 6 months of treatment. Pooled analyses of smaller, short-term trials 8
<http://jama.ama-assn.org/issues/v287n7/rfull/#r8> , 9
<http://jama.ama-assn.org/issues/v287n7/rfull/#r9>  and recent reports from
observational studies 10-12
<http://jama.ama-assn.org/issues/v287n7/rfull/#r10>  also suggest a
transient increase in risk of recurrent CV events after initiation of HRT in
postmenopausal women.
The Multiple Outcomes of Raloxifene Evaluation (MORE) trial was a double
blind, placebo-controlled trial designed to determine the effect of
raloxifene, a selective estrogen receptor modulator, on bone mineral density
and vertebral fractures in 7705 postmenopausal women with osteoporosis. 13
<http://jama.ama-assn.org/issues/v287n7/rfull/#r13> , 14
<http://jama.ama-assn.org/issues/v287n7/rfull/#r14>  Raloxifene reduced the
risk of osteoporotic vertebral fractures and newly diagnosed breast cancer
without increasing the risk of endometrial cancer but increased the risk of
venous thromboembolic events to an extent similar to that of HRT and
tamoxifen. 13-17 <http://jama.ama-assn.org/issues/v287n7/rfull/#r13>
Although raloxifene has favorable effects on low-density lipoprotein
cholesterol (LDL-C) and other risk factors for CHD 18
<http://jama.ama-assn.org/issues/v287n7/rfull/#r18> , 19
<http://jama.ama-assn.org/issues/v287n7/rfull/#r19>  and improves vascular
endothelial function in postmenopausal women, 20
<http://jama.ama-assn.org/issues/v287n7/rfull/#r20>  the effect of
raloxifene on the risk of arterial CV events is unknown. Whether raloxifene
affects the risk of CV events in postmenopausal women is currently being
tested prospectively in the Raloxifene Use for The Heart (RUTH) trial, which
enrolled 10 101 postmenopausal women with established CHD or with multiple
risk factors for acute CHD events; however, results will not be available
for several years. 21 <http://jama.ama-assn.org/issues/v287n7/rfull/#r21>
In the MORE trial, CV events and mortality were reported as adverse events
and serum lipid concentrations were measured. We used these data to
determine the early and overall effects of 4 years of raloxifene therapy on
the occurrence of major arterial CV events, including coronary and
cerebrovascular events. Since the MORE cohort was at relatively low risk for
CV events overall, we also determined the effect of raloxifene on coronary
and cerebrovascular events among women at increased risk for these events as
defined by the presence of multiple CV risk factors or evidence of
established CHD at baseline.



METHODS



Study Design and Participants

Study participants were enrolled in the MORE study at 180 sites in 25
countries. Details of the study design, eligibility criteria, and flow of
participants through the trial have been published. 13-15
<http://jama.ama-assn.org/issues/v287n7/rfull/#r13>  Patients were at least
2 years postmenopausal and had osteoporosis documented either by prior
vertebral fracture or bone mineral density T score of less than -2.5. Women
were excluded if they had a history of stroke or venous thromboembolic
disease during the past 10 years. Women with coronary artery disease,
hypertension, hypercholesterolemia, type 2 diabetes mellitus, or other risk
factors for CV disease were not excluded. The primary outcomes of MORE were
bone mineral density and vertebral fractures. Measurement of serum lipid
concentrations was a secondary objective of the trial, and assessment of
major CV events (including CV death) was a safety objective of the trial.
The protocol was approved by the ethical review board at each site. All
women gave written informed consent to participate in the study in
accordance with the ethical principles stated in the Declaration of
Helsinki.
Randomization and Blinding

Details of the randomization and blinding procedures have been published.
13-15 <http://jama.ama-assn.org/issues/v287n7/rfull/#r13>  Briefly, women
were stratified by site and randomly assigned to receive raloxifene 60 mg/d
or 120 mg/d or placebo ( Figure 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f1.html> ). The
sponsor supplied randomly numbered kits containing identically appearing
raloxifene and placebo tablets. The women received 2 tablets daily: 2
placebo, 1 placebo and 1 tablet of 60-mg raloxifene, or 2 tablets of 60-mg
raloxifene. In addition, all women were provided with 500 mg of calcium and
400 to 600 IU of cholecalciferol per day, beginning at study entry.
Participants, investigators, laboratory staff, and those who adjudicated the
CV outcomes were blinded to treatment assignment.
Baseline Assessments and Follow-up

At baseline, information on age, ethnicity, years of education, reproductive
history, prior postmenopausal hormone use, cigarette smoking, alcohol
consumption, and health conditions was reported ( Table 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t1.html> ). In
addition, women were asked whether they had previously experienced any of
the following CV events or procedures: myocardial infarction (MI), coronary
artery bypass graft (CABG) surgery, percutaneous coronary intervention, or
stroke. Other baseline clinical assessments included height, weight, blood
pressure, pulse, a standard 12-lead electrocardiogram (ECG), and measurement
of serum lipid concentrations (total cholesterol, high-density lipoprotein
cholesterol [HDL-C], and triglycerides) and fasting blood glucose. Use of
all prescription and over-the-counter medications was recorded at baseline.
Follow-up visits were scheduled at 3 and 6 months after enrollment and every
6 months thereafter.
CV Risk Assessment

We assessed CV risk for each woman by assigning a risk score based on
evidence of established CHD or the presence of CV risk factors, adapted from
criteria established previously for enrollment of women in the RUTH trial.
21 <http://jama.ama-assn.org/issues/v287n7/rfull/#r21>  Women with a prior
MI, percutaneous coronary intervention, or CABG surgery received a score of
4 points, regardless of the timing of the event or procedure relative to
enrollment. Women with diabetes mellitus (fasting blood glucose >140 mg/dL
[>7.8 mmol/L]) or taking hypoglycemic medication) received 3 points. Women
70 years or older received 2 points; those between age 65 and 70 years
received 1 point. Women who were current smokers (10 or more cigarettes per
day for 6 months before enrollment), had hypertension (systolic blood
pressure >160 mm Hg, diastolic blood pressure >95 mm Hg, or taking
antihypertensive medication), or had hyperlipidemia (LDL-C >160 mg/dL ([4.14
mmol/L] or HDL-C <45 mg/dL [1.17 mmol/L] with triglycerides >250 mg/dL [2.82
mmol/L], or receiving hypolipidemic therapy) received 1 point for each
condition. The combination of current smoking, hypertension, and
hyperlipidemia was scored 4 points. Women with 4 or more risk points were
considered to be at increased risk for CV events. 21
<http://jama.ama-assn.org/issues/v287n7/rfull/#r21>  Based on these
criteria, 1035 (13.4%) were defined as being at high risk for CV events, 202
with a prior acute coronary event or procedure and the remainder with no
prior history of acute coronary events but with multiple CV risk factors.
Ascertainment and Adjudication of CV Events

Cardiovascular events were collected by asking women at each visit if they
had an MI, CABG surgery, percutaneous coronary intervention, or a stroke
since the previous visit and by recording unsolicited reports of adverse CV
events. For each reported event, the investigator reviewed available
documentation, including ECG reports, cardiac enzyme results, procedure
reports, hospitalization records, discharge summaries, or letters from
physicians and provided a written summary of these findings to the sponsor.
All summaries of reported CV events through 4 years of follow-up were
reviewed and adjudicated by a board-certified cardiologist, who was
contracted by the sponsor, was not associated with the trial, and was
blinded to treatment assignment. No original source documents were available
to the adjudicator.
All reported deaths were adjudicated to be due to either CV or non-CV
causes. Cardiovascular deaths included coronary death (fatal MI, sudden
death, unwitnessed death in the absence of other likely noncoronary causes,
and death related to a coronary artery procedure) and cerebrovascular death
(fatal stroke). Nonfatal CV outcomes included coronary events (MI, unstable
angina, or coronary ischemia) and cerebrovascular events (stroke or
transient ischemic attack). Based on review of the case summaries, the
adjudicator determined that MI was present in women with symptoms compatible
with MI (ie, chest pain or anginal equivalent) who had documentation of
either elevated cardiac enzyme levels (ie, cardiac enzymes >2-fold above the
upper limit of normal or deemed consistent with MI by the local laboratory)
or positive ECG findings compatible with MI (ie, pathological Q waves, ST
depression or elevation or T-wave inversion); in hospitalized patients with
a discharge diagnosis of MI; or in patients who had an MI according to a
physician's statement in the medical record. Unstable angina was deemed to
be present in hospitalized patients with anginal symptoms (ie, chest pain or
anginal equivalent) with a primary diagnosis of this condition. A diagnosis
of coronary ischemia was made for women hospitalized with chest symptoms
consistent with angina but without additional supporting evidence of
unstable angina or other acute coronary syndromes.
Cerebrovascular events included stroke and transient ischemic attack. Stroke
was deemed to be present in patients with symptoms compatible with stroke
who had documentation of either positive computed tomographic scan result
(read by a physician as consistent with recent stroke) or with persistent
neurological deficit (>24 hours); in hospitalized patients with a discharge
diagnosis of stroke; or in patients who had a stroke according to a
physician's statement. Transient ischemic attack was deemed to have occurred
in hospitalized patients with symptoms compatible with stroke but for whom a
diagnosis of stroke was not established or for patients with a primary
diagnosis of transient ischemic attack.
Serial ECGs

A standard 12-lead ECG was obtained at baseline and at 24 and 48 months or
at early discontinuation. The ECG results were coded by the Epidemiological
Cardiology Research Center (EPICARE) at Wake Forest University, using the
NOVACODE serial classification system. 22
<http://jama.ama-assn.org/issues/v287n7/rfull/#r22>  Persons interpreting
the ECGs had no knowledge of treatment assignment, clinical symptoms, or
cardiac enzymes. Incident MI and myocardial ischemia based on ECG changes
were defined by changes in Q wave and ST-T wave scores. Incident MI required
an increase in Q wave score of at least 25 points (grade 2), or at least 15
points (grade 1) with clinically significant ST-T wave evolution from the
baseline ECG (change in ST-T score of at least 10). Myocardial ischemia was
defined as borderline isolated Q-wave evolution without evolving ST-T waves
or ischemic evolution of ST-T waves without a significant Q wave. 23
<http://jama.ama-assn.org/issues/v287n7/rfull/#r23>  Serial ECGs were
obtained independent of the reporting of clinical CV events and were not
used in the adjudication of the clinical CV events because of the lack of
temporal association between them. Cardiovascular events identified from
serial ECGs were not included in the analysis of clinically reported events.
Measurement of Serum Lipids

Total cholesterol, HDL-C, and triglyceride concentrations were measured in
all women at baseline. Follow-up measurements were performed in serum
obtained at 6, 12, 24, 36, and 48 months using previously described methods.
18 <http://jama.ama-assn.org/issues/v287n7/rfull/#r18>  Total cholesterol
was measured at follow-up visits in all women, whereas only samples from the
2738 women enrolled at the 14 study sites that had at least 100 enrolled
patients were assayed for HDL-C and triglyceride concentrations. Low-density
lipoprotein cholesterol levels were calculated using the Friedewald
equation. 24 <http://jama.ama-assn.org/issues/v287n7/rfull/#r24>
Statistical Analyses

Procedures in SAS Version 6.09 were used to perform all statistical analyses
(SAS Institute Inc, Cary, NC). Analyses were performed using the
intention-to-treat principle with patients allocated to assigned treatment
regardless of compliance or other postbaseline factors. Unless otherwise
stated, all hypotheses were tested at the .05 (2-sided) level of
significance. No adjustments were made for multiple comparisons.
Baseline patient characteristics were compared using analysis of variance
for continuous data and Pearson chi2 test for categorical data. Kaplan-Meier
curves were generated to assess the cumulative incidence of all CV events
(coronary and cerebrovascular events combined), and coronary and
cerebrovascular events separately. To examine early and longer-term effects
of raloxifene on CV risk, we compared across treatment groups the number of
women with a CV event (coronary and cerebrovascular events combined and
analyzed individually) after 1 and 4 years of treatment using Pearson chi2
test, and calculated relative risks (RRs) and 95% confidence intervals (CIs)
for each raloxifene treatment group compared with placebo. Post hoc power
calculations, performed using a 1-sided, continuity-corrected Pearson chi2
test, suggested that the data provided 80% power to detect a 1.9-fold
increase in risk of CV events in the first year with raloxifene 60 mg/d
(1.8-fold when raloxifene dose groups were pooled) over placebo in the total
cohort and a 2.5-fold increase in risk in the first year with raloxifene 60
mg/d (2.3-fold when the raloxifene dose groups were pooled) over placebo in
the high-risk subset. Through 4 years, the data provided 80% power to
observe a 29% and 41% RR reduction in the total cohort and high-risk subset,
respectively.
We assessed whether treatment group imbalances in baseline characteristics
confounded the observed results by performing multivariate regression
analysis, including factors for treatment, baseline characteristics, and the
interaction between treatment and baseline characteristics in the model.
For analysis of serum lipid concentrations, percentage changes in these
markers were compared between treatment groups using an analysis of variance
on rank transformed data from all women with a baseline and at least 1
postbaseline measurement. Missing end point observations were imputed by
carrying forward the last available postbaseline observation.



RESULTS



Baseline Characteristics

In the overall study cohort, women in the placebo and raloxifene groups were
generally similar at baseline with regard to demographics, CHD risk factors,
and prior evidence of CHD ( Table 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t1.html> ). The
only exception was a greater proportion of women in the raloxifene groups
compared with placebo with evidence of diabetes at baseline (P = .02). Use
of CV medications also was balanced across treatment groups except for
lipid-lowering therapy, which was greater at baseline among women assigned
to receive raloxifene compared with placebo ( Table 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t1.html> ). On
average, women were 67 years old, 19 years postmenopausal, primarily white
(96%), and had a mean body mass index of 25 kg/m2. Women had a
borderline-high mean serum concentration of total cholesterol (238 mg/dL
[6.15 mmol/L] and LDL-C (155 mg/dL [4.01 mmol/L]). Most women were
normotensive and without established coronary artery disease, with less than
3% reporting a prior coronary event or procedure ( Table 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t1.html> ). There
was no significant difference across treatment groups among women in the
high-CV risk subset in age, other demographics, or in most CHD risk factors
( Table 1
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t1.html> ). As in
the overall cohort, there were more women in the high-risk group with
evidence of diabetes in the raloxifene 60 mg/d compared with the other
groups, although the difference was not statistically significant (P = .10)
in this subset. There also tended to be fewer women with a prior history of
MI (P = .08) or CABG (P = .07) in the raloxifene 60 mg/d group compared with
the other groups. Use of lipid-lowering therapy and most other CV
medications was balanced across treatment groups at baseline except for
calcium channel blockers, which was less (P = .04) in the raloxifene 60 mg/d
group.
Data were analyzed through 48 months of follow-up, during which time 1996
participants (25.9%) discontinued from the study before its completion: 679
in the placebo group, 670 in the raloxifene 60 mg/d group, and 647 in the
raloxifene 120 mg/d group. At 4 years, vital status was known for 5709 (74%)
of the 7705 women originally enrolled in the trial. Women for whom vital
status could not be confirmed included those who discontinued the trial
early, primarily due to an adverse event (n = 910, 46% of discontinuations)
or personal decision (n = 696, 35% of discontinuations). Compared with women
who remained in the trial at 4 years, those who discontinued early were
slightly older (67.2 vs 66.2 years; P<.001), were more years past menopause
(19.9 vs 18.3 years; P<.001), and had slightly higher serum triglyceride
levels (115.9 mg/dL [1.31 mmol/L] and 111.5 mg/dL [1.26 mmol/L]; P = .004).
Among the women who discontinued early, there were no significant
differences across the treatment groups in baseline characteristics or most
risk factors for CV disease (P.30), with the exceptions of total cholesterol
(placebo, 238.2 [6.17]; 60-mg/d raloxifene, 240.5 [6.23]; 120-mg/d
raloxifene, 234.7 mg/dL [6.08 mmol/L]; P = .03) and LDL-C (placebo, 154.8
[4.01]; 60-mg/d raloxifene, 157.5 [4.08]; 120 mg/d raloxifene, 151.7 mg/dL
[3.93 mmol/L]; P = .01).
Ninety-two percent of participants took at least 80% of the study
medication, and 83% took at least 90% of the study medication. Compliance
did not differ significantly across treatment groups (P = .99 for 80%; P =
.55 for 90% compliance).
CV Events

Placebo Event Rates
Figure 2 <http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f2.html>
summarizes the incidence of CV events among women assigned to receive
placebo in the overall cohort and among subsets of women with increasing CV
risk scores. Among all participants, the placebo rate of CV events was 3.7%
for all CV events, 2.1% for coronary events, and 1.6% for cerebrovascular
events. The rate of coronary and cerebrovascular events increased
progressively as the CV risk score increased from 3 to 6 ( Figure 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f2.html> ). The
placebo event rate for women with a CV risk score of 4 or more was
approximately 3.5-fold higher than in the overall cohort: 12.9% for all CV
events, 7.6% for coronary events, and 5.4% for cerebrovascular events.
Treatment Comparisons Overall and in Subsets at Higher CV Risk
A total of 272 women experienced at least 1 major CV event during the trial:
156 experienced at least 1 coronary event (31 fatal; 125 nonfatal) and 117
experienced at least 1 cerebrovascular event (14 fatal,102 nonfatal, and 1
nonfatal event that was subsequently fatal) ( Table 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t2.html> ).
Overall, coronary events included MI or coronary death (44%), unstable
angina (39%), and coronary ischemia (17%); cerebrovascular events included
nonfatal or fatal strokes (68%) and transient ischemic attacks (32%). The
distribution of events was similar among women in the high-risk subset (data
not shown).
Kaplan-Meier survival curves show the cumulative incidence of coronary and
cerebrovascular events together and coronary and cerebrovascular events
alone over time ( Figure 3
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f3.html> ). For the
total study population, there was no evidence for a time trend of an early
increase or a later decrease in risk of CV events with raloxifene ( Figure 3
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f3.html> ). During
the first year of the trial, there were no significant differences across
treatment groups in the number of women with CV events (placebo, 23 [0.8%];
raloxifene 60 mg/d, 25 [0.9%]; raloxifene 120 mg/d, 23 [0.8%]; P = .94);
coronary events (placebo, 15 [0.5%]; raloxifene 60 mg/d, 18 [0.7%];
raloxifene 120 mg/d, 13 [0.5%]; P = .65), or cerebrovascular events
(placebo, 8 [0.3%]; raloxifene 60 mg/d, 7 [0.2%]; raloxifene 120 mg/d, 10
[0.3%]; P = .76). Similar results were obtained when nonfatal and fatal
events were analyzed separately (P.17). Pooling raloxifene dose groups, the
RR of experiencing any CV event during the first year in the overall study
population was 1.05 (95% CI, 0.64-1.72).
In the subset of 1035 women with increased CV risk, there also was no
evidence for an early increase in risk of CV events with raloxifene ( Figure
3 <http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f3.html> ). The
number of CV events during the first year among women in the high-risk
subset was not significantly different across the treatment groups: 10
(3.1%), placebo; 11 (3.0%), raloxifene 60 mg/d; and 9 (2.5%), raloxifene 120
mg/d (P = .86). Pooling the raloxifene dose groups, the RR of experiencing
any CV event during the first year among women in the high-risk subset was
0.88 (95% CI, 0.42-1.87). The Kaplan-Meier curves in the high-risk subset
diverged beginning after approximately the first year, which continued
through 4 years with fewer events in the raloxifene groups compared with
placebo ( Figure 3
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f3.html> ).
At 4 years, the incidence of CV events (coronary and cerebrovascular events
combined or analyzed individually) among all 7705 participants did not
differ significantly across the treatment groups ( Table 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t2.html> ). Similar
results were observed when nonfatal and fatal events were analyzed
separately and for the combined outcomes of coronary death/MI/unstable
angina, and all (fatal plus nonfatal) strokes ( Table 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t2.html> ). Among
the 1035 women in the high-risk subset, there were significantly fewer CV
events among women assigned to raloxifene (RR, 0.60; 95% CI, 0.38-0.95 for
both raloxifene groups; Table 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t2.html> ). The
number of nonfatal CV events also was significantly less in the raloxifene
groups (RR, 0.61; 95% CI, 0.37-0.99 for the 60-mg/d group and RR, 0.50; 95%
CI, 0.30-0.85 for the 120-mg/d group) in this subset, but the number of
fatal CV events was small and not significantly different. Similar trends
were observed when coronary and cerebrovascular events were analyzed
individually; however, most of the differences were not statistically
significant and most of the 95% CIs included 1.0 ( Table 2
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_t2.html> ). The
exception was the combined outcome of all strokes, for which there were
significantly fewer strokes among women receiving raloxifene compared with
those receiving placebo (RR, 0.38; 95% CI, 0.15-0.94). The proportion of
women in the high-risk subset with a prior MI or evidence of diabetes at
baseline, both important risk factors for CV events, was relatively high
(approximately 17% for each), and was slightly imbalanced across treatment
groups. However, adjusting the analyses for these baseline variables did not
change the inference.
To determine whether the significant reduction in CV events observed with
raloxifene among those with 4 or more risk points also was detectable in
subsets of women at lower or higher CV risk, we analyzed the RR of CV events
in the raloxifene groups compared with placebo across the range of CV risk
scores from 3 or more points through 6 or more points ( Figure 4
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f4.html> ). For the
raloxifene 60-mg/d group, the risk of CV events among women with a risk
score of at least 3 was similar to that in the overall cohort and not
reduced significantly compared with placebo. However, as observed for women
with at least 4 CV risk points, raloxifene 60 mg/d was associated with
significantly lower risk of CV events among subsets of women with risk
scores of at least 5 (RR, 0.35; 95% CI, 0.18-0.70) and at least 6 (RR, 0.40;
95% CI, 0.18-0.87). The reduction in risk of CV events with raloxifene 60
mg/d tended to be greater as the CV risk score increased from at least 3
through at least 6 points ( Figure 4
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f4.html> ).
Although the risk of CV events tended to be lower in the raloxifene 120-mg/d
group compared with placebo among women with a risk score at least 5 or at
least 6, the CIs were wide and none were significantly different from
placebo ( Figure 4
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f4.html> ).
Only 202 women who were enrolled in the MORE trial had established CHD
(prior MI, CABG surgery, or percutaneous coronary intervention): 66,
placebo; 56, raloxifene 60 mg/d; and 80, raloxifene 120 mg/d. Among these
women, 4 (7.1%) in the raloxifene 60-mg/d group vs 15 (22.7%) in the placebo
group experienced a coronary or cerebrovascular event during the trial (RR,
0.31; 95% CI, 0.12-0.82) compared with 12 (15%) in the raloxifene 120-mg/d
group (RR, 0.66; 95% CI, 0.33-1.31). Similar results were obtained when
coronary events were analyzed separately, but the differences were not
statistically significant (RR, 0.39; 95% CI, 0.12-1.31 for raloxifene 60
mg/d; RR, 0.92; 95% CI, 0.39-2.13 for raloxifene 120 mg/d). There were 9
cerebrovascular events among women with established CHD: 6 (9%) in the
placebo, 1 (1.7%) in the raloxifene 60-mg/d (RR, 0.20; 95% CI, 0.03-1.25),
and 2 (2.5%) in the raloxifene 120-mg/d (RR, 0.28; 95% CI, 0.06-1.18)
groups. There were 11 CV events among women with established CHD during the
first year of the trial: 5 (7.5%) in the placebo, 3 (5.3%) in the raloxifene
60-mg/d, and 3 (3.7%) in the raloxifene 120-mg/d groups (RR, 0.71; 95% CI,
0.18-2.82 for raloxifene 60 mg/d and RR, 0.50; 95% CI, 0.13-1.94 for
raloxifene 120 mg/d; P = .60).
CV Events Identified From Serial ECGs
Serial ECGs obtained at baseline and at 2 and 4 years were analyzed for
changes consistent with incident Q wave, MI, or myocardial ischemia. The
number of women with incident MI (placebo, 9 [0.4%]; raloxifene 60 mg/d, 8
[0.4%]; raloxifene 120 mg/d, 5 [0.2%]; P = .22) or either incident MI or
myocardial ischemia (placebo, 110 [4.8%], raloxifene 60 mg/d, 89 [3.9%];
raloxifene 120 mg/d, 89 [3.9%]; P = .56) identified from ECG changes from
baseline to 4 years was not significantly different between treatment
groups.
Serum Lipids

Compared with placebo, raloxifene treatment for 4 years resulted in
significant reductions in total and LDL-C but not HDL-C levels ( Figure 5
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f5.html> ).
Reductions in total cholesterol and LDL-C levels were observed in the
raloxifene 60-mg/d and 120-mg/d groups as early as 6 months (median
percentage change: total cholesterol, -5% and -6%; LDL-C, -8% and –9%; both
P<.001 vs placebo), and those levels were maintained throughout the 4 years
of the study ( Figure 5
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f5.html> ). The
median 2.4% decrease in serum triglyceride levels in the placebo group was
significantly different from the median 1.1% and 1.5% increase in serum
triglyceride levels in the raloxifene 60-mg/d and 120-mg/d groups,
respectively (P = .004 for overall treatment group difference). The absolute
median increase in serum triglyceride levels in both raloxifene groups was
0.89 mg/dL (0.01 mmol/L) over 4 years. Relative to placebo, the effects of
raloxifene on total, LDL-C, and HDL-C levels among women in the high-risk
subset were comparable to those observed in the whole cohort although no
significant change in triglyceride levels was observed in this subset (
Figure 5
<http://jama.ama-assn.org/issues/v287n7/fig_tab/joc11015_f5.html> ).
Overall, 1069 women (13.9%) reported taking lipid-lowering therapy at
baseline or at any time during the trial. Excluding these women from the
analyses, raloxifene decreased total cholesterol and LDL-C levels by an
amount comparable to that observed in the overall cohort (data not shown).
Significantly fewer women treated with raloxifene initiated new
lipid-lowering therapy during the course of the study both in the entire
study cohort (placebo, 232 [9.7%]; raloxifene 60 mg/d, 137 [5.9%];
raloxifene 120 mg/d, 122 [5.1%]; P<.001) and among women in the high-risk
subset (placebo, 37 [14.5%]; raloxifene 60 mg/d, 28 [9.8%]; raloxifene 120
mg/d, 22 [7.5%]; P = .03).



COMMENT



In this secondary analysis of data from this large study of osteoporotic
postmenopausal women, raloxifene did not significantly reduce the risk of CV
events in the overall study population, which was at relatively low risk for
CV events, but it did significantly lower the risk of CV events in a subset
of women at high risk for acute coronary events and in those with
established CHD. The number of CV events was small, but there was no
evidence that raloxifene treatment was associated with an early increase in
CV morbidity or mortality in the overall cohort, among women at high risk
for or among women with established CHD.
Women enrolled in the MORE trial were selected for increased risk for
osteoporotic fracture rather than for an increased risk of CV disease 13
<http://jama.ama-assn.org/issues/v287n7/rfull/#r13> ; thus, the overall
study cohort was predominantly women without established CHD and was at
relatively low risk for CV events. Overall, CV mortality in MORE (1.5 per
1000 women-years) was less than that reported for women in the general US
population (2.95 per 1000 women-years). 25
<http://jama.ama-assn.org/issues/v287n7/rfull/#r25>  That MORE enrolled
women worldwide rather than only in the United States may have contributed
to this difference. However, the rate of acute coronary events and mortality
in MORE was in the range of those reported previously for postmenopausal
women in primary CV prevention studies such as the Women's Health Study 26
<http://jama.ama-assn.org/issues/v287n7/rfull/#r26>  and the Air Force/Texas
Coronary Atherosclerosis Prevention Study 27
<http://jama.ama-assn.org/issues/v287n7/rfull/#r27>  and is comparable to
that observed in the Nurses Health Study. 28
<http://jama.ama-assn.org/issues/v287n7/rfull/#r28>  We adapted criteria
established previously for identification of postmenopausal women at high
risk for acute coronary events for enrollment in the RUTH trial 21
<http://jama.ama-assn.org/issues/v287n7/rfull/#r21>  in order to identify
1035 women at increased CV risk in the MORE trial, 202 having experienced a
prior coronary event or undergone a procedure and 833 with no prior coronary
events but with multiple risk factors. The annual coronary event rate in
this subset was about 3.5-fold higher than in the overall cohort, and the
progressive increase in coronary and cerebrovascular event rates as the CV
risk score increased further supports the validity of the method used to
identify women at increased risk. As expected, the incidence of CV events
was highest (23%) among the 202 women who entered the MORE trial with
established CHD and is comparable with the combined incidence of 28% for
coronary and cerebrovascular events observed in HERS. 3
<http://jama.ama-assn.org/issues/v287n7/rfull/#r3>
During the first year of the trial, the number of fatal or nonfatal events
due to coronary artery or cerebrovascular disease did not differ
significantly across treatment groups in the overall study population, among
the 1035 women at high risk or among the 202 women who entered the study
with established CHD. This provides no evidence of an early increase in risk
for CV events although there were too few events during the first year to
draw more definitive conclusions. Hypotheses suggested to explain the early
increase in risk of CV events with HRT observed in HERS, 3
<http://jama.ama-assn.org/issues/v287n7/rfull/#r3>  WEST, 7
<http://jama.ama-assn.org/issues/v287n7/rfull/#r7>  and other studies 5
<http://jama.ama-assn.org/issues/v287n7/rfull/#r5> , 6
<http://jama.ama-assn.org/issues/v287n7/rfull/#r6> , 9
<http://jama.ama-assn.org/issues/v287n7/rfull/#r9>  include a prothrombotic
effect on the arterial system similar to the effect of HRT to increase the
risk of venous thrombolic events or a proinflammatory effect at the vessel
wall that could promote plaque destabilization in women with susceptible
lesions. 29 <http://jama.ama-assn.org/issues/v287n7/rfull/#r29>  The former
seems unlikely given the preliminary evidence that neither raloxifene nor
tamoxifen 30 <http://jama.ama-assn.org/issues/v287n7/rfull/#r30>  cause an
early increase in CV risk, and yet both increase the risk of venous
thromboembolism by a magnitude comparable to that of HRT. 13
<http://jama.ama-assn.org/issues/v287n7/rfull/#r13> , 16
<http://jama.ama-assn.org/issues/v287n7/rfull/#r16> , 17
<http://jama.ama-assn.org/issues/v287n7/rfull/#r17>  The finding that, in
contrast to HRT, 31 <http://jama.ama-assn.org/issues/v287n7/rfull/#r31>
neither raloxifene 19 <http://jama.ama-assn.org/issues/v287n7/rfull/#r19>
nor tamoxifen 32 <http://jama.ama-assn.org/issues/v287n7/rfull/#r32>
increase C-reactive protein levels in postmenopausal women together with
studies showing that HRT increases circulating levels of matrix
metalloproteinase-9 (MMP-9) 33
<http://jama.ama-assn.org/issues/v287n7/rfull/#r33>  lend support to a
proinflammatory hypothesis related to plaque destabilization. However,
direct evidence to support a role for C-reactive protein, MMP-9, or other
markers of inflammation in early CV risk associated with HRT is lacking.
Although raloxifene did not significantly affect the overall risk of CV
events in the total study population, it was associated with a significant
40% reduction in risk of coronary and cerebrovascular events in the 13% of
women at high risk. Because of the post hoc nature of these analyses and the
relatively small number of events involved, the possibility that the
significant risk reduction with raloxifene observed in the high-risk subset
was a chance finding cannot be discounted. However, the reduction in risk
for CV events was statistically significant for raloxifene 60 mg/d across
several subsets of women with increasing risk for CV, was consistent over
time, and was significant in the small subset of women with established CHD.
The separate incidences of coronary and cerebrovascular events were fewer in
the raloxifene 60 mg/d group compared with placebo in the high-risk subset
although these differences did not reach statistical significance. The
effects observed with raloxifene 120 mg/d were less consistent, but whether
this represents a true difference in effect between the doses is unknown.
Overall, raloxifene improved total cholesterol and LDL-C levels with no
effect on HDL-C levels, effects comparable to those previously reported in a
6-month trial of 390 healthy postmenopausal women. 18
<http://jama.ama-assn.org/issues/v287n7/rfull/#r18>  The effects of
raloxifene on serum lipid concentrations in the high-risk subset were
comparable to those observed for the total cohort. Thus, the effect of
raloxifene to lower the risk of CV events in this subset, if real, is not
likely explained by a greater improvement in serum lipid concentrations
among these women.
There are several limitations to our study. The MORE trial was not designed
to test the effect of raloxifene on CV outcomes, and larger trials with
greater numbers of events are needed to confirm these findings.
Cardiovascular events were assessed by self-report and complete supporting
evidence for adjudication of events was not available in many cases. The
study was not designed to confirm vital status or to obtain information on
CV events occurring in women who discontinued the trial early. The overall
incidence and total number of CV events in this trial were small, providing
limited power to detect treatment group differences, especially in the
number of events through the first year of the trial. Finally, all of the
women in MORE had osteoporosis defined by bone mineral density or previous
vertebral fracture, so whether these findings can be generalized to
nonosteoporotic women is uncertain.
In summary, there was no evidence that raloxifene caused an early increase
in risk of CV events, either overall or among postmenopausal women at high
risk for or with CHD. Raloxifene therapy for 4 years did not significantly
affect the overall risk of CV events in the total MORE cohort but did
significantly reduce the risk of CV events among women at high risk for and
among those with established CHD. Before raloxifene is used for prevention
of CV events, these findings must be confirmed by an adequately powered,
randomized trial with CV events as predefined outcomes.



Author/Article Information


Author Affiliations: Division of Epidemiology, Department of Family and
Preventive Medicine, University of California, San Diego, La Jolla (Dr
Barrett-Connor); Department of Epidemiology and Biostatistics, University of
California, San Francisco (Dr Grady); Lilly Research Laboratories,
Indianapolis, Ind (Drs Sashegyi, Anderson, Cox, and Harper); Medical Center,
Railway Hospital, Warsaw, Poland (Dr Hoszowski); and Department of Public
Health Services, Wake Forest University, Winston-Salem, NC (Dr Rautaharju).

Corresponding Author and Reprints: Elizabeth Barrett-Connor, MD, Division of
Epidemiology, Department of Family and Preventive Medicine, University of
California, San Diego School of Medicine, 9500 Gilman Dr, No. 0607, La
Jolla, CA 92093-0607 (e-mail: [log in to unmask]
<mailto:[log in to unmask]> ).
Financial Disclosures: Dr Barrett-Connor has served as a consultant for and
has received research support from Eli Lilly & Co. Dr Grady receives or has
received salary support via contracts with the University of California, San
Francisco, from Berlex Laboratories, Eli Lilly, Pfizer, and Wyeth-Ayerst
Research. Drs Sashegi, Anderson, Cox, and Harper are full-time employees of
and own stock in Eli Lilly & Co.
Author Contributions: Dr Barrett-Connor had full access to all the data in
this study and takes responsibility for the integrity of the data and
accuracy of the data analysis.
Study concept and design: Grady, Anderson, Cox, Harper.
Acquisition of data: Barrett-Connor, Sashegyi, Hoszowski, Rautaharju.
Analysis and interpretation of data: Barrett-Connor, Grady, Anderson, Cox,
Rautaharju, Harper.
Drafting of the manuscript: Barrett-Connor, Anderson, Cox.
Critical revision of the manuscript for important intellectual content:
Barrett-Connor, Grady, Sashegyi, Anderson, Cox, Hoszowski, Rautaharju,
Harper.
Statistical expertise: Grady, Sashegyi.
Obtained funding: Hoszowski.
Administrative, technical, or material support: Anderson, Cox, Rautaharju,
Harper.
Study supervision: Barrett-Connor, Harper.
Medical expertise: Anderson.
Principal investigator of ECG laboratory: Rautaharju.
Funding/Support: This study was funded by Eli Lilly & Co.
Role of the Sponsor: The MORE trial protocol was designed by the sponsor in
consultation with the coordinating center at the University of California,
San Francisco. Study medication and randomization codes were provided by the
sponsor. Data on cardiovascular events, on which the present study is based,
were by investigators at their respective sites without regard to causality
or treatment group assignment. The cardiologists who adjudicated the
cardiovascular events were compensated for their time but were not otherwise
involved in the trial or associated with the sponsor. Data analyses were
performed by the statistical group of the sponsor (Dr Sashegyi) with review
by and input from the corresponding author (Dr Barrett-Connor). The
manuscript was written by Drs Barrett-Connor and Cox and was reviewed by
each of the coauthors. The manuscript also was reviewed before submission
both by a study publications committee consisting of a majority of
nonsponsor investigators and by sponsor staff.
Acknowledgment: We are indebted to Kathryn Krueger, MD, for implementation
of the study and contributions to the data analysis plan, Somnath Sarkar,
PhD, for contributions to the statistical analysis plans and for his
guidance and consultancy during the statistical analyses, and to Yiyong Fu,
MS, for extensive statistical programming support. We also thank Imre Pavo,
MD, for critically reviewing the manuscript, William Groh, MD, and Chamoun
Chamoun, MD (Krannert Institute of Cardiology, Indiana University School of
Medicine) for adjudication of the cardiovascular events, and all of the MORE
trial investigators who made this study possible.
A list of the MORE Investigators has been published previously (JAMA.
1999;282:637-656).




REFERENCES



1. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr1>
Mosca L, Manson JE, Sutherland SE, Langer RD, Manolio T, Barrett-Connor E.
Cardiovascular disease in women.
Circulation.
1997;96:2468-2482.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9337227>
2. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr2>
Barrett-Connor E, Grady D.
Hormone replacement therapy, heart disease, and other considerations.
Annu Rev Public Health.
1998;19:55-72.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9611612>
3. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr3>
Hulley S, Grady D, Bush T, et al.
Randomized trial of estrogen plus progestin for secondary prevention of
coronary heart disease in postmenopausal women.
JAMA.
1998;280:605-613.
ABSTRACT <http://jama.ama-assn.org/issues/v280n7/abs/joc80678.html>   |
FULL TEXT <http://jama.ama-assn.org/issues/v280n7/rfull/joc80678.html>   |
PDF <http://jama.ama-assn.org/issues/v280n7/rpdf/joc80678.pdf>   |   MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9718051>
4. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr4>
Herrington DM, Reboussin DM, Brosnihan KB, et al.
Effects of estrogen replacement on the progression of coronary-artery
atherosclerosis.
N Engl J Med.
2000;343:522-529.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10954759>
5. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr5>
Lenfant C.
Statement from Claude Lenfant, MD, Director, National Heart, Lung, and Blood
Institute on preliminary trends in the Women's Health Initiative.
WHI HRT Update. Bethesda, Md: National Heart, Lung, and Blood Institute;
April 6, 2000. Available at: http://www.nhlbi.nih.gov/whi/hrt-en.htm
<http://www.nhlbi.nih.gov/whi/hrt-en.htm> . Accessed January 22, 2002.
6. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr6>
Rossouw JE.
Early risk of cardiovascular events after commencing hormone replacement
therapy.
Curr Opin Lipidol.
2001;12:371-375.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11507320>
7. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr7>
Viscoli CM, Brass LM, Kernan WN, Sarrel PM, Suissa S, Horwitz RI.
A clinical trial of estrogen-replacement therapy after ischemic stroke.
N Engl J Med.
2001;345:1243-1249.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11680444>
8. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr8>
Hemminki E, McPherson K.
Impact of postmenopausal hormone therapy on cardiovascular events and
cancer: pooled data from clinical trials.
BMJ.
1997;315:149-153.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9251544>
9. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr9>
Hemminki R, McPherson K.
Value of drug-licensing documents in studying the effect of postmenopausal
hormone therapy on cardiovascular disease.
Lancet.
2000;355:566-569.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10683020>
10. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr10>
Grodstein F, Manson JE, Stampfer MJ.
Postmenopausal hormone use and secondary prevention of coronary events in
the Nurses' Health Study: a prospective, observational study.
Ann Intern Med.
2001;135:1-8.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11434726>
11. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr11>
Heckbert SR, Kaplan RC, Weiss NS, et al.
Risk of recurrent coronary events in relation to use and recent initiation
of postmenopausal hormone therapy.
Arch Intern Med.
2001;161:1709-1713.
ABSTRACT <http://archinte.ama-assn.org/issues/v161n14/abs/ioi00867.html>   |
FULL TEXT <http://archinte.ama-assn.org/issues/v161n14/rfull/ioi00867.html>
|   PDF <http://archinte.ama-assn.org/issues/v161n14/rpdf/ioi00867.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11485503>
12. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr12>
Alexander KP, Newby LK, Hellkamp AS, et al.
Initiation of hormone replacement therapy after acute myocardial infarction
is associated with more cardiac events during follow-up.
J Am Coll Cardiol.
2001;38:1-7.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11451256>
13. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr13>
Cummings SR, Eckert S, Krueger KA, et al.
The effect of raloxifene on risk of breast cancer in postmenopausal women.
JAMA.
1999;281:2189-2197.
ABSTRACT <http://jama.ama-assn.org/issues/v281n23/abs/joc90196.html>   |
FULL TEXT <http://jama.ama-assn.org/issues/v281n23/rfull/joc90196.html>   |
PDF <http://jama.ama-assn.org/issues/v281n23/rpdf/joc90196.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10376571>
14. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr14>
Ettinger B, Black DM, Mitlak BH, et al.
Reduction of vertebral fracture risk in postmenopausal women with
osteoporosis treated with raloxifene: results from a 3-year randomized
clinical trial.
JAMA.
1999;282:637-645.
ABSTRACT <http://jama.ama-assn.org/issues/v282n7/abs/joc90496.html>   |
FULL TEXT <http://jama.ama-assn.org/issues/v282n7/rfull/joc90496.html>   |
PDF <http://jama.ama-assn.org/issues/v282n7/rpdf/joc90496.pdf>   |   MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10517716>
15. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr15>
Cauley JA, Norton L, Lippman ME, et al.
Continued breast cancer risk reduction in postmenopausal women treated with
raloxifene.
Breast Cancer Res Treat.
2001;65:125-134.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11261828>
16. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr16>
Grady D, Wenger NK, Herrington D, et al.
Postmenopausal hormone therapy increases risk for venous thromboembolic
disease.
Ann Intern Med.
2000;132:689-696.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10787361>
17. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr17>
Fisher B, Costantino JP, Wickerham DL, et al.
Tamoxifen for prevention of breast cancer: report of the National Surgical
Adjuvant Breast and Bowel Project P-1 Study.
J Natl Cancer Inst.
1998;90:1371-1388.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9747868>
18. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr18>
Walsh BW, Kuller LH, Wild RA, et al.
Effects of raloxifene on serum lipids and coagulation factors in healthy
postmenopausal women.
JAMA.
1998;279:1445-1451.
ABSTRACT <http://jama.ama-assn.org/issues/v279n18/abs/joc72104.html>   |
FULL TEXT <http://jama.ama-assn.org/issues/v279n18/rfull/joc72104.html>   |
PDF <http://jama.ama-assn.org/issues/v279n18/rpdf/joc72104.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9600478>
19. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr19>
Walsh BW, Paul S, Wild RA, et al.
The effects of hormone replacement therapy and raloxifene on C-reactive
protein and homocysteine in healthy postmenopausal women: a randomized,
controlled trial.
J Clin Endocrinol Metab.
2000;85:214-218.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10634389>
20. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr20>
Saitta A, Altavilla D, Cucinotta D, et al.
Randomized, double-blind, placebo controlled study on the effects of
raloxifene and hormone replacement therapy on plasma nitric oxide
concentrations, endothelin-1 levels and endothelium-dependent vasodilation
in postmenopausal women.
Arterioscler Thromb Vasc Biol.
2001;21:1512-1519.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11557681>
21. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr21>
Mosca L, Barrett-Connor E, Wenger NK, et al.
Design and methods of the Raloxifene Use for The Heart (RUTH) study.
Am J Cardiol.
2001;88:392-395.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11545760>
22. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr22>
Rautaharju PM, Calhoun HP, Chaitman BR.
NOVACODE serial ECG classification system for clinical trials and
epidemiologic studies.
J Electrocardiol.
1992;24(suppl):179-187.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
1552254>
23. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr23>
Rautaharju PM, Park LP, Chaitman BR, Rautaharju F, Zhang ZM.
The NOVACODE criteria for classification of ECG abnormalities and their
clinically significant progression and regression.
J Electrocardiol.
1998;31:157-187.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9682893>
24. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr24>
Friedewald WT, Levy RI, Fredrickson DS.
Estimation of the concentration of low-density lipoprotein cholesterol in
plasma, without use of the preparative ultracentrifuge.
Clin Chem.
1972;18:499-502.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
4337382>
25. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr25>
Biostatistical Fact Sheet.
Women and cardiovascular disease.
Washington, DC: American Heart Association. Available at:
http://216.185.102.50/statistics/biostats/biowo.htm
<http://216.185.102.50/statistics/biostats/biowo.htm> . Accessed October 22,
2001.
26. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr26>
Lee IM, Cook NR, Manson JE, Buring JE, Hennekens CH.
Beta-carotene supplementation and incidence of cancer and cardiovascular
disease: the Women's Health Study.
J Natl Cancer Inst.
1999;91:2102-2106.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10601381>
27. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr27>
Downs JR, Clearfield M, Weis S, et al.
Primary prevention of acute coronary events with lovastatin in men and women
with average cholesterol levels.
JAMA.
1998;279:1615-1622.
ABSTRACT <http://jama.ama-assn.org/issues/v279n20/abs/joc80151.html>   |
FULL TEXT <http://jama.ama-assn.org/issues/v279n20/rfull/joc80151.html>   |
PDF <http://jama.ama-assn.org/issues/v279n20/rpdf/joc80151.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9613910>
28. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr28>
Grodstein F, Manson JE, Colditz GA, Willett WC, Speizer FE, Stampfer MJ.
A prospective, observational study of postmenopausal hormone therapy and
primary prevention of cardiovascular disease.
Ann Intern Med.
2000;133:933-941.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11119394>
29. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr29>
Herrington DM.
The HERS trial results: paradigms lost?
Ann Intern Med.
1999;131:463-466.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10498564>
30. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr30>
Reis SE, Costantino JP, Wickerham DL, Tan-Chiu E, Wang J, Kavanah M.
Cardiovascular Effects of Tamoxifen in Women With and Without Heart Disease:
Breast Cancer Prevention Trial.
J Natl Cancer Inst.
2001;93:16-21.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11136837>
31. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr31>
Cushman M, Legault C, Barrett-Connor E, et al.
Effect of postmenopausal hormones on inflammation-sensitive proteins: the
postmenopausal Estrogen/Progestin interventions (PEPI) study.
Circulation.
1999;100:717-722.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10449693>
32. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr32>
Cushman M, Costantino JP, Tracy RP, et al.
Tamoxifen and cardiac risk factors in healthy women: suggestion of an
anti-inflammatory effect.
Arterioscler Thromb Vasc Biol.
2001;21:255-261.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11156862>
33. <http://jama.ama-assn.org/issues/v287n7/rfull/#rr33>
Zanger D, Yang BK, Ardans J, et al.
Divergent effects of hormone therapy on serum markers of inflammation in
postmenopausal women with coronary artery disease on appropriate medical
management.
J Am Coll Cardiol.
2000;36:1797-1802.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11092646>


Edward E. Rylander, M.D.
Diplomat American Board of Family Practice.
Diplomat American Board of Palliative Medicine.