Vous êtes sur la page 1sur 8

Cardiovascular Research 53 (2002) 550–557

www.elsevier.com / locate / cardiores

Review

Hormones, genetic factors, and gender differences in cardiovascular


disease
Jacques E. Rossouw*
Women’ s Health Initiative, National Heart, Lung and Blood Institute, 1 Rockledge Center, Suite 300, 6705 Rockledge Drive, Bethesda,
MD 20892 -7966, USA

Received 6 July 2001; accepted 30 August 2001

Keywords: Gender; Gene therapy

1. Gender differences in age of onset of coronary heart due to the lifelong difference in HDL cholesterol levels;
disease however, this difference is a consequence of having the Y
chromosome. During fetal development, the Y chromo-
On average, women develop heart disease some 10–15 some directs the formation of testes rather than ovaries,

Downloaded from by guest on September 12, 2015


years later than men. This raises the question of whether and the testes in turn produce testosterone and dihydrotes-
there is some aspect of ‘femaleness’ which reduces risk, or tosterone rather than estrone and estradiol as the primary
whether there is some aspect of ‘maleness’ that raises risk. sex steroids [6]. The high levels of male sex hormones
To date, most attention has been focused on the hypothesis drive down the HDL cholesterol levels, with consequent
that endogenous estrogen is cardioprotective in women [1]. higher early risk of CHD. Thus, the gender difference in
Rising rates of coronary heart disease (CHD) after the CHD may well be due the most basic genetic difference
menopause, and after oophorectomy, are among the strands between men and women, which is the presence of the Y
of evidence in humans that endogenous estrogen may chromosome.
prevent CHD [2]. However, upon closer examination this Some of the observational studies in adult males suggest
evidence is not persuasive, and in fact the evidence is that higher levels of testosterone are associated with higher
amenable to alternative explanations. (rather than lower) levels of HDL cholesterol, and with
During the first 3 decades of adult life, low-density lower risk of CHD [7]. Intracoronary infusion of testo-
lipoprotein (LDL) cholesterol levels are lower in women sterone induces coronary artery dilatation and increases
than men, and this may contribute to the delayed onset of coronary blood flow in men with coronary artery disease
CHD in women. A more widely held explanation for the [8]. These observations do not fit the concept that testo-
later onset of CHD in women is their higher high-density sterone is harmful to the male cardiovascular system. On
lipoprotein (HDL) cholesterol levels, attributed to higher the other hand, androgen deprivation in men is associated
endogenous estrogen levels in women. However, the with enhanced endothelium-dependent dilatation in men
difference in HDL cholesterol between women and men is successfully treated for prostate cancer, while dilatation is
an androgen effect, not an estrogen effect. Up to puberty, reduced in genetic females taking high dose androgens
young men and women have similar HDL cholesterol [9,10]. In the laboratory, androgen receptor expression is
levels. At puberty, concurrent with the rise in endogenous greater in macrophages from male than from female
testosterone levels, the HDL cholesterol levels in young donors, and lipid loading of male (but not female) macro-
men decline to the adult level [3,4]. A 20% difference in phages was increased by dihydrotestosterone [11].
HDL cholesterol levels predicts at least a 20% difference Dihydrotestosterone also increases the adhesion of mono-
in CHD rates in the short term, and may predict even cytes to endothelial cells, increases the expression of
larger differences in CHD rates over a lifetime [5]. Thus, endothelial vascular cell adhesion molecule-1, increases
the entire gender difference in CHD risk may indeed be platelet expression of thromboxane receptors, and in-
creases platelet aggregation [12,13]. However, there are

*Tel.: 11-301-435-6669; fax: 11-301-480-5158.


E-mail address: rossouwj@nih.gov (J.E. Rossouw). Time for primary review 31 days.

0008-6363 / 02 / $ – see front matter  2002 Elsevier Science B.V. All rights reserved.
PII: S0008-6363( 01 )00478-3
J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557 551

even more gaps in our knowledge about the real effects of lower risk is found in those who have recently started
testosterone than there are for estrogen. As in the case of estrogen as well as in long-term users [16].
estrogen, clinical trials with hard endpoints using an- These findings from observational epidemiology provide
drogens, androgen antagonists, or selective androgen re- the rationale for clinical trials testing whether and to what
ceptor modulators will ultimately be needed to resolve this degree current use of postmenopausal hormone therapy
issue, but it is difficult to anticipate that such trials will be prevents a first heart attack. However, the observational
done in the near future. epidemiology is not sufficient to prove the case, because
The statement that CHD rates in women rise steeply even the best studies may be subject to a variety of
after the age of menopause, and the corollary that this is systematic biases that could lead to an overestimation of
due to lower levels of estrogen after that age, is also open benefit and an underestimation of harm from hormone
to question. In actuality, there is no evidence for an therapy, hence the need for an unbiased estimate from
increase in the year-on-year rate of increase in CHD clinical trials. These biases in observational studies include
around the age of menopause. The linear relationship healthy user selection bias, compliance bias, surveillance
between age and CHD incidence as seen on a semilogarit- bias, and survivor bias [14,17].
hmic plot shows that there is a constant proportional In combination, these biases will lead to a systematic
increase in CHD incidence with age, with no inflection overestimation of benefit, and an underestimation of risk in
upward at the average age of menopause [14]. This is observational studies. Adjusting for baseline differences in
evidence for an age effect, and evidence against an effect risk factors will mitigate healthy user selection bias, but
of menopause. The Nurses’ Health Study investigators will not correct for compliance, surveillance, or survivor
have reported that, after controlling for age and smoking bias. Thus, the real benefit for CHD may be much less than
status, the natural menopause is not associated with an predicted by the observational studies, or there may be no
increased risk for CHD [2]. The same investigators have benefit at all [14]. The clinical trials to date have failed to
reported that, in contrast to the natural menopause, bilater- show overall benefit for CHD over the short term (ongoing
al oophorectomy is associated with an increased risk for trials will ascertain the long term effects particularly in

Downloaded from by guest on September 12, 2015


CHD in women who had never taken estrogen after women without prevalent CHD) [18].
menopause. However, the study had very few cases of
CHD in women with oophorectomy, and the increased risk
was no longer significant in the multivariate analysis.
Nonetheless, the use of estrogens appeared to eliminate 3. Biological mechanisms for cardioprotection by
this increased risk. It is possible that women who have a estrogen
hysterectomy and bilateral oophorectomy (often done for
menorrhagia / metrorrhagia associated with endometrial The possibility that estrogen may reduce CHD risk has
hyperplasia) are at higher risk for CHD because of the stimulated a wide variety of studies that attempt to explain
co-existence of metabolic risk factors such as central the presumed benefit. Because it was unexpected, fewer
obesity, high blood pressure, lipid disorders, and glucose studies have been done to explain the apparent excess risk
intolerance. The finding of an apparent lower risk in early in the course of treatment, found in recent clinical
women who subsequently used estrogen would be subject trials (see below). Effects of estrogen that may predict
to the biases discussed below. In summary, the post- benefit include: lowered LDL cholesterol and lipoprotein
menopausal increase in risk is most likely due to age and (a) levels, raised HDL cholesterol levels, reduced fibrino-
not the menopause, and the increase in risk in women after gen levels and enhanced fibrinolysis (reduced plasminogen
oophorectomy may be due to confounding by other risk activator inhibitor-1 (PAI-1) and increased D-dimer levels),
factors. reduced homocysteine levels, antioxidant properties, and
improved endothelial function (e.g. reduced E-selectin
levels and enhanced flow-mediated dilatation) [1]. Es-
trogen and progesterone reduce lipid accumulation in
2. Observational studies of estrogen users macrophages from female, but not male, donors [19]. On
the other hand, several mechanisms that might increase
By far the most persuasive evidence in favor of a risk after estrogen administration have been found: tri-
protective effect for estrogen comes from the large number glycerides increase, some coagulation markers increase
of cohort studies comparing CHD risk in postmenopausal (e.g. Factor VII, prothrombin fragments 112, activated
women currently using estrogen to never-users. These protein C resistance), and the inflammatory marker C-
studies have shown consistently that CHD risk is 35–50% reactive protein increases [20–23]. Some observational
lower in estrogen users, after adjusting for other risk studies have suggested that certain of the lipid markers
factors [15,16]. The lower risk has been found in studies of changed by estrogen administration are associated with
estrogen alone, as well as in studies of estrogen in higher relative risks for CHD in women than in men,
combination with a progestin [16]. For healthy women, the including HDL cholesterol and triglycerides [23,24]. How-
552 J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557

ever, particularly in respect of coagulation and inflamma- 4. Genetic mechanisms for cardioprotection by
tion, laboratory measurements do not predict whether the estrogen
predominant effect will be favorable or unfavorable.
Studies of venous thromboembolism (including clinical The role of the major monogenic disorders such as
trials) provide unequivocal evidence that the overall effect familial hypercholesterolemia and familial hyper-
is indeed procoagulant [25,26]. homocysteinemia in arterial disease is well established.
Compounding this difficulty in interpreting laboratory Even within kindreds with familial hypercholesterolemia,
measurements is the fact that progestins counteract some affected females develop CHD later than in males, pre-
of the estrogen effects, and that the clinical expression of sumably because the same (unknown) factors that delay
metabolic changes may be time-dependent. The early onset in non-affected females continue to operate in
excess risk for arterial disease observed in the Heart and affected females [38]. Though estrogen administration
Estrogen / Progestin Replacement Study (HERS) may have lowers LDL cholesterol in women with familial hyper-
been due to an initial procoagulant or inflammatory effect cholesterolemia [39], this pharmacologic property does not
on susceptible plaques, while the favorable effects in the necessarily mean that endogenous estrogen has the same
survivors may be due to the later assertion of the generally effect. There is no sex difference in the elevated LDL
favorable lipid effects [27]. In HERS, participants with cholesterol levels of individuals with familial hypercholes-
higher levels at baseline had the largest decrease in terolemia [38]. Rather, as in non-affected individuals,
lipoprotein (a) on treatment, and had a more favorable factors that blunt the impact of a given level of LDL
clinical outcome than participants with lower baseline cholesterol in females (or accelerate it in males) need to be
levels [28]. The role of estrogen’s direct vascular effect is sought.
unclear, since impaired endothelial function has not yet The importance to arterial disease of polymorphisms in
been established as a risk factor for CHD. Interestingly, the genes that code for coagulation proteins and markers of
current estrogen users do not appear to have a lower risk inflammation is not clear. For example, a variety of
for angina, as one would expect if direct vascular effects polymorphisms in the fibrinogen gene are known to affect

Downloaded from by guest on September 12, 2015


were important [16]. On the other hand, in a clinical trial circulating fibrinogen levels, and fibrinogen levels have
sublingual estrogen relieved exercise-induced angina [29]. been associated with arterial disease [40]. However,
Overall, the studies of mechanisms have not resolved the studies seeking to quantify the association of a fibrinogen
core issue of whether estrogens protect against CHD. gene polymorphism with arterial disease have generally
It is important to realize that almost all the studies of yielded disappointing results. This may be because any
mechanism used oral estrogen preparations, and may turn individual polymorphism has a modest influence on fibrin-
out to have little relevance towards explaining the gender ogen levels, and the levels are simultaneously affected by
difference in CHD. Ovarian estrogen directly enters the both other genetic factors, and importantly, by environ-
systemic circulation through the inferior vena cava, unlike mental factors [40]. Hence, it is difficult to discern the
oral estrogens, which enter the portal vein and undergo direct effect of any individual polymorphism on clinical
first pass hepatic circulation. Because of extensive metabo- disease. In addition to fibrinogen, polymorphisms in the
lism in the liver, in order to achieve similar blood levels genes for FVII, FXIII, FV, prothrombin, thrombomodulin,
the dose of oral estrogen needs to be approximately ten tissue plasminogen activator, PAI-1, and platelet-mem-
times that of non-oral (e.g transdermal) estrogen. These brane glycoproteins have been described [40]. Except for
doses of estrogens profoundly influence the hepatic metab- fibrinogen, the relationships of the phenotypic factors
olism of a variety of proteins, including lipid apoproteins, affected by these polymorphisms with arterial disease are
coagulation proteins, and (probably) C-reactive protein uncertain. A possible interaction of gender with poly-
[20–22,30–34]. The large effects on lipids and coagulation morphisms in the FVII gene has been described, in that
proteins described for oral estrogens are greatly attenuated, plasma activity of FVII in males varies markedly accord-
absent, or in the opposite direction with non-oral estrogens. ing to the presence of three of these polymorphisms, but in
Non-oral estrogens have very modest effects on lowering females these polymorphisms are associated with much
LDL-cholesterol and lipoprotein (a), have no effect or smaller changes in FVII activity [41]. Estrogen affects
reduce triglycerides, have no effect on HDL-cholesterol, many of these phenotypic factors [20–22,32–35], but in
and have a modest or no effect on levels of coagulation general it is not known whether there is a protective or
proteins [30–34]. Non-oral estrogens retain the ability to harmful interaction of estrogen with the polymorphisms as
improve endothelial function [35]. Additional mechanistic risk factors for arterial disease.
studies, as well as epidemiologic studies and clinical trials, Both males and females have a- and b-estrogen re-
that focus on the role of non-oral estrogen preparations are ceptors in the vascular endothelium, smooth muscle, and
needed. Of interest, epidemiologic studies in post- myocardium, though receptor numbers may be higher in
menopausal women have not shown an association of females because estrogen induces their expression [1,42].
endogenous estrogen levels with CHD [36,37]. One group of investigators found a gender difference for
J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557 553

the non-genomic effects of estrogen on the response of causing the harm. It remains possible that the lipid changes
coronary arteries to acetylcholine. Males did not show the induced by HRT may result in long-term benefit for
reversal of acetylcholine-induced vasoconstriction in cor- women who survive the first years. This question remains
onary arteries after acute estrogen administration found in unanswered at this point, but for many women the answer
females [43]. On the other hand, genetic males receiving may be moot if ways of avoiding the early harm are not
long-term estrogen therapy show enhanced flow-mediated found.
dilatation [44]. The presence of higher numbers of func-
tional estrogen receptors in females may relate to the
inhibition of injury-induced vascular intimal thickening by 6. Mechanisms for explaining an increased risk for
estrogen in female arteries, and the lower prevalence of left arterial disease shortly after commencing HRT
ventricular hypertrophy in women compared to men
[1,45,46]. Estrogen receptor numbers are lower in female Hypotheses for explaining early harm center around a
atherosclerotic arteries [46]. There is some potential for potential interaction of HRT with coagulation and / or
polymorphisms of the estrogen receptors themselves to inflammation mechanisms in a subset of women with
affect the clinical outcome of estrogen administration. For vulnerable plaques. According to these hypotheses, expo-
example, the common ER Pvu II and Xbal genotypes are sure to HRT for certain women with a susceptibility factor
not associated with differences in HDL cholesterol levels, in the form of an elevated blood or tissue level, or with an
but after estrogen administration women who are hetero- augmented response to HRT perhaps due to a genetic
zygous for the genotype have a greater elevation of HDL mutation, may result in plaque destabilization and throm-
cholesterol [47]. The clinical relevance of this finding is bosis.
unknown. HRT induces a wide variety of changes in coagulation
factors in healthy women. Some of these are likely to be
procoagulant, while others are likely to profibrinolytic,
5. Evidence of early harm from clinical trials hence the clinical effect cannot be predicted from labora-

Downloaded from by guest on September 12, 2015


tory studies. Because of an emphasis on a few factors, in
The evidence emerging from trials of hormone replace- particular reductions in levels of fibrinogen and PAI-1,
ment therapy (HRT) to prevent coronary heart disease many scientists believed that HRT exerted a favorable
(CHD) continues to confound the belief that HRT is effect on coagulation. As noted above, clinical trials and
cardioprotective. On the contrary, a fairly consistent epidemiologic studies confirm that the net effect of HRT is
pattern of early harm is emerging from secondary and, to a to promote coagulation in the venous system [25,26].
lesser extent, from primary prevention trials. Long term There is a growing suspicion that coagulation factors
effects of HRT on the cardiovascular system, and also on beyond platelets may be more important in arterial disease
the overall health of postmenopausal women, are being than previously appreciated. Among the coagulation mech-
evaluated in the ongoing primary prevention trials [18]. anisms the Factor V Leiden (FVL) mutation and the
The findings from four clinical trials of HRT in women prothrombin G20210A mutation are currently of most
are consistent with an early excess of arterial cardiovascu- interest for both venous and arterial disease.
lar events: HERS, the Papworth HRT and Atherosclerosis FVL produces a coagulation factor V which resists the
Survival Enquiry (PHASE), the Women’s Estrogen for action of the major natural anticoagulant, activated protein
Stroke Trial (WEST), and the Women’s Health Initiative C. Hence, the phenotypic expression can be measured as
(WHI) [27,48–50]. In addition, a combined analysis of activated protein C (APC) resistance. The prevalence of
short-term trials (typically lasting a year) in healthy heterozygous FVL in whites is about 4.8% (range 3–7%),
women showed a non-significant 39% excess risk for CHD with a much lower prevalence of 0.05% in Blacks and
[51]. Support for short-term harm comes also from the Asians [57]. FVL is associated with venous thromboembo-
early trials in men, and several observational studies of lism (VTE), and is found in about 18% of unselected cases
new users of HRT among women with prior CHD [52–55] of VTE and about 40% of selected cases with presumed
and one of healthy women in the Group Health Co- familial thrombophilia. The risk for VTE associated with
operative study [56]. The evidence comes from secondary FVL is amplified by exposure to oral contraceptives (OCs)
and primary prevention trials, trials using conjugated and the risk associated with APC resistance is amplified by
equine estrogens (CEE) with and without medroxyproges- HRT use [57–59]. In contrast, the association of FVL with
terone (MPA), and trials of estradiol (oral and transder- CHD has not been found in men, and in women it has been
mal). Many of the trials have weaknesses (and in the case inconsistent [60–63]. In a case–control study of younger
of WHI the data have not been published); individually women, FVL was associated with MI in those with other
their findings can be rationalized away, but in aggregate risk factors for CHD, particularly smoking [62]. However,
the findings are persuasive. Therefore, the question is no two other case–control studies in women failed to show an
longer whether the early harm is real, but rather what is association with CHD, or an interaction with HRT use
554 J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557

[60,65]. In one cross-sectional study of women referred to On the other hand, for women who survive the acute phase
a lipid clinic for management of dyslipidemia, HRT use in or who do not have vulnerable plaques, the long-term
the absence of FVL was associated with a reduced risk for effect of elevated levels of MMP-9 might be favorable,
atherothrombotic arterial disease, while HRT use in the since the proteinases might prevent the accumulation of
presence of FVL was associated with an increased risk matrix proteins and improve arterial compliance.
[63]. Thus, the status of FVL as a risk factor for CHD is These intriguing observations and hypotheses have great
much less certain than for VTE, and only one study potential relevance to women, as they affect the future use
suggested an interaction with HRT. On the other hand, of exogenous estrogens in the form of HRT. It is unknown
there is insufficient data to exclude a possible role for FVL whether endogenous estrogens affect these markers of
in CHD in women, particularly in the context of CHD inflammation and coagulation, and therefore unknown
associated with HRT use. whether these factors have a role in explaining the gender
The prothrombin G20210A mutation is procoagulant difference in cardiovascular disease. If anything, the
because it increases prothrombin levels. The prevalence is known and suspected interactions of estrogen with these
lower than FVL, being found in about 2.7% of whites markers go against the observation of delayed onset in
(range 1–4%). As for FVL, the prevalence is much lower women, since they raise the risk for arterial disease in
in Blacks and Asians (0.06%) [57]. It is associated with women. Younger women may have higher rates of VTE
VTE, being found in 7% of unselected cases and 16% of events than men, but it is not known whether endogenous
selected cases of VTE. In women, but not in men, the estrogen or use of oral contraceptives explain that observa-
mutation has been found to be associated with CHD tion [72].
[64–67]. A cross-sectional analysis of women with
dyslipidemia found that the HRT use was associated with
decreased risk for atherothrombotic disease in the absence 7. Conclusions
of the G20210A mutation, and increased risk in its
presence [66]. A case–control study of non-fatal MI in The gender difference in the age of onset of CHD is as

Downloaded from by guest on September 12, 2015


women with hypertension (a parallel study of women yet unexplained. A natural starting point is that the
without hypertension was negative) found that presence of biologic effects of the XX and XY chromosomes are
the mutation alone was associated with an odds ratio for expressed through sex hormones, and that these explain the
MI of 1.5 (95% CI 0.6–1.4) and current use of HRT alone gender difference in CHD. However, the respective roles
was associated with an odds ratio of 0.9 (0.3–7.7) [64]. of female and male sex hormones in this regard remain
However, the presence of both the mutation and use of unclear. To date, research has focused overwhelmingly on
HRT was associated with an odds ratio of 10.9 (2.2–55.2, estrogen. The substantial evidence in favor of a protective
P50.002). This supra-multiplicative effect of the com- effect of estrogen in women is being called into question
bined factors represents true synergism. If confirmed, a by recent clinical trial data indicating that HRT is not
large synergistic effect between HRT and a prothrombotic protective, and at least in women with existing heart
factor in the range of 3–5% could explain the pattern of disease may initially increase risk. Because of the in-
early harm and late benefit seen in the HERS trial [64]. creased in risk in secondary prevention trials, and the
HRT also affects several markers of inflammation, some absence of published clinical trial data for primary preven-
in a potentially unfavorable direction and some in a tion, HRT is no longer being recommended for prevention
potentially favorable direction. HRT increases circulating of CHD [73]. It remains possible that HRT given for many
levels of C-reactive protein and matrix metalloproteinases years may be cardioprotective.
including matrix metalloproteinase-9 (MMP-9) [22,68,69]. Oral estrogen has pronounced effects on blood markers
On the other hand, HRT decreases circulating levels of cell and cell function, including endothelial cells and mono-
adhesion molecules E-selectin, intercellular adhesion cyte-derived macrophages. Some of these effects may be
molecule-1, and vascular cell adhesion molecule-1, and in favorable, others unfavorable, and thus laboratory studies
the short term improves endothelial function [70,71]. An cannot predict clinical outcomes. Clinical trials indicate
intriguing hypothesis put forward by Cannon. et al. centers that the net effect of estrogen is prothrombotic. The
on the effects of estrogen on MMP-9 [68]. According to metabolic effects of oral estrogen are very different from
this hypothesis, acute administration may be harmful, and those of non-oral (e.g. transdermal) estrogen; the latter is
chronic administration may be beneficial. Estrogen de- more likely to mimic the effects of endogenous estrogen,
creases PAI-1 levels and increases plasmin activity, which and thus may be more informative about the mechanisms
in turn increases the expression of MMP-9. Local eleva- underlying the gender difference in CHD. More studies on
tions of matrix metalloproteinases in the vulnerable plaque the effects of non-oral estrogens are needed, including
are associated with weakening and rupture of the thin trials with clinical outcomes.
fibrous cap, which is thought to precipitate the thrombotic It is possible, but unproven, that in part the gender
occlusion of the vessel [69]. Thus, the acute effect of HRT difference may be due to adverse effects of androgens in
might be unfavorable in women with vulnerable plaques. men, and more studies of androgens, antiandrogens, and
J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557 555

selective androgen receptor modulators are needed. Males [12] McCrohon JA, Jessup W, Handelsman DJ, Celermajer DS. An-
drogen exposure increases human monocyte adhesion to vascular
and females not only differ in their levels of sex hormones,
endothelium and endothelial cell expression of vascular cell adhe-
the effects of those sex hormones also differ by gender. It sion molecule-1. Circulation 1999;99:2317–2322.
appear likely that more research will reveal that the gender [13] Ayashi AAL, Mathur R, Halushka PV. Testosterone increases
difference has male as well as female components. human platelet thromboxane A2 receptor density and aggregation
In addition to effects of the sex chromosomes, it is responses. Circulation 1995;91:2742–2747.
[14] Rossouw JE. What we still need to learn about hormone replacement
possible that autosomal mutations may influence the risk therapy. Infertil Reprod Med Clin North Am 1999;10:189–209.
for CHD. By definition, autosomal mutations are as [15] Barrett-Connor E, Grady D. Hormone replacement therapy, heart
frequent in males as in females, hence the search should be disease, and other considerations. Annu Rev Public Health
focused on mutations that modify risk and interact with sex 1998;19:55–72.
[16] Grodstein F, Stampfer MJ, Manson JE et al. Postmenopausal
hormones. This research is in its infancy, but a good estrogen and progestin use and the risk of cardiovascular disease.
example of its potential is the interaction between prot- New Engl J Med 1996;335:453–461.
hrombotic mutations and estrogen (in oral contraceptives [17] Sotelo MM, Johnson SR. The effects of hormone replacement
and HRT formulations) that underlie a substantial propor- therapy on coronary heart disease. Endocrinol Metab Clin North Am
1977;26:313–328.
tion of VTE in families with thrombophilia, and also in
[18] The Women’s Health Initiative Study Group. Design of the women’s
unrelated individuals with VTE. The most pressing sci- health initiative clinical trial and observational study. Control Clin
entific challenge of the moment is to find ways of Trials 1998;19:61–109.
identifying the women who are at increased risk for CHD [19] McCrohon JA, Nakhla S, Jessup W, Stanley KK, Celermajer DS.
when they commence HRT. The prothrombotic mutations Estrogen and progesterone reduce lipid accumulation in human
monocyte-derived macrophages. Circulation 1999;100:2319–2325.
are being investigated in this regard, as are several other [20] Winkler UH, Aitkemper R, Kwee B, Helmond FA, Coelingh
markers of coagulation and inflammation. If it becomes Bennink HJT. Effects of tibolone and continuous combined hor-
possible to identify susceptible women, they could be mone replacement therapy on parameters of the clotting cascade:
counseled against HRT, while the non-susceptible women multicenter, double-blind, randomized study. Fertil Steril
2000;74:10–19.
would have a higher chance of experiencing benefit from

Downloaded from by guest on September 12, 2015


[21] van Baal MW, Emeis JJ, van der Mooren MJ et al. Impaired
HRT. procoagulant–anticoagulant balance during hormone replacement
therapy? A randomized, placebo-controlled 12-week study. Thromb
Haemost 2000;83:29–34.
[22] Cushman M, Legault C, Barrett-Connor E et al. Effect of post-
References menopausal hormones on inflammation-sensitive proteins: the post-
menopausal estrogen / progestin interventions (PEPI) study. Circula-
[1] Mendelsohn ME, Karas RH. The protective effects of estrogen on tion 1999;100:717–722.
the cardiovascular system. New Engl J Med 1999;340:1801–1811. [23] Manolio TA, Pearson TA, Wenger NK et al. Cholesterol and heart
[2] Colditz GA, Willett WC, Stampfer MJ et al. Menopause and the risk disease in older persons and women: review of an NHLBI Work-
of coronary disease in women. New Engl J Med 1987;316:1105– shop. Ann Epidemiol 1992;2:161–176.
1110. [24] Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a
[3] Kirkland R, Keenan BS, Probstfield JL et al. Decrease in plasma cardiovascular risk factor. Am J Cardiol 1998;81(4A):7B–12B.
high-density lipoprotein cholesterol levels at puberty in boys with [25] Grady D, Wenger NK, Herrington D et al. Postmenopausal hormone
delayed adolescence. Correlation with plasma testosterone levels. J therapy increases risk for venous thromboembolic disease. The
Am Med Assoc 1987;257:502–507. Heart and Estrogen / progestin Replacement Study. Ann Intern Med
[4] National Heart, Lung, and Blood Institute. In: The lipid research 2000;132:689–696.
clinics population studies data book, vol 1, Bethesda, MD: US [26] Castellsague J, Perez Gutthann S, Garcia Rodriguez LA. Recent
Department of Health and Human Services, National Institutes of epidemiological studies of the association between hormone replace-
Health, 1980. ment therapy and venous thromboembolism. Drug Safety
[5] Gordon DJ, Probstfield JL, Garrison RJ et al. High-density lipopro- 1998;18:117–123.
tein cholesterol and cardiovascular disease. Four prospective Ameri- [27] Hulley S, Grady D, Bush TL et al. Randomized trial of estrogen plus
can studies. Circulation 1989;79:8–15. progestin for secondary prevention of coronary heart disease in
[6] De La Chapelle A. The etiology of maleness in XX men. Hum postmenopausal women. J Am Med Assoc 1998;280:605–613.
Genet 1981;51:105–116. [28] Shlipak MG, Simon JA, Vittinghof E et al. Estrogen and progestin,
[7] Crook D. Androgen therapy in the aging male: assessing the effect lipoprotein (a), and the risk of recurrent coronary heart disease
on heart disease. Aging Male 1999;2:151–156. events after the menopause. J Am Med Assoc 2000;283:1845–1852.
[8] Webb CM, McNeill JG, Hayward CS, de Ziegler D, Collins P. [29] Rosano GMC, Sarrel PM, Poole-Wilson PA, Collins P. Beneficial
Effects of testosterone on coronary vasomotor regulation in men effect of oestrogen on exercise-induced myocardial ischaemia in
with coronary heart disease. J Am Coll Cardiol 1999;83:437–439. women with coronary artery disease. Lancet 1993;342:133–136.
[9] Herman MS, Robinson JTC, McCredie RJ et al. Androgen depriva- [30] Crook D, Cust MP, Gangar KF et al. Comparison of transdermal and
tion is associated with enhanced endothelium-dependent dilatation in and oral estrogen–progestin replacement: effects on serum lipids
adult men. Arterioscler Thromb Vasc Biol 1997;17:2004–2009. and lipoproteins. Am J Obstet Gynecol 1992;166:960–965.
[10] McCredie RJ, McCrohon JA, Turner L et al. Vascular reactivity is [31] Crook D. The metabolic consequences of treating post-menopausal
impaired in genetic females taking high-dose androgens. J Am Coll women with non-oral hormone replacement therapy. Br J Obstet
Cardiol 1998;32:1331–1335. Gynecol 1997;104(suppl 16):4–13.
[11] McCrohon JA, Death AK, Nakhla S et al. Androgen receptor [32] Meschia M, Bruschi F, Soma M et al. Effects of oral and
expression is greater in macrophages from male than from female transdermal hormone replacement therapy on lipoprotein (a) and
donors. Circulation 2000;101:224–226. lipids: a randomized controlled trial. Menopause 1998;5:157–162.
556 J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557

[33] Kroon UB, Silfverstolpe G, Tengborn L. The effects of transdermal replacement therapy in patients with coronary heart disease. J Am
estradiol and oral conjugated estrogens on hemostasis variables. Med Assoc 2000;284:41–43.
Thromb Haemost 1994;71:420–423. [53] Grodstein F, Manson JE. Postmenopausal hormone use and sec-
[34] Scarabin P-Y, Alhenc-Gelas M, Plu-Bureau G et al. Effects of oral ondary prevention of coronary events in the Nurses’ Health Study.
and transdermal estrogen / progesterone concentrations on blood Ann Intern Med 2001;135:1–8.
coagulation and fibrinolysis in postmenopausal women. A random- [54] Alexander KP, Newby KL, Hellkamp AS et al. Initiation of hormone
ized controlled trial. Arterioscler Thromb Vasc Biol 1997;17:3071– replacement therapy after acute myocardial infarction is associated
3078. with more cardiac events during follow-up. J Am Coll Cardiol
[35] Gerhard M, Walsh BW, Tawakol A et al. Estradiol therapy combined 2001;38:1–7.
with progesterone and endothelium-dependent vasodilation in post- [55] Heckbert SR, Kaplan RC, Weiss NS et al. Risk of recurrent coronary
menopausal women. Circulation 1998;98:1158–1163. events in relation to use and recent initiation of postmenopausal
[36] Barrett-Connor E, Goodman-Gruen D. Prospective study of endog- hormone therapy. Arch Intern Med 2001;161:1709–1713.
enous sex hormones and fatal cardiovascular disease in post- [56] Heckbert SR, Weiss NS, Psaty BM. Hormone replacement therapy
menopausal women. Br Med J 1995;311:1193–1196. for secondary prevention of coronary heart disease (letter). J Am
[37] Cauley JA, Gutai JP, Glynn NW et al. Serum estrone concentrations Med Assoc 1999;281:795–796.
and coronary artery disease in postmenopausal women. Arterioscler [57] Seligson U, Lubetsky A. Genetic susceptibility to venous throm-
bosis. New Engl J Med 2001;344:1222–1231.
Thromb 1994;12:14–18.
[58] Vandenbroucke JP, Koster T, Brier E et al. Increased risk of deep
[38] Miserez AR, Keller U. Differences in the phenotypic characteristics
venous thrombosis in oral-contraceptive users who are carriers of
of subjects with familial defective apolipoprotein B-100 and familial
Factor V Leiden mutation. Lancet 1994;344:1453–1457.
hypercholesterolemia. Arterioscl Thromb Vasc Biol 1995;15:1719–
[59] Lowe G, Woodward M, Vessey M et al. Thrombotic variables and
1729.
risk of idiopathic venous thromboembolism in women age 45–64
[39] Wakatsuki A, Okatani Y, Ikenoue N. Effects of combination therapy
years. Thromb Haemost 2000;83:530–535.
with estrogen plus simvastatin on lipoprotein metabolism in post-
[60] Cushman M, Rosendaal FR, Psaty BM et al. Factor V Leiden is not
menopausal women with type IIa hypercholesterolemia. Athero-
a risk factor for arterial vascular disease in the elderly: results from
sclerosis 2000;150:103–111. the Cardiovascular Health Study. Thromb Haemost 1998;79:912–
[40] Lane DA, Grant PJ. Role of hemostatic gene polymorphisms in 915.
venous and arterial disease. Blood 2000;95:1517–1532. [61] Ridker PM, Hennekens CH, Lindpaintner K et al. Mutation in the
[41] Di Castelnuovo A, D’Orazio A, Amore C et al. Genetic modulation

Downloaded from by guest on September 12, 2015


gene encoding for coagulation factor V and the risk of myocardial
of coagulation factor. VII. Plasma levels: contribution of different infarction, stroke, and venous thrombosis in apparently healthy men.
polymorphisms and gender-related effects. Thromb Haemost New Engl J Med 1995;332:912–917.
1998;80:592–597. [62] Rosendaal FR, Siscovick DS, Swartz SM et al. Factor V Leiden
[42] Grohe C, Kahlert S, Lobbert K et al. Cardiac myocytes and (resistance to activated protein C) increases the risk of myocardial
fibroblasts contain functional estrogen receptors. FEBS Lett infarction in young women. Blood 1997;89:2817–2821.
1997;416:107–112. [63] Glueck CJ, Wang P, Fontaine RN et al. Effect of exogenous estrogen
[43] Collins P, Rosano GMC, Sarrell PM et al. Oestradiol-17b attenuates on atherothrombotic vascular disease risk related to the presence or
acetylcholine induced coronary arterial vasoconstriction in women absence of the Factor V Leiden mutation (resistance to activated
but not in men with coronary artery disease. Circulation protein C). Am J Cardiol 1999;84:549–554.
1995;92:24–30. [64] Psaty BM, Smith NL, Lemaitre RN et al. Hormone replacement
[44] McCrohon JA, Walters WAW, Robinson JTC et al. Arterial reactivity therapy, prothrombotic mutations, and the risk of incident myocar-
is enhanced in genetic males taking high dose estrogens. J Am Coll dial infarction in postmenopausal women. J Am Med Assoc
Cardiol 1997;29:1432–1436. 2001;285:906–913.
[45] Morales DE, McGowan KA, Grant DS et al. Estrogen promotes [65] Rosendaal FR, Siscovick DS, Schartz SM et al. A common
angiogenic activity in human umbilical vein endothelial cells in vitro prothrombin variant (20210 G to A) increased the risk of myocardial
and in a murine model. Circulation 1995;91:755–763. infarction in young women. Blood 1997;90:1747–1750.
[46] Losordo DW, Kearney M, Kim EA, Jekanowski J, Isner JM. Variable [66] Glueck CJ, Wang P, Fontaine RN, Sieve-Smith L, Lang JE.
expression of the estrogen receptor in normal and atherosclerotic Interaction of estrogen replacement therapy with the thrombophilic
coronary arteries of premenopausal women. Circulation 20210 G /A prothrombin gene mutation for atherothrombotic vascu-
1994;89:1501–1510. lar disease: a cross-sectional study of 275 hyperlipidemic women.
[47] Herrington DM, Howard T, Florance A et al. Estrogen receptor Metabolism 2001;50:360–365.
polymorphisms associated with enhanced response of HDL to [67] Ridker PM, Hennekens CH, Miletich JP. G20210A mutation in
estrogen replacement therapy in postmenopausal women. Circula- prothrombin gene and risk of myocardial infarction, stroke and
tion 2001;103:1353, abstract. venous thrombosis in a large cohort of US men. Circulation
[48] Clarke S, Kelleher J, Lloyd-Jones H et al. Transdermal hormone 1999;99:999–1004.
replacement therapy for the secondary prevention of coronary artery [68] Cannon R, Yang BK, Ardans J et al. Increased serum matrix
disease in postmenopausal women. Eur Heart J 2000;21:212, metalloproteinase-9 expression in postmenopausal women on es-
Abstract. trogen therapy. J Am Coll Cardiol 1995;35(Suppl 2):303, Abstract.
[49] Viscoli CM, Brass LM, Kernan WN, Sarrel PM, Horwitz R. [69] Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Libby P.
Estrogen after ischemic stroke effect of estrogen replacement on risk Enhanced expression of vascular matrix metalloproteinases induced
of stroke and death in Women’s Estrogen for Stroke Trial (WEST). in vitro by cytokines and in regions of human atherosclerotic
Stroke 2001;32:329, Abstract. lesions. Ann NY Acad Sci 1995;748:501–507.
[50] http: / / www.nhlbi.nih.gov / whi / hrt-en.htm [70] Zanger D, Yang BK, Ardans J et al. Divergent effects of hormone
[51] Hemminki E, McPherson K. Impact of postmenopausal hormone therapy on serum markers of inflammation in postmenopausal
therapy on cardiovascular events and cancer: pooled data from women with coronary artery disease on appropriate medical man-
clinical trials. Br Med J 1997;315:149–153. agement. J Am Coll Cardiol 2000;36:1797–1802.
[52] Wenger NK, Knatterud GL, Canner PL. Early risks of hormone [71] Koh KK, Cardillo C, Bui MN et al. Vascular effects of estrogen and
J.E. Rossouw / Cardiovascular Research 53 (2002) 550 – 557 557

cholesterol-lowering therapies in hypercholesterolemic post- [73] Mosca L, Collins P, Herrington DM et al. Hormone replacement
menopausal women. Circulation 1999;99:354–360. therapy and cardiovascular disease. A statement for healthcare
[72] Rosendaal FR. Thrombosis in the young: epidemiology and risk professionals from the American Heart Association. Circulation
factors. Thromb Haemost 1997;78:1–6. 2001;104:499–503.

Downloaded from by guest on September 12, 2015

Vous aimerez peut-être aussi