Abstract: Cardiovascular disease (CVD) is the most common cause of death in women. Before the Women's Health Initiative (WHI) hormone trials, evidence favored the concept that menopausal hormone treatment (MHT) protects against CVD. WHI studies failed to demonstrate CVD benefit, with worse net outcomes for MHT versus placebo in the population studied. We review evidence regarding the relationship between MHT and CVD with consideration of mechanisms and risk factors for atherogenesis and cardiac events, results of observational case-control and cohort studies, and outcomes of randomized trials. Estrogen effects on CVD risk factors favor delay or amelioration of atherosclerotic plaque development but may increase risk of acute events when at-risk plaque is present. Long-term observational studies have shown ∼40% reductions in risk of myocardial infarction and all-cause mortality. Analyses of data from randomized control trials other than the WHI show a ∼30% cardioprotective effect in recently menopausal women. Review of the literature as well as WHI data suggests that younger and/or more recently menopausal women may have a better risk-benefit ratio than older or remotely menopausal women and that CVD protection may only occur after >5 years; WHI women averaged 63 years of age (12 years postmenopausal) and few were studied for >6 years. Thus, a beneficial effect of long-term MHT on CVD and mortality is still an open question and is likely to remain controversial for the foreseeable future.


Atherosclerotic cardiovascular disease (CVD) remains the leading cause of death in women >50 years of age, accounting for ∼40% of mortality versus about 5% for breast cancer (1, 2). This remains the case despite trends for improvement in CVD incidence rates and reductions in CVD death rates in the population overall and women in particular (3, 4, 5). Risk for new-onset CVD increases after menopause (6), and considerable evidence suggests that the decrease in estrogen experienced by menopausal women contributes to this increase. Nonetheless, after >50 years of research on female sex steroid hormones and atherosclerosis, the questions of whether estrogen deficiency accelerates development of CVD and whether menopausal hormone treatment (MHT) can ameliorate CVD risk remain controversial.

Because the numbers of postmenopausal women in the United States population is large and growing (7), CVD risk assessment and prevention in middle-aged and elderly women is of increasing clinical importance. In this review, we first examine reports of the effects of estrogens and progestogens on factors known or thought to influence development of atherosclerosis and risk of CVD events, then examine the epidemiologic evidence derived from a studies reporting rates of CVD events in menopausal women using and not using MHT, and finally provide a critique of results of recent clinical trials of MHT in which CVD outcomes were primary or secondary endpoints.


Pathogenesis of atherosclerosis

As outlined in a review by Mendelsohn and Karas (8), atherogenesis is a progressive sequence of overlapping stages with characteristic factors influencing each stage. Estrogens and, to a lesser extent, progestogens have been shown to influence factors involved at every stage of the atherogenic process.


Stage 1: Endothelial Injury

The initial step in atherosclerosis involves injury to endothelial cells, most often at sites made vulnerable by disruption of laminar flow (e.g., branch points) or increased blood pressure. Arterial flexibility and vasodilation response may be impaired at such sites owing to reduced production and action of nitric oxide (NO). Lipids, such as oxidized low-density lipoprotein (LDL) cholesterol and lipoprotein (Lp) (a), may also cause endothelial injury.

Flow-mediated vasodilation (FMD) is a reflex relaxation of arterial smooth muscle after a period of arterial occlusion with reduced or absent blood flow. FMD is mediated by endothelial NO production. FMD can be quantified after compression of the brachial artery for several minutes with a blood pressure cuff with the use of Doppler ultrasound or detection systems that respond to blood flow in the digits. Impaired FMD is an indicator of endothelial dysfunction and is associated with increased CVD risk (9). Estrogens have been reported to improve FMD (10, 11, 12, 13) and arterial compliance (14, 15, 16) in a number of studies, whereas progestogens may oppose this effect (17). Higher levels of endogenous E2 are associated with better FMD response (18), and estrogen treatment increases NO synthase activity (19, 20). This may be due to a direct action of estrogens to induce endothelial NO synthase, but estrogens may also act indirectly via effects on asymmetric dimethyl arginine (ADMA), blood pressure, or Lp(a).

ADMA, an amino acid derivative produced by endothelial cell injury (21, 22), is an NO synthase inhibitor. ADMA is increased in hypertension (23) and CVD (24, 25, 26). It is an independent predictor of CVD mortality (27) and worsening of congestive heart failure (28). Estradiol inhibits endothelial cell ADMA production (21, 22, 29). Postmenopausal women have increased ADMA levels (30) and reduced FMD (31), and estrogen treatment decreases ADMA levels (18, 32). Oral estrogen may be more potent than transdermal estrogen in lowering circulating ADMA (33).

Blood pressure (BP) is a major factor in inducing endothelial injury and plays a role in arterial smooth muscle proliferation and thus arterial wall thickening. Oral MHT has been reported to increase BP in younger but not older menopausal women (34) as well as to have neutral effects or even to improve BP (35, 36, 37). In one study, transdermal MHT decreased BP in postmenopausal women without altering angiotensin II, and oral HRT increased angiotensin II but did not affect BP (38). Further studies comparing route of administration showed reduction in BP during MHT with transdermal but not with oral estrogen (39). In a longitudinal study, average systolic BP increased less in MHT users than in nonusers (40). However, data from the largest clinical trial of MHT to date show no overall effect of oral conjugated estrogens with constant low-dose medroxyprogesterone acetate on BP (41) and elevations on the order of 1 mm Hg in women on oral conjugated estrogen alone (42).

Lipoprotein(a) is a lipid fraction that contributes to CVD risk independently from LDL and high-density lipoprotein (HDL) cholesterol levels (43, 44, 45, 46). High Lp(a) levels are associated with reduced FMD, suggesting that Lp(a) mediates endothelial injury (47). In a prospective study, Lp(a) levels were more predictive of CVD events in women than in men (48). MHT has been reported to reduce levels of Lp(a) (49), with a greater decrease with oral than with transdermal estrogen (50). Also, oral MHT appears to reduce CVD events more in women who have high initial Lp(a) levels than in those who do not (44).

Stage 2: Plaque Initiation

The second stage is plaque formation due to lipid deposition in the arterial wall. During this stage, microcrystals of cholesterol and cholesterol esters from circulating Lp particles accumulate at sites of endothelial injury and are phagocytosed by macrophages. These then form clumps of lipid-engorged foam cells in the arterial intima. Factors contributing to this stage include levels of circulating Lps and endothelial adhesion factors that recruit macrophages to transit the endothelium from the arterial lumen. Plaque progression may be reduced by HDL cholesterol via reverse transport of lipid from the arterial wall to the liver (51). As plaques enlarge, increasing numbers of inflammatory cells and fibroblasts are attracted, leading to formation of a fibrous cap over the lipid deposits.

In numerous studies, high total and LDL cholesterol and triglyceride levels and low levels of HDL cholesterol have been associated with increased CVD risk (52, 53). Agents that lower LDL cholesterol have been shown to decrease CVD events in persons with (54, 55) and without (56, 57, 58, 59) prevalent CVD. However, secondary prevention may depend in part on non–lipid-lowering (antiinflammatory and plaque stabilizing) effects of these agents (60, 61, 62, 63). Whether interventions that increase HDL cholesterol are also protective remains an unanswered question, because recent large-scale studies examining this issue have been either negative or equivocal (51, 64, 65).

As reviewed by Tikkanen (66), estrogens lower both total and LDL cholesterol and raise HDL cholesterol levels (67, 68, 69, 70, 71, 72, 73), although the transdermal route may have less effect on HDL cholesterol (72, 74, 75). Estrogen-induced increases in HDL appear to be due mainly to elevation of the cardioprotective HDL-2 subfraction (67, 74).


Stage 3: Inflammation

The third stage of atherogenesis is characterized by increasing inflammation. As plaques reach a critical size, necrosis of foam cells, invasion by inflammatory cells, and neovascularization with invasion and smooth muscle proliferation in the arterial media occur. The end stage of this phase is the “at risk” plaque partially occluding the arterial lumen containing a core of necrotic material and infiltrated with inflammatory cells. Investigations in the past 10 years have provided strong evidence that inflammatory processes are important contributors to atherosclerosis (76, 77, 78).

Inflammatory cells and activated platelets amplify the atherosclerotic process by releasing cytokines, including interleukin (IL) 6 and tumor necrosis factor (TNF) α, which attract and activate additional cells as well as stimulate smooth muscle hyperplasia (79). A variety of circulating cytokines, including IL-6 and C-reactive protein (CRP), have been shown to predict CVD event risk independently from lipids (80, 81, 82). High CRP predicts CVD event risk in both men (83, 84, 85, 86, 87, 88, 89, 90) and women (91, 92, 93). CRP was the best nonlipid biochemical predictor of CVD events in National Health Service data (92) and in the Women's Health Study (94). Some data have suggested that CRP may be a better marker for CVD event risk than for prevalent atherosclerosis (95). IL-6 is the proximate stimulus for CRP production, and in one large prospective study, IL-6, but not CRP, levels independently predicted CVD event incidence, although coronary artery calcium scores were negatively correlated with CRP concentrations (96). Thus, some evidence suggests that IL-6 rather than CRP is the true “villain” in increasing plaque inflammation. However, in another study, neither CRP nor IL-6 was associated with CVD event risk (97). In a prospective nested case-control study of women in the WHI hormone trial, median baseline levels of CRP and IL-6 were significantly higher in women experiencing CVD events (91).

Both IL-6 and CRP increase during menopause (98). In the WHI trials, MHT was associated with significant elevation of median CRP but not IL-6 levels (91). In other studies in which oral estrogen increased CRP levels, no effect was seen on IL-6 (93, 99, 100), nor were changes in IL-6 observed during continuous transdermal MHT (100, 101). Thus, neither oral nor transdermal estrogen appears to affect IL-6. Estrogen effects on CRP depend on route of administration. In the Postmenopausal Estrogen/Progestin Interventions (PEPI) trial (102), oral conjugated equine estrogens (CEE) increased CRP by 85%. In other studies comparing oral and transdermal estrogens, oral treatment increased levels of CRP, whereas transdermal estrogen did not (75, 100, 101, 103). However, in one study of oral E2 combined with cyclic P, no CRP elevation was seen (104). Taken together, these findings suggest that the oral estrogen–induced increase in CRP is an “artifact” of hepatic first-pass effect on protein synthesis. Whether such an elevation of CRP in the absence of increased IL-6 exacerbates CVD risk is unknown.

Prothrombin activator inhibitor (PAI) 1, an acute-phase reactant involved in coagulation, also has been shown to be elevated in patients with prevalent CVD compared with healthy control subjects (105) and to be associated with increased CVD risk (106). In one study, MHT users had lower PAI-1 levels than never users (107). In prospective studies, oral estrogen caused significant decreases in PAI-1 (108, 109) but transdermal estrogen did not (108).

At sites of endothelial injury, E-selectin, ICAM-1, and other adhesion molecules attach circulating leukocytes to the vascular wall. Attached mononuclear cells then transit the endothelium to enter the vascular intima, where they phagocytose lipid particles and transform into foam cells, creating a fatty streak (110). E-Selectin, produced by vascular endothelium in response to CRP, TNF-α, IL-6, and other cytokines (111), interacts with ligands on leukocyte cell membranes (112) to recruit leukocytes to the endothelial surface at sites of inflammation or injury (113, 114). In case-control studies, patients with angiographic evidence of atherosclerosis (115, 116) or myocardial infarction (MI) (117) had significantly elevated E-selectin levels compared with control subjects, whereas in another angiographic study (118) this was not the case. In a prospective study (119), baseline E-selectin levels were higher in subjects who subsequently experienced CVD events and were independently associated with degree of atherosclerosis estimated from carotid artery intima-media thickness. E-Selectin has been reported to be increased in postmenopausal versus cycling women (120), and both oral (102, 104, 121, 122) and transdermal (101, 123) MHT reduced E-selectin levels in prospective studies.

Soluble intercellular adhesion molecule (ICAM) 1, a member of the immunoglobulin gene superfamily, also mediates vascular adhesion and migration of leukocytes. In in vitro studies, CRP and TNF-α up-regulate ICAM-1 expression in coronary endothelial cells (111, 124). In carotid endarterectomy specimens, ICAM-1 mRNA expression was higher in plaques than in normal endothelium and ICAM-1 protein was increased on the surface of high-grade versus low-grade lesions (125). Plasma ICAM-1 levels have been found to be elevated in individuals with versus without angiographic evidence of CVD (116, 126), but this finding was not replicated in another angiographic study (115). Also, higher ICAM-1 levels are found in patients with MI versus control subjects (127). In the Physicians' Health Study, men with ICAM-1 levels in the highest quartile had a risk ratio (RR) of 1.8 (95% confidence interval [CI] 1.1–2.8) for future MI (124). In the Atherosclerosis Risk in Communities (ARIC) study, ICAM-1 levels strongly predicted both MI and stroke; men and women in the highest quartile of ICAM-1 had a hazard ratio of 2.64 for CVD and 5.53 for evidence of coronary atherosclerosis by ultrasound (119). In the WHI, women in the highest quartile of ICAM-1 had an RR of 2.6 (95% CI 1.3–5.1) for CVD events (94). Both oral (93, 104, 128) and transdermal (101, 123) estrogen treatment have been reported to decrease circulating ICAM-1 levels. However, in one RCT, oral E2 did not alter ICAM-1 levels (99).

Stage 4: Plaque Rupture and Thrombosis

In the fourth stage of atherosclerosis, enzymes from inflammatory cells contribute to lysis of the fibrous cap (129, 130), allowing necrotic and inflamed tissue to contact blood. This in turn leads to thrombosis and vascular occlusion. The balance of circulating thrombotic and thrombolytic factors also plays a critical role in this process (131).

Matrix metalloproteinase (MMP) 9 is one of a family of enzymes that break down collagen, allowing remodeling of tissues and migration of cells. In the final stages of plaque evolution, metalloproteinases, notably MMP-9, are released from inflammatory cells (132, 133). The role of MMPs in rupture of the fibrous cap is supported by observations in both experimental (134) and clinical (130, 132) atherosclerosis. High levels of local intralesional MMP expression are characteristic of advanced, but not early, plaques (130). Elevated plasma MMP-9 levels are strongly associated with prevalent symptomatic CVD (135, 136) and are predictive of CVD event risk (132, 137).

Although estrogens appear to be potent inducers of several different MMPs in a variety of cell types (138, 139, 140), MHT has been variously reported to have no effect on (101), to increase significantly (100, 141), and to decrease (93) circulating levels of MMP-9 in women. A likely source of the wide variation in the results of these studies is that variability in plasma MMP-9 levels may reflect variations in both production by vascular inflammatory cells and release of this enzyme from platelets during sample processing (142). The theory that estrogen induction of MMP-9 in at-risk plaques promotes plaque rupture followed by thrombosis and downstream ischemic damage due to luminal obstruction is thus plausible but not fully substantiated.

Effects of oral estrogen on thrombotic, antithrombotic, and fibrinolytic factors favor thrombosis and therefore could elevate risk of a CVD event. This is likely due to first-pass actions of high concentrations of estrogen on the liver after absorption into the portal circulation. These effects are much reduced or absent when estrogen is administered by a nonoral route.

Epidemiologic evidence

Figure 1 shows published results for HRs from 12 prospective cohort and retrospective observational studies examining risk for CVD events (143, 144, 145, 146, 147, 148, 149, 150, 151, 152), stroke (153), and all-cause mortality (154). Eleven of the 12 studies show trends for reduced risk, and in five of those the reductions observed were statistically significant and averaged 40%–50%. There are observational studies not shown in Figure 1 reporting similar reductions in rates of CVD incidence (155, 156, 157, 158) as well as additional reports of reductions in all-cause mortality (157, 159). Moreover, in one study (160) the observed decrease in mortality was significantly greater in current than in past MHT users and in women with longer duration of use, so that current users with >15 years of MHT exposure had a 40% reduction in overall mortality rates.


Figure 1
Mean relative risk (squares) and 95% confidence intervals (black lines) for cardiovascular events in women taking versus not taking menopausal hormone treatment from results of 12 observational and cohort studies published from 1985 to 1996. Only the 1985 study by Wilson et al. shows increased risk, and in five of the 13 the 95% confidence intervals do not overlap 1.0 indicating that the decrease in risk was statistically significant the ≤.05 level.


It is also of note that, whereas women in the Nurses Health Study starting menopausal hormone treatment at or near menopause had significantly reduced CVD event rates (HR 0.66, 95% CI 0.54–0.80, for E alone; HR 0.72, 95% CI 0.56–0.92, for E+P), those who initiated MHT ≥10 years after menopause did not (HR 0.87, 95% CI 0.69–1.10 for E alone; HR 0.90, 95% CI 0.62–1.29 for E+P) (161). This finding is consistent with the “timing hypothesis,” that MHT prevents CVD only if administered during a critical window of opportunity before atherosclerosis is fully established.

Another factor, not widely considered in comparing results of RCTs of MHT, is duration of treatment. If beneficial effects of estrogen on atherogenesis risk factors occur mainly in the early to middle stages of plaque development, then it would follow that any event reductions would have to await the maturation (or lack thereof) of plaques exposed to estrogen treatment in their earlier stages. Some published results of observational studies do indicate that CVD protection may become apparent only after 5–7 years of MHT (162, 163). A case-control study of acute MI (163) found a significant risk reduction in MHT-treated women only after >60 months of treatment (163). Also consistent with a duration effect, postmenopausal women undergoing coronary angiography showed a strong inverse relationship between number of years of MHT and severity of stenosis (164).

In summary, the overwhelming evidence from observational studies comparing many thousands of women choosing versus not choosing to use MHT strongly favors the concept that prolonged use by women starting MHT early in menopause reduces CVD, CVD death, and all-cause mortality rates by ∼40%.

Bias in Epidemiological and Observational Studies

Results of nonrandomized studies need to be interpreted with caution, because a variety of factors may bias results to produce apparent significant differences between treated and untreated groups where none actually exist. These biases include (but are not limited to):

•Selection bias, in which women prescribed MHT are healthier and at lower risk at the outset than those not receiving MHT.
•Prevention bias, in which monitoring and treatment is more intensive in women prescribed MHT, leading to better outcomes in that group.
•Compliance bias, in which patients with better adherence to treatments of all kinds, including MHT, have better outcomes.
•Survivor bias, in which MHT may have been stopped owing to intervening illness and subsequently patients are classified as nonusers when outcomes are assessed.
•Prevalence-incidence bias, in which adverse events or deaths early in the period of MHT use are discounted and the user does not become part of the cohort analyzed.


Well designed RCTs have the advantage of reducing or eliminating the kinds of bias enumerated above and therefore are generally given greater credence than observational studies. However, there have been relatively few RCTs of MHT.


Clinical trials evidence

Before 2002 there were a number of small to medium-size randomized trials of MHT reporting CVD outcomes. Twenty-three of those trials met criteria for inclusion in a meta-analysis by Salpeter et al. (165), who analyzed results according to the ages of the women studied. The authors concluded that in younger women there was significant protection from CVD events with an odds ratio (OR) of 0.68 (95% CI 0.48–0.96), whereas in older women the OR was 1.03 (95% CI 0.91–1.16), such that no effect on CVD risk was apparent. A Bayesian meta-analysis of all-cause mortality looking at randomized trials of MHT of ≥6 months' duration with mean age <60 years that reported at least one death found that in 19 randomized trials, including 16,000 women (83,000 patient-years) with a mean age of 55 years, the relative risk of mortality was 0.73 (95% CI 0.52–0.96). Thus, before 2002, the RCT data, though limited, seemed to be consistent with the data from long-term observational studies, albeit showing somewhat less protective effect, on the order of 30%, perhaps reflecting the effects of the aforementioned biases in the latter studies.

In apparent contrast to the above, results of the large randomized placebo-controlled WHI Estrogen plus Progestin (E+P) (137) and Estrogen Alone (EA) (42) trials found no evidence of CVD protection in women aged 50–79 years and showed increases in breast cancer and thromboembolic disease that, taken together, led to a conclusion that MHT produces net harm. Publication of the WHI findings led millions of menopausal women either to discontinue MHT or to avoid starting it (166, 167).

One potentially critical difference between the WHI and observational studies of MHT is that women enrolled in the WHI were an average of 63 years old, ∼12 years postmenopausal (41, 42, 137, 168). In contrast, enrollees in the observational studies tended to start MHT at or near the menopause, at an average age of 51 years (169).

The idea that differences in age and/or time since menopause may account for differences in cardiovascular outcomes has become known as the “timing hypothesis” (8, 170, 171, 172). The credibility of the timing hypothesis is owed in part to the known effects of estrogen on CVD risk factors. That is, estrogens have effects on lipids, the endothelium, adhesion factors, and inflammatory factors that might be expected to retard early development of plaque. On the other hand, estrogen effects on MMP-9 secretion and, in the case of oral estrogens, thrombotic and thrombolytic factors, could promote plaque rupture and thrombosis, in women harboring mature at-risk plaques, leading to an increase in ischemia and infarction early in the course of treatment.

Subgroup analyses of WHI data provide support for this hypothesis. For example, in the E+P trial a nonsignificant trend toward cardiovascular protection was seen in women who were <10 years postmenopausal, whereas significant excess risk occurred in women >20 years postmenopausal (41). Similarly, in the EA trial (42, 173) there was a trend for cardiovascular protection in women 50–59 years old, but increased risk in women >70 years old. Consistent with the differential effects of estrogen on risk factors for atherogenesis versus plaque rupture and thrombosis, the excess in CVD events in the estrogen-treated groups tended to occur in the first 18–24 months, after which the rates equalized. Similar trends were seen when data from both E+P and EA trials were pooled (174). Finally, in a follow-up examination of women in the EA trial (175), at a mean of 7.4 years of treatment and 1.3 years after the trial was completed, coronary calcification was significantly less in women randomized to CEE versus placebo. This difference was magnified when only subjects compliant with study medication were compared.

A post hoc analysis of data from the WHI E+P trial (176) also supports the concept that emergence of CVD protective effects of MHT requires sufficient duration of treatment. In this analysis, compliant women initiating MHT <10 years after menopause had slightly lower CVD event–free survival rates than women on placebo during the first 5 years. However, after 6 years the placebo and treatment group curves crossed with a nonsignificant, late trend toward better event-free survival in the active MHT group. Specifically, the HR was 1.29 (95% CI 0.52–3.18), a nonsignificant increase in risk, during the first 2 years. In contrast, after 2 years the HR was 0.63 (95% CI 0.27–1.52), suggesting a trend toward decrease in risk. Interestingly, the difference between the ≤2-year and the >2-year HRs was statistically significant (P=.038), consistent with a duration effect.

In an effort to shed further light on the effect of treatment duration, we have performed an independent analysis (177) on published data from the WHI EA trial (42), in which women were followed ∼2 years longer than in the E+P trial. We calculated annual incidence rates for CVD events during years 1–8+ as well as rate ratios and 95% CIs for rates pooled from years 1–6 and >6 with the use of a regression model that accounted for person-years at risk in both groups. Our calculated annual CVD event incidence rates, shown in Figure 2, were slightly greater for CEE- versus placebo-treated women in years 1–2 and 5–6, but declined in the CEE-treated women after year 6. Comparison of the rate ratios for years 1–6 versus 7–8+ showed a statistically significant (P=.003) reduction in CVD risk after >6 years of use of CEE versus placebo.

Figure 2
Plot of the mean numbers of events per 1,000 women per year in the Women's Health Initiative estrogen-alone study comparing conjugated equine estrogens (CEE) with placebo (values taken from Anderson et al., Fig. 3 [42]). There is a small excess in the CEE group in years 1 and 2 and again in years 4 and 5, but values from years 1 to 5 are relatively similar, whereas after year 5, they diverge, falling in the CEE group while remaining fairly stable in the placebo group. Calculated hazard ratio for pooled CVD events (20 in the CEE group versus 51 in the placebo group during a total of 14,633 woman-years) in years 6–8+ is 0.46 (95% confidence interval 0.28–0.78).


Relevant data from another much smaller RCT, the Danish Osteoporosis Prevention Study (DOPS) (178), were published in 2012. That trial was designed to test long-term effects of MHT on osteoporosis in women of average age 50 years at initiation of treatment. Women with an intact uterus were randomized to triphasic E2 and norethisterone acetate or placebo, and hysterectomized women were given 2 mg oral E2 or placebo. Although the treatment phase was discontinued after an average of just over 10 years, data from 1,006 women after an average of 16 years of follow-up showed a reduced incidence of a composite CVD end point consisting of death from any cause, hospital admission for congestive heart failure, and confirmed MI with an HR of 0.61 (95% CI 0.39–0.94; P=.02). There was an apparent excess of CVD deaths (23 of 40) in the control group compared with the MHT group (6 of 27), but no statistical analysis of this difference was provided. Although no differences in estrogen-related adverse events (strokes, all cancers, breast cancer, pulmonary embolus, or thrombophlebitis) were observed, these events were relatively rare (on the order of ≤20 per group) and the study was not powered to detect differences in rare adverse events.


Summary and discussion

Well demonstrated effects of estrogens on a plethora of known and suspected CVD risk factors strongly suggest that MHT should be protective against atherosclerosis if initiated early but is potentially harmful if administered to women who already have mature at-risk plaque. The majority of long-term large observational studies and a number of small RCTs are consistent with this interpretation. Nonetheless, data remain insufficiently definitive to provide decision makers with a high degree of comfort in advising women regarding their risk-benefit ratio for use of MHT.

The present review does not evaluate known or suspected risks of MHT, which include breast cancer, thromboembolic disease, stroke, and cholelithiasis, nor does it take into account established benefits such as relief of menopausal vasomotor symptoms or dyspareunia, reduced risk of osteoporotic fractures, and protection from colon cancer and diabetes. Even taking these risks and benefits as known quantities (which is also a questionable proposition), it remains the case that any valid risk-benefit analysis for MHT depends crucially on whether MHT provides significant CVD protection and/or contributes excess risk and, if so, the magnitude and timing of these effects. An analysis published in 1997 found that when a 40% reduction in CVD event risk is assumed, MHT is likely to be beneficial overall for most women with the exception of women at highest risk for breast cancer (179). This analysis is echoed by a more recent evaluation (180), in which a CVD benefit, at least for younger women, was also assumed.

Contrary to the CVD benefit assumed in the risk-benefit calculations cited, the WHI hormone trials, which represent the largest RCT to date, have been generally interpreted as showing no CVD benefit or even an increase in CVD risk. Limitations of this interpretation include evidence that those conclusions may apply only to older, more distantly postmenopausal women and may be valid only for a relatively short duration (<6 years) of treatment. However, there is no consensus, even among WHI investigators, as to the validity or clinical value of these criticisms.

Thus, in the final analysis, the current state of knowledge regarding the clinical effects of MHT on risk of CVD events, and risk-benefit ratios, remains controversial and in flux, nor is this situation likely to change in the near future. More information is needed regarding effects on both potential risks and benefits of MHT by age and menopausal duration, for oral versus transdermal or subcutanous routes of estrogen administration, and of the various progestational agents used for protection against endometrial hyperplasia. Yet, even with better data on these issues, without a new RCT of the magnitude of the WHI, doubt and controversy will undoubtedly continue.


References

  1. Murphy, S. Deaths: final data for 1998. in: National vital statistics reports. Vol. 48, no. 11. National Center for Health Statistics, Hyattsville, Maryland; 2000
  2. National Heart, Lung, and Blood Institute. Incidence and prevalence: 2006 chart book on cardiovascular and lung diseases. National Institutes of Health, Bethesda, Maryland; 2006
  3. Ma, Y., Hebert, J.R., Balasubramanian, R., Wedick, N.M., Howard, B.V., Rosal, M.C. et al. All-cause, cardiovascular, and cancer mortality rates in postmenopausal white, black, Hispanic, and Asian women with and without diabetes in the United States: the Women's Health Initiative, 1993–2009. Am J Epidemiol. 2013; 178: 1533–1541
  4. Gillum, R.F., Mehari, A., Curry, B., and Obisesan, T.O. Racial and geographic variation in coronary heart disease mortality trends. BMC Public Health. 2012; 12: 410
  5. Hu, F.B., Stampfer, M.J., Manson, J.E., Grodstein, F., Colditz, G.A., Speizer, F.E. et al. Trends in the incidence of coronary heart disease and changes in diet and lifestyle in women. N Engl J Med. 2000;343: 530–537
  6. Pai, J.K. and Manson, J.E. Acceleration of cardiovascular risk during the late menopausal transition.Menopause. 2013; 20: 1–2
  7. Hill, K. The demography of menopause. Maturitas. 1996; 23: 113–127
  8. Mendelsohn, M.E. and Karas, R.H. Molecular and cellular basis of cardiovascular gender differences.Science. 2005; 308: 1583–1587
  9. Jambrik, Z., Venneri, L., Varga, A., Rigo, F., Borges, A., and Picano, E. Peripheral vascular endothelial function testing for the diagnosis of coronary artery disease. Am Heart J. 2004; 148: 684–689
  10. Herrington, D.M., Werbel, B.L., Riley, W.A., Pusser, B.E., and Morgan, T.M. Individual and combined effects of estrogen/progestin therapy and lovastatin on lipids and flow-mediated vasodilation in postmenopausal women with coronary artery disease. J Am Coll Cardiol. 1999; 33: 2030–2037
  11. Sherwood, A., Bower, J.K., McFetridge-Durdle, J., Blumenthal, J.A., Newby, L.K., and Hinderliter, A.L.Age moderates the short-term effects of transdermal 17β-estradiol on endothelium-dependent vascular function in postmenopausal women. Arterioscler Thromb Vasc Biol. 2007; 27: 1782–1787
  12. Stevenson, J.C., Oladipo, A., Manassiev, N., Whitehead, M.I., Guilford, S., and Proudler, A.J.Randomized trial of effect of transdermal continuous combined hormone replacement therapy on cardiovascular risk markers. Br J Haematol. 2004; 124: 802–808
  • Wakatsuki, A., Okatani, Y., Ikenoue, N., and Fukaya, T. Effect of medroxyprogesterone acetate on endothelium-dependent vasodilation in postmenopausal women receiving estrogen. Circulation. 2001;104: 1773–1778
  • Rajkumar, C., Kingwell, B.A., Cameron, J.D., Waddell, T., Mehra, R., Christophidis, N. et al. Hormonal therapy increases arterial compliance in postmenopausal women. J Am Coll Cardiol. 1997; 30: 350–356
  1. Kawecka-Jaszcz, K., Czarnecka, D., Olszanecka, A., Rajzer, M., and Jankowski, P. The effect of hormone replacement therapy on arterial blood pressure and vascular compliance in postmenopausal women with arterial hypertension. J Hum Hypertens. 2002; 16: 509–516
  2. Moreau, K.L., Donato, A.J., Seals, D.R., DeSouza, C.A., and Tanaka, H. Regular exercise, hormone replacement therapy and the age-related decline in carotid arterial compliance in healthy women.Cardiovasc Res. 2003; 57: 861–868
  3. Adams, M.R., Register, T.C., Golden, D.L., Wagner, J.D., and Williams, J.K. Medroxyprogesterone acetate antagonizes inhibitory effects of conjugated equine estrogens on coronary artery atherosclerosis. Arterioscler Thromb Vasc Biol. 1997; 17: 217–221
  4. Li, X.P., Zhou, Y., Zhao, S.P., Gao, M., Zhou, Q.C., and Li, Y.S. Effect of endogenous estrogen on endothelial function in women with coronary heart disease and its mechanism. Clin Chim Acta. 2004;339: 183–188
  5. Mendelsohn, M.E. Genomic and nongenomic effects of estrogen in the vasculature. Am J Cardiol. 2002; 90: 3F–6F
  6. Nuedling, S., Karas, R.H., Mendelsohn, M.E., Katzenellenbogen, J.A., Katzenellenbogen, B.S., Meyer, R. et al. Activation of estrogen receptor beta is a prerequisite for estrogen-dependent upregulation of nitric oxide synthases in neonatal rat cardiac myocytes. FEBS Lett. 2001; 502: 103–108
  7. Dai, Z., Zhu, H.Q., Jiang, D.J., Jiang, J.L., Deng, H.W., and Li, Y.J. 17beta-estradiol preserves endothelial function by reduction of the endogenous nitric oxide synthase inhibitor level. Int J Cardiol. 2004; 96: 223–227
  8. Ishibahshi, T., Obayashi, S., Sakamoto, S., Aso, T., Ishizaka, M., and Azuma, H. Estrogen replacement effectively improves the accelerated intimal hyperplasia following balloon injury of carotid artery in the ovariectomized rats. J Cardiovasc Pharmacol. 2006; 47: 37–45
  9. Wang, D., Strandgaard, S., Iversen, J.S., and Wilcox, C.S. Asymmetric dimethylarginine, oxidative stress and vascular nitric oxide synthase in essential hypertension. Am J Physiol Regul Integr Comp Physiol. 2008; 296: R195–R200
  10. Wang, J., Sim, A.S., Wang, X.L., Salonikas, C., Naidoo, D., and Wilcken, D.E. Relations between plasma asymmetric dimethylarginine (ADMA) and risk factors for coronary disease. Atherosclerosis. 2006; 184: 383–388
  11. Lu, T.M., Ding, Y.A., Charng, M.J., and Lin, S.J. Asymmetrical dimethylarginine: a novel risk factor for coronary artery disease. Clin Cardiol. 2003; 26: 458–464
  12. Ilhan, N., Seckin, D., and Ozbay, Y. Abnormal asymmetric dimethylarginine/nitric oxide balance in patients with documented coronary artery disease: relation to renal function and homocysteine. J Thromb Thrombolysis. 2007; 23: 205–211
  13. Zeller, M., Korandji, C., Guilland, J.C., Sicard, P., Vergely, C., Lorgis, L. et al. Impact of asymmetric dimethylarginine on mortality after acute myocardial infarction. Arterioscler Thromb Vasc Biol. 2008; 28: 954–960
  14. Tang, W.H., Tong, W., Shrestha, K., Wang, Z., Levison, B.S., Delfraino, B. et al. Differential effects of arginine methylation on diastolic dysfunction and disease progression in patients with chronic systolic heart failure. Eur Heart J. 2008; 29: 2506–2513
  15. Monsalve, E., Oviedo, P.J., Garcia-Perez, M.A., Tarin, J.J., Cano, A., and Hermenegildo, C. Estradiol counteracts oxidized LDL-induced asymmetric dimethylarginine production by cultured human endothelial cells. Cardiovasc Res. 2007; 73: 66–72
  16. Verhoeven, M.O., van der Mooren, M.J., Teerlink, T., Verheijen, R.H., Scheffer, P.G., and Kenemans, P.The influence of physiological and surgical menopause on coronary heart disease risk markers.Menopause. 2009; 16: 37–49
  17. Kalantaridou, S.N., Naka, K.K., Papanikolaou, E., Kazakos, N., Kravariti, M., Calis, K.A. et al. Impaired endothelial function in young women with premature ovarian failure: normalization with hormone therapy. J Clin Endocrinol Metab. 2004; 89: 3907–3913
  18. Post, M.S., Verhoeven, M.O., van der Mooren, M.J., Kenemans, P., Stehouwer, C.D., and Teerlink, T.Effect of hormone replacement therapy on plasma levels of the cardiovascular risk factor asymmetric dimethylarginine: a randomized, placebo-controlled 12-week study in healthy early postmenopausal women. J Clin Endocrinol Metab. 2003; 88: 4221–4226
  19. Verhoeven, M.O., Hemelaar, M., van der Mooren, M.J., Kenemans, P., and Teerlink, T. Oral, more than transdermal, oestrogen therapy lowers asymmetric dimethylarginine in healthy postmenopausal women: a randomized, placebo-controlled study. J Intern Med. 2006; 259: 199–208
  20. Steiner, A.Z., Hodis, H.N., Lobo, R.A., Shoupe, D., Xiang, M., and Mack, W.J. Postmenopausal oral estrogen therapy and blood pressure in normotensive and hypertensive subjects: the Estrogen in the Prevention of Atherosclerosis Trial. Menopause. 2005; 12: 728–733
  21. McCubbin, J.A., Helfer, S.G., Switzer, F.S. 3rd, and Price, T.M. Blood pressure control and hormone replacement therapy in postmenopausal women at risk for coronary heart disease. Am Heart J. 2002;143: 711–717
  22. Angerer, P., Stork, S., and von Schacky, C. Influence of 17beta-oestradiol on blood pressure of postmenopausal women at high vascular risk. J Hypertens. 2001; 19: 2135–2142
  23. Harvey, P.J., Wing, L.M., Savage, J., and Molloy, D. The effects of different types and doses of oestrogen replacement therapy on clinic and ambulatory blood pressure and the renin-angiotensin system in normotensive postmenopausal women. J Hypertens. 1999; 17: 405–411
  • Ichikawa, J., Sumino, H., Ichikawa, S., and Ozaki, M. Different effects of transdermal and oral hormone replacement therapy on the renin-angiotensin system, plasma bradykinin level, and blood pressure of normotensive postmenopausal women. Am J Hypertens. 2006; 19: 744–749
  • Ichikawa, A., Sumino, H., Ogawa, T., Ichikawa, S., and Nitta, K. Effects of long-term transdermal hormone replacement therapy on the renin-angiotensin- aldosterone system, plasma bradykinin levels and blood pressure in normotensive postmenopausal women. Geriatr Gerontol Int. 2008; 8: 259–264
  1. Scuteri, A., Bos, A.J., Brant, L.J., Talbot, L., Lakatta, E.G., and Fleg, J.L. Hormone replacement therapy and longitudinal changes in blood pressure in postmenopausal women. Ann Intern Med. 2001;135: 229–238
  2. Manson, J.E., Hsia, J., Johnson, K.C., Rossouw, J.E., Assaf, A.R., Lasser, N.L. et al. Estrogen plus progestin and the risk of coronary heart disease. N Engl J Med. 2003; 349: 523–534
  3. Anderson, G.L., Limacher, M., Assaf, A.R., Bassford, T., Beresford, S.A., Black, H. et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA. 2004; 291: 1701–1712
  4. Futterman, L.G. and Lemberg, L. Lp(a) lipoprotein—an independent risk factor for coronary heart disease after menopause. Am J Crit Care. 2001; 10: 63–67
  5. Shlipak, M.G., Simon, J.A., Vittinghoff, E., Lin, F., Barrett-Connor, E., Knopp, R.H. et al. Estrogen and progestin, lipoprotein(a), and the risk of recurrent coronary heart disease events after menopause.JAMA. 2000; 283: 1845–1852
  6. Candido, A.P., Ferreira, S., Lima, A.A., Nicolato, R.L., Freitas, S.N., Brandao, P. et al. Lipoprotein(a) as a risk factor associated with ischemic heart disease: Ouro Preto Study. Atherosclerosis. 2007; 191: 454–459
  7. Albers, J.J., Slee, A., O'Brien, K.D., Robinson, J.G., Kashyap, M.L., Kwiterovich, P.O. Jr. et al.Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes: the AIM-HIGH trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes). J Am Coll Cardiol. 2013; 62: 1575–1579
  8. Wu, H.D., Berglund, L., Dimayuga, C., Jones, J., Sciacca, R.R., Di Tullio, M.R. et al. High lipoprotein(a) levels and small apolipoprotein(a) sizes are associated with endothelial dysfunction in a multiethnic cohort. J Am Coll Cardiol. 2004; 43: 1828–1833
  9. Lundstam, U., Herlitz, J., Karlsson, T., Linden, T., and Wiklund, O. Serum lipids, lipoprotein(a) level, and apolipoprotein(a) isoforms as prognostic markers in patients with coronary heart disease. J Intern Med. 2002; 251: 111–118
  10. Perry, W. and Wiseman, R.A. Combined oral estradiol valerate-norethisterone treatment over 3 years in postmenopausal women: effect on lipids, coagulation factors, haematology and biochemistry.Maturitas. 2002; 42: 157–164
  11. Seed, M., Sands, R.H., McLaren, M., Kirk, G., and Darko, D. The effect of hormone replacement therapy and route of administration on selected cardiovascular risk factors in post-menopausal women. Fam Pract. 2000; 17: 497–507
  12. Sorci-Thomas, M.G. and Thomas, M.J. Why targeting HDL should work as a therapeutic tool, but has not. J Cardiovasc Pharmacol. 2013; 62: 239–246
  13. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Final report. Circulation. 2002; 106: 3143–3421
  14. Ginsberg, H.N. Is hypertriglyceridemia a risk factor for atherosclerotic cardiovascular disease? A simple question with a complicated answer. Ann Intern Med. 1997; 126: 912–914
  15. Crouse, J.R. 3rd, Byington, R.P., Bond, M.G., Espeland, M.A., Craven, T.E., Sprinkle, J.W. et al.Pravastatin, Lipids, and Atherosclerosis in the Carotid Arteries (PLAC-II). Am J Cardiol. 1995; 75: 455–459
  16. Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. N Engl J Med. 1998; 339: 1349–1357
  17. Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA. 1984; 251: 365–374
  18. Shepherd, J., Cobbe, S.M., Ford, I., Isles, C.G., Lorimer, A.R., MacFarlane, P.W. et al. West of Scotland Coronary Prevention Study Group. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med. 1995; 333: 1301–1307
  19. Downs, J.R., Clearfield, M., Weis, S., Whitney, E., Shapiro, D.R., Beere, P.A. et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study. JAMA. 1998; 279: 1615–1622
  20. Knopp, R.H. Drug treatment of lipid disorders. N Engl J Med. 1999; 341: 498–511
  21. Ridker, P.M., Rifai, N., Pfeffer, M.A., Sacks, F., Braunwald, E., and Cholesterol and Recurrent Events (CARE) Investigators. Long-term effects of pravastatin on plasma concentration of C-reactive protein.Circulation. 1999; 100: 230–235
  22. Bea, F., Blessing, E., Bennett, B., Levitz, M., Wallace, E.P., and Rosenfeld, M.E. Simvastatin promotes atherosclerotic plaque stability in apoE-deficient mice independently of lipid lowering. Arterioscler Thromb Vasc Biol. 2002; 22: 1832–1837
  23. Hwang, J., Hodis, H.N., Hsiai, T.K., Asatryan, L., and Sevanian, A. Role of annexin II in estrogen-induced macrophage matrix metalloproteinase-9 activity: the modulating effect of statins.Atherosclerosis. 2006; 189: 76–82
  24. Kanadasi, M., Cayli, M., Demirtas, M., Inal, T., Demir, M., Koc, M. et al. The effect of early statin treatment on inflammation and cardiac events in acute coronary syndrome patients with low-density lipoprotein cholesterol. Heart Vessels. 2006; 21: 291–297
  • Guyton, J.R., Slee, A.E., Anderson, T., Fleg, J.L., Goldberg, R.B., Kashyap, M.L. et al. Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol. 2013; 62: 1580–1584
  • Hps Thrive Collaborative Group. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J. 2013; 34: 1279–1291
  1. Tikkanen, M.J. Estrogens, progestins and lipid metabolism. Maturitas. 1996; 23: S51–S55
  2. Miller, V.T., Muesing, R.A., LaRosa, J.C., Stoy, D.B., Phillips, E.A., and Stillman, R.J. Effects of conjugated equine estrogen with and without three different progestogens on lipoproteins, high-density lipoprotein subfractions, and apolipoprotein A-I. Obstet Gynecol. 1991; 77: 235–240
  3. Writing Group for the PEPI Trial. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. JAMA. 1995; 273: 199–208
  4. Darling, G.M., Johns, J.A., McCloud, P.I., and Davis, S.R. Estrogen and progestin compared with simvastatin for hypercholesterolemia in postmenopausal women. N Engl J Med. 1997; 337: 595–601
  5. Paganini-Hill, A., Dworsky, R., and Krauss, R.M. Hormone replacement therapy, hormone levels, and lipoprotein cholesterol concentrations in elderly women. Am J Obstet Gynecol. 1996; 174: 897–902
  6. Pickar, J.H., Wild, R.A., Walsh, B., Hirvonen, E., Lobo, R.A., and Menopause Study Group. Effects of different hormone replacement regimens on postmenopausal women with abnormal lipid levels.Climacteric. 1998; 1: 26–32
  7. Sendag, F., Karadadas, N., Ozsener, S., and Bilgin, O. Effects of sequential combined transdermal and oral hormone replacement therapies on serum lipid and lipoproteins in postmenopausal women.Arch Gynecol Obstet. 2002; 266: 38–43
  8. Tugrul, S., Yildirim, G., Pekin, O., Uslu, H., Kutlu, T., and Eren, S. Comparison of two forms of continuous combined hormone replacement therapy with respect to metabolic effects. Arch Gynecol Obstet. 2007; 275: 335–339
  9. Slowinska-Srzednicka, J., Zgliczynski, S., Chotkowska, E., Srzednicki, M., Stopinska-Gluszak, U., Jeske, W. et al. Effects of transdermal 17 beta-oestradiol combined with oral progestogen on lipids and lipoproteins in hypercholesterolaemic postmenopausal women. J Intern Med. 1993; 234: 447–451
  10. Vehkavaara, S., Silveira, A., Hakala-Ala-Pietila, T., Virkamaki, A., Hovatta, O., Hamsten, A. et al. Effects of oral and transdermal estrogen replacement therapy on markers of coagulation, fibrinolysis, inflammation and serum lipids and lipoproteins in postmenopausal women. Thromb Haemost. 2001;85: 619–625
  11. Ridker, P.M., Genest, J., and Libby, P. Risk factors for atherosclerotic disease. in: E. Braunwald, D.P. Zipes, P. Libby (Eds.) Heart Disease: A Textbook of Cardiovascular Medicine. W.B. Saunders,Philadelphia; 2001: 1010–1039
  12. Tracy, R.P. Emerging relationships of inflammation, cardiovascular disease and chronic diseases of aging. Int J Obes. 2003; 27: S29–S34
  13. Willerson, J.T. Systemic and local inflammation in patients with unstable atherosclerotic plaques.Prog Cardiovasc Dis. 2002; 44: 469–478
  14. Prasongsukarn, K., Chaisri, U., Chartburus, P., Wetchabut, K., Benjathummarak, S., Khachansaksumet, V. et al. Phenotypic alterations in human saphenous vein culture induced by tumor necrosis factor-alpha and lipoproteins: a preliminary development of an initial atherosclerotic plaque model. Lipids Health Dis. 2013; 12: 132
  15. Ajani, U.A., Ford, E.S., and McGuire, L.C. Distribution of lifestyle and emerging risk factors by 10-year risk for coronary heart disease. Eur J Cardiovasc Prev Rehabil. 2006; 13: 745–752
  16. St-Pierre, A.C., Cantin, B., Bergeron, J., Pirro, M., Dagenais, G.R., Despres, J.P. et al. Inflammatory markers and long-term risk of ischemic heart disease in men A 13-year follow-up of the Quebec Cardiovascular Study. Atherosclerosis. 2005; 182: 315–321
  17. Cesari, M., Penninx, B.W., Newman, A.B., Kritchevsky, S.B., Nicklas, B.J., Sutton-Tyrrell, K. et al.Inflammatory markers and cardiovascular disease (the Health, Aging and Body Composition [Health ABC] Study). Am J Cardiol. 2003; 92: 522–528
  18. Lagrand, W.K., Visser, C.A., Hermens, W.T., Niessen, H.W., Verheugt, F.W., Wolbink, G.J. et al. C-Reactive protein as a cardiovascular risk factor: more than an epiphenomenon?. Circulation. 1999; 100: 96–102
  19. Bassuk, S.S., Rifai, N., and Ridker, P.M. High-sensitivity C-reactive protein: clinical importance. Curr Probl Cardiol. 2004; 29: 439–493
  20. Roivainen, M., Viik-Kajander, M., Palosuo, T., Toivanen, P., Leinonen, M., Saikku, P. et al. Infections, inflammation, and the risk of coronary heart disease. Circulation. 2000; 101: 252–257
  21. Danesh, J., Whincup, P., Walker, M., Lennon, L., Thomson, A., Appleby, P. et al. Low grade inflammation and coronary heart disease: prospective study and updated meta-analyses. BMJ. 2000;321: 199–204
  22. Ridker, P.M., Cushman, M., Stampfer, M.J., Tracy, R.P., and Hennekens, C.H. Inflammation, aspirin, and the risk of cardiovascular disease in apparently healthy men. N Engl J Med. 1997; 336: 973–979
  23. Koenig, W., Sund, M., Frohlich, M., Fischer, H.G., Lowel, H., Doring, A. et al. C-Reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from the MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Cohort Study, 1984 to 1992. Circulation. 1999; 99: 237–242
  24. Kuller, L.H., Tracy, R.P., Shaten, J., and Meilahn, E.N. Relation of C-reactive protein and coronary heart disease in the MRFIT nested case-control study. Multiple Risk Factor Intervention Trial. Am J Epidemiol. 1996; 144: 537–547
  25. Thompson, S.G., Kienast, J., Pyke, S.D., Haverkate, F., van de Loo, J.C., and European Concerted Action on Thrombosis and Disabilities Angina Pectoris Study Group. Hemostatic factors and the risk of myocardial infarction or sudden death in patients with angina pectoris. N Engl J Med. 1995; 332: 635–641
  26. Pradhan, A.D., Manson, J.E., Rossouw, J.E., Siscovick, D.S., Mouton, C.P., Rifai, N. et al.Inflammatory biomarkers, hormone replacement therapy, and incident coronary heart disease: prospective analysis from the Women's Health Initiative observational study. JAMA. 2002; 288: 980–987
  27. Pai, J.K., Pischon, T., Ma, J., Manson, J.E., Hankinson, S.E., Joshipura, K. et al. Inflammatory markers and the risk of coronary heart disease in men and women. N Engl J Med. 2004; 351: 2599–2610
  28. Hu, P., Greendale, G.A., Palla, S.L., Reboussin, B.A., Herrington, D.M., Barrett-Connor, E. et al. The effects of hormone therapy on the markers of inflammation and endothelial function and plasma matrix metalloproteinase-9 level in postmenopausal women: the Postmenopausal Estrogen Progestin Intervention (PEPI) trial. Atherosclerosis. 2006; 185: 347–352
  29. Ridker, P.M., Hennekens, C.H., Buring, J.E., and Rifai, N. C-Reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000; 342: 836–843
  30. Folsom, A.R., Pankow, J.S., Tracy, R.P., Arnett, D.K., Peacock, J.M., Hong, Y. et al. Association of C-reactive protein with markers of prevalent atherosclerotic disease. Am J Cardiol. 2001; 88: 112–117
  31. Godsland, I.F., Elkeles, R.S., Feher, M.D., Nugara, F., Rubens, M.B., Richmond, W. et al. Coronary calcification, homocysteine, C-reactive protein and the metabolic syndrome in type 2 diabetes: the Prospective Evaluation of Diabetic Ischaemic Heart Disease by Coronary Tomography (PREDICT) Study. Diabet Med. 2006; 23: 1192–1200
  32. Lowe, G.D., Rumley, A., McMahon, A.D., Ford, I., O'Reilly, D.S., and Packard, C.J. Interleukin-6, fibrin D-dimer, and coagulation factors VII and XIIa in prediction of coronary heart disease. Arterioscler Thromb Vasc Biol. 2004; 24: 1529–1534
  33. Hager, K., Machein, U., Krieger, S., Platt, D., Seefried, G., and Bauer, J. Interleukin-6 and selected plasma proteins in healthy persons of different ages. Neurobiol Aging. 1994; 15: 771–772
  34. Manning, P.J., Sutherland, W.H., Allum, A.R., de Jong, S.A., and Jones, S.D. Effect of hormone replacement therapy on inflammation-sensitive proteins in post-menopausal women with type 2 diabetes. Diabet Med. 2002; 19: 847–852
  35. Sumino, H., Ichikawa, S., Kasama, S., Takahashi, T., Kumakura, H., Takayama, Y. et al. Different effects of oral conjugated estrogen and transdermal estradiol on arterial stiffness and vascular inflammatory markers in postmenopausal women. Atherosclerosis. 2006; 189: 436–442
  • Sumino, H., Ichikawa, S., Kasama, S., Kumakura, H., Takayama, Y., Sakamaki, T. et al. Effect of transdermal hormone replacement therapy on carotid artery wall thickness and levels of vascular inflammatory markers in postmenopausal women. Hypertens Res. 2005; 28: 579–584
  1. Cushman, M., Legault, C., Barrett-Connor, E., Stefanick, M.L., Kessler, C., Judd, H.L. et al. Effect of postmenopausal hormones on inflammation-sensitive proteins: the Postmenopausal Estrogen/Progestin Interventions (PEPI) Study. Circulation. 1999; 100: 717–722
  2. Zegura, B., Keber, I., Sebestjen, M., and Koenig, W. Double blind, randomized study of estradiol replacement therapy on markers of inflammation, coagulation and fibrinolysis. Atherosclerosis. 2003;168: 123–129
  3. Stork, S., von Schacky, C., and Angerer, P. The effect of 17beta-estradiol on endothelial and inflammatory markers in postmenopausal women: a randomized, controlled trial. Atherosclerosis. 2002;165: 301–307
  4. Okopien, B., Haberka, M., Cwalina, L., Kowalski, J., Belowski, D., Madej, A. et al. Plasma cytokines as predictors of coronary heart disease. Res Commun Mol Pathol Pharmacol. 2002; 112: 5–15
  5. Marcucci, R., Brogi, D., Sofi, F., Giglioli, C., Valente, S., Liotta, A.A. et al. PAI-1 and homocysteine, but not lipoprotein (a) and thrombophilic polymorphisms, are independently associated with the occurrence of major adverse cardiac events after successful coronary stenting. Heart. 2006; 92: 377–381
  6. Zhang, Y., Howard, B.V., Cowan, L.D., Welty, T.K., Schaefer, C.F., Wild, R.A. et al. Associations of postmenopausal hormone therapy with markers of hemostasis and inflammation and lipid profiles in diabetic and nondiabetic American Indian women: the Strong Heart study. J Womens Health (Larchmt). 2004; 13: 155–163
  7. Scarabin, P.Y., Alhenc-Gelas, M., Plu-Bureau, G., Taisne, P., Agher, R., and Aiach, M. Effects of oral and transdermal estrogen/progesterone regimens on blood coagulation and fibrinolysis in postmenopausal women. A randomized controlled trial. Arterioscler Thromb Vasc Biol. 1997; 17: 3071–3078
  8. Brussaard, H.E., Leuven, J.A., Krans, H.M., and Kluft, C. The effect of 17 beta-oestradiol on variables of coagulation and fibrinolysis in postmenopausal women with type 2 diabetes mellitus.Vascul Pharmacol. 2002; 39: 141–147
  9. Libby, P. Changing concepts of atherogenesis. J Intern Med. 2000; 247: 349–358
  10. Pasceri, V., Willerson, J.T., and Yeh, E.T. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation. 2000; 102: 2165–2168
  11. Price, D.T. and Loscalzo, J. Cellular adhesion molecules and atherogenesis. Am J Med. 1999; 107: 85–97
  • Beekhuizen, H. and van Furth, R. Monocyte adherence to human vascular endothelium. J Leukoc Biol. 1993; 54: 363–378
  1. Bevilacqua, M.P., Stengelin, S., Gimbrone, M.A. Jr., and Seed, B. Endothelial leukocyte adhesion molecule 1: an inducible receptor for neutrophils related to complement regulatory proteins and lectins. Science. 1989; 243: 1160–1165
  2. Semaan, H.B., Gurbel, P.A., Anderson, J.L., Muhlestein, J.B., Carlquist, J.F., Horne, B.D. et al. Soluble VCAM-1 and E-selectin, but not ICAM-1 discriminate endothelial injury in patients with documented coronary artery disease. Cardiology. 2000; 93: 7–10
  3. Oishi, Y., Wakatsuki, T., Nishikado, A., Oki, T., and Ito, S. Circulating adhesion molecules and severity of coronary atherosclerosis. Coron Artery Dis. 2000; 11: 77–81
  4. Elhadd, T.A., Kennedy, G., McLaren, M., Stonebridge, P.A., Shaw, W.J., and Belch, J.J. Elevated levels of soluble E-selectin in diabetic patients with severe symptomatic peripheral arterial occlusive disease requiring angioplasty. A possible role in diabetic vascular disease?. Int Angiol. 2000; 19: 171–175
  5. Porsch-Oezcueruemez, M., Kunz, D., Kloer, H.U., and Luley, C. Evaluation of serum levels of solubilized adhesion molecules and cytokine receptors in coronary heart disease. J Am Coll Cardiol. 1999; 34: 1995–2001
  6. Hwang, S.J., Ballantyne, C.M., Sharrett, A.R., Smith, L.C., Davis, C.E., Gotto, A.M. Jr. et al.Circulating adhesion molecules VCAM-1, ICAM-1, and E-selectin in carotid atherosclerosis and incident coronary heart disease cases: the Atherosclerosis Risk In Communities (ARIC) Study.Circulation. 1997; 96: 4219–4225
  7. Kennedy, G., McLaren, M., Belch, J.J., and Seed, M. Elevated levels of sE-selectin in post-menopausal females are decreased by hormone replacement therapy to levels observed in pre-menopausal females. Thromb Haemost. 1999; 82: 1433–1436
  8. Zanger, D., Yang, B.K., Ardans, J., Waclawiw, M.A., Csako, G., Wahl, L.M. 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
  9. Koh, K.K., Cardillo, C., Bui, M.N., Hathaway, L., Csako, G., Waclawiw, M.A. et al. Vascular effects of estrogen and cholesterol-lowering therapies in hypercholesterolemic postmenopausal women.Circulation. 1999; 99: 354–360
  10. Seljeflot, I., Arnesen, H., Hofstad, A.E., and Os, I. Reduced expression of endothelial cell markers after long-term transdermal hormone replacement therapy in women with coronary artery disease.Thromb Haemost. 2000; 83: 944–948
  11. Ridker, P.M., Hennekens, C.H., Roitman-Johnson, B., Stampfer, M.J., and Allen, J. Plasma concentration of soluble intercellular adhesion molecule 1 and risks of future myocardial infarction in apparently healthy men. Lancet. 1998; 351: 88–92
  • DeGraba, T.J. Expression of inflammatory mediators and adhesion molecules in human atherosclerotic plaque. Neurology. 1997; 49: S15–S19
  • Ikata, J., Wakatsuki, T., Oishi, Y., Oki, T., and Ito, S. Leukocyte counts and concentrations of soluble adhesion molecules as predictors of coronary atherosclerosis. Coron Artery Dis. 2000; 11: 445–449
  • Balbay, Y., Tikiz, H., Baptiste, R.J., Ayaz, S., Sasmaz, H., and Korkmaz, S. Circulating interleukin-1 beta, interleukin-6, tumor necrosis factor-alpha, and soluble ICAM-1 in patients with chronic stable angina and myocardial infarction. Angiology. 2001; 52: 109–114
  1. Koh, K.K., Jin, D.K., Yang, S.H., Lee, S.K., Hwang, H.Y., Kang, M.H. et al. Vascular effects of synthetic or natural progestagen combined with conjugated equine estrogen in healthy postmenopausal women. Circulation. 2001; 103: 1961–1966
  2. Loftus, I.M., Naylor, A.R., Goodall, S., Crowther, M., Jones, L., Bell, P.R. et al. Increased matrix metalloproteinase-9 activity in unstable carotid plaques. A potential role in acute plaque disruption.Stroke. 2000; 31: 40–47
  3. de Nooijer, R., Verkleij, C.J., von der Thusen, J.H., Jukema, J.W., van der Wall, E.E., van Berkel, T.J. et al. Lesional overexpression of matrix metalloproteinase-9 promotes intraplaque hemorrhage in advanced lesions but not at earlier stages of atherogenesis. Arterioscler Thromb Vasc Biol. 2006; 26: 340–346
  4. Corti, R., Farkouh, M.E., and Badimon, J.J. The vulnerable plaque and acute coronary syndromes.The Am J Med. 2002; 113: 668–680
  5. Watanabe, N. and Ikeda, U. Matrix metalloproteinases and atherosclerosis. Curr Atheroscler Rep. 2004; 6: 112–120
  6. Kunz, J. Matrix metalloproteinases and atherogenesis in dependence of age. Gerontology. 2006; 53: 63–73
  7. Gough, P.J., Gomez, I.G., Wille, P.T., and Raines, E.W. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest. 2006; 116: 59–69
  8. Zeng, B., Prasan, A., Fung, K.C., Solanki, V., Bruce, D., Freedman, S.B. et al. Elevated circulating levels of matrix metalloproteinase-9 and -2 in patients with symptomatic coronary artery disease. Intern Med J. 2005; 35: 331–335
  9. Ferroni, P., Basili, S., Martini, F., Cardarello, C.M., Ceci, F., di Franco, M. et al. Serum metalloproteinase 9 levels in patients with coronary artery disease: a novel marker of inflammation. J Investig Med. 2003; 51: 295–300
  10. Trials TWGftW. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA. 2002; 288: 321–333
  11. Zhang, X., Christenson, L.K., and Nothnick, W.B. Regulation of MMP-9 expression and activity in the mouse uterus by estrogen. Mol Reprod Dev. 2007; 74: 321–331
  • Lu, T., Achari, Y., Sciore, P., and Hart, D.A. Estrogen receptor alpha regulates matrix metalloproteinase-13 promoter activity primarily through the AP-1 transcriptional regulatory site.Biochim Biophys Acta. 2006; 1762: 719–731
  1. Suzuki, T. and Sullivan, D.A. Comparative effects of estrogen on matrix metalloproteinases and cytokines in immortalized and primary human corneal epithelial cell cultures. Cornea. 2006; 25: 454–459
  2. Lewandowski, K.C., Komorowski, J., Mikhalidis, D.P., Bienkiewicz, M., Tan, B.K., O'Callaghan, C.J. et al. Effects of hormone replacement therapy type and route of administration on plasma matrix metalloproteinases and their tissue inhibitors in postmenopausal women. J Clin Endocrinol Metab. 2006; 91: 3123–3130
  3. R&D Systems. Human MMP-9 (total) immunoassay. R&D Systems, Minneapolis; 2006
  4. Stampfer, M.J., Willett, W.C., Colditz, G.A., Rosner, B., Speizer, F.E., and Hennekens, C.H. A prospective study of postmenopausal estrogen therapy and coronary heart disease. N Engl J Med. 1985; 313: 1044–1049
  5. Wilson, P.W., Garrison, R.J., and Castelli, W.P. Postmenopausal estrogen use, cigarette smoking, and cardiovascular morbidity in women over 50. The Framingham Study. N Engl J Med. 1985; 313: 1038–1043
  6. Bush, T.L., Barrett-Connor, E., Cowan, L.D., Criqui, M.H., Wallace, R.B., Suchindran, C.M. et al.Cardiovascular mortality and noncontraceptive use of estrogen in women: results from the Lipid Research Clinics Program Follow-up Study. Circulation. 1987; 75: 1102–1109
  7. Petitti, D.B., Perlman, J.A., and Sidney, S. Noncontraceptive estrogens and mortality: long-term follow-up of women in the Walnut Creek Study. Obstet Gynecol. 1987; 70: 289–293
  8. Henderson, B.E., Paganini-Hill, A., and Ross, R.K. Estrogen replacement therapy and protection from acute myocardial infarction. Am J Obstet Gynecol. 1988; 159: 312–317
  9. Wolf, P.H., Madans, J.H., Finucane, F.F., Higgins, M., and Kleinman, J.C. Reduction of cardiovascular disease-related mortality among postmenopausal women who use hormones: evidence from a national cohort. Am J Obstet Gynecol. 1991; 164: 489–494
  10. Falkeborn, M., Persson, I., Adami, H.O., Bergstrom, R., Eaker, E., Lithell, H. et al. The risk of acute myocardial infarction after oestrogen and oestrogen-progestogen replacement. Br J Obstet Gynaecol. 1992; 99: 821–828
  11. Psaty, B.M., Heckbert, S.R., Atkins, D., Lemaitre, R., Koepsell, T.D., Wahl, P.W. et al. The risk of myocardial infarction associated with the combined use of estrogens and progestins in postmenopausal women. Arch Intern Med. 1994; 154: 1333–1339
  12. Folsom, A.R., Mink, P.J., Sellers, T.A., Hong, C.P., Zheng, W., and Potter, J.D. Hormonal replacement therapy and morbidity and mortality in a prospective study of postmenopausal women.Am J Public Health. 1995; 85: 1128–1132
  • Grodstein, F., Stampfer, M.J., Manson, J.E., Colditz, G.A., Willett, W.C., Rosner, B. et al.Postmenopausal estrogen and progestin use and the risk of cardiovascular disease. N Engl J Med. 1996; 335: 453–461
  1. Boysen, G., Nyboe, J., Appleyard, M., Sorensen, P.S., Boas, J., Somnier, F. et al. Stroke incidence and risk factors for stroke in Copenhagen, Denmark. Stroke. 1988; 19: 1345–1353
  2. Criqui, M.H., Suarez, L., Barrett-Connor, E., McPhillips, J., Wingard, D.L., and Garland, C.Postmenopausal estrogen use and mortality. Results from a prospective study in a defined, homogeneous community. Am J Epidemiol. 1988; 128: 606–614
  3. Stampfer, M.J., Colditz, G.A., Willett, W.C., Manson, J.E., Rosner, B., Speizer, F.E. et al.Postmenopausal estrogen therapy and cardiovascular disease. Ten-year follow-up from the nurses' health study. N Engl J Med. 1991; 325: 756–762
  4. Ettinger, B., Friedman, G.D., Bush, T., and Quesenberry, C.P.J. Reduced mortality associated with long-term postmenopausal estrogen therapy. Obstet Gynecol. 1996; 87: 6–12
  5. Grodstein, F., Stampfer, M.J., Colditz, G.A., Willett, W.C., Manson, J.E., Joffe, M. et al.Postmenopausal hormone therapy and mortality. N Engl J Med. 1997; 336: 1769–1775
  6. Grodstein, F., Manson, J.E., Colditz, G.A., Willett, W.C., Speizer, F.E., and Stampfer, M.J. A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease. Ann Intern Med. 2000; 133: 933–941
  7. Bush, T.L., Cowan, L.D., Barrett-Connor, E., Criqui, M.H., Karon, J.M., Wallace, R.B. et al. Estrogen use and all-cause mortality. Preliminary results from the Lipid Research Clinics Program Follow-Up Study. JAMA. 1983; 249: 903–906
  8. Henderson, B.E., Paganini-Hill, A., and Ross, R.K. Decreased mortality in users of estrogen replacement therapy. Arch Intern Med. 1991; 151: 75–78
  9. Grodstein, F., Manson, J.E., and Stampfer, M.J. Hormone therapy and coronary heart disease: The role of time since menopause and age at hormone initiation. J Womens Health (Larchmt). 2006; 15: 35–44
  10. Grodstein, F., Manson, J.E., and Stampfer, M.J. 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
  11. Chilvers, C.E., Knibb, R.C., Armstrong, S.J., Woods, K.L., and Logan, R.F. Post menopausal hormone replacement therapy and risk of acute myocardial infarction—a case control study of women in the East Midlands, UK. Eur Heart J. 2003; 24: 2197–2205
  12. Bairey Merz, C.N., Johnson, B.D., Berga, S.L., Braunstein, G.D., Azziz, R., Yang, Y. et al. Total estrogen time and obstructive coronary disease in women: insights from the NHLBI-sponsored Women's Ischemia Syndrome Evaluation (WISE). J Womens Health (Larchmt). 2009; 18: 1315–1322
  • Salpeter, S.R., Walsh, J.M., Ormiston, T.M., Greyber, E., Buckley, N.S., and Salpeter, E.E. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab. 2006; 8: 538–554
  • Barber, C.A., Margolis, K., Luepker, R.V., and Arnett, D.K. The impact of the Women's Health Initiative on discontinuation of postmenopausal hormone therapy: the Minnesota Heart Survey (2000–2002). J Womens Health. 2004; 13: 975–985
  1. Kelly, J.P., Kaufman, D.W., Rosenberg, L., Kelley, K., Cooper, S.G., and Mitchell, A.A. Use of postmenopausal hormone therapy since the Women's Health Initiative findings. Pharmacoepidemiol Drug Saf. 2005; 14: 837–842
  2. Hsia, J., Criqui, M.H., Rodabough, R.J., Langer, R.D., Resnick, H.E., Phillips, L.S. et al. Estrogen plus progestin and the risk of peripheral arterial disease: the Women's Health Initiative. Circulation. 2004; 109: 620–626
  3. Grodstein, F., Clarkson, T.B., and Manson, J.E. Understanding the divergent data on postmenopausal hormone therapy. N Engl J Med. 2003; 348: 645–650
  4. Naftolin, F., Taylor, H.S., Karas, R., Brinton, E., Newman, I., Clarkson, T.B. et al. The Women's Health Initiative could not have detected cardioprotective effects of starting hormone therapy during the menopausal transition. Fertil Steril. 2004; 81: 1498–1501
  5. Brinton, E.A., Hodis, H.N., Merriam, G.R., Harman, S.M., and Naftolin, F. Can menopausal hormone therapy prevent coronary heart disease?. Trends Endocrinol Metab. 2008; 19: 206–212
  6. Harman, S.M. and Brinton, E.A. Biphasic effects of hormone treatment on risk of cardiovascular disease: resolving the paradox in postmenopausal women. Menopausal Med. 2009; 17: S5–S8
  7. Hsia, J., Langer, R.D., Manson, J.E., Kuller, L., Johnson, K.C., Hendrix, S.L. et al. Conjugated equine estrogens and coronary heart disease: the Women's Health Initiative. Arch Intern Med. 2006; 166: 357–365
  8. Rossouw, J.E., Prentice, R.L., Manson, J.E., Wu, L., Barad, D., Barnabei, V.M. et al. Postmenopausal hormone therapy and risk of cardiovascular disease by age and years since menopause. JAMA. 2007;297: 1465–1477
  9. Manson, J.E., Allison, M.A., Rossouw, J.E., Carr, J.J., Langer, R.D., Hsia, J. et al. Estrogen therapy and coronary-artery calcification. N Engl J Med. 2007; 356: 2591–2602
  10. Toh, S., Hernandez-Diaz, S., Logan, R., Rossouw, J.E., and Hernan, M.A. Coronary heart disease in postmenopausal recipients of estrogen plus progestin therapy: does the increased risk ever disappear? A randomized trial. Ann Intern Med. 2010; 152: 211–217
  11. Harman, S.M., Vittinghoff, E., Brinton, E.A., Budoff, M.J., Cedars, M.I., Lobo, R.A. et al. Timing and duration of menopausal hormone treatment may affect cardiovascular outcomes. Am J Med. 2011; 124: 199–205
  • Schierbeck, L.L., Rejnmark, L., Tofteng, C.L., Stilgren, L., Eiken, P., Mosekilde, L. et al. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ. 2012; 345: e6409
  1. Col, N.F., Eckman, M.H., Karas, R.H., Pauker, S.G., Goldberg, R.J., Ross, E.M. et al. Patient-specific decisions about hormone replacement therapy in postmenopausal women. JAMA. 1997; 277: 1140–1147
  2. Salpeter, S.R., Buckley, N.S., Liu, H., and Salpeter, E.E. The cost-effectiveness of hormone therapy in younger and older postmenopausal women. Am J Med. 2009; 122: 42–52.e2