Menu

An expert resource for medical professionals
Provided FREE as a service to women’s health

The Alliance for
Global Women’s Medicine
A worldwide fellowship of health professionals working together to
promote, advocate for and enhance the Welfare of Women everywhere

An Educational Platform for FIGO

The Global Library of Women’s Medicine
Clinical guidance and resourses

A vast range of expert online resources. A FREE and entirely CHARITABLE site to support women’s healthcare professionals

The Global Academy of Women’s Medicine
Teaching, research and Diplomates Association

This chapter should be cited as follows:
Santoro, N, Stephens, S, Glob. libr. women's med.,
(ISSN: 1756-2228) 2012; DOI 10.3843/GLOWM.10083
This chapter was last updated:
December 2012

Postmenopausal Hormone Therapy

Authors

INTRODUCTION

There is little question that women who suffer from vasomotor symptoms (VMS) or vaginal atrophy can be relieved by use of estrogen. However, over recent decades, the focus of menopausal hormone therapy (MHT) has shifted significantly. In the 1980s and 1990s, based upon observational cohort data that implied that women who used MHT had fewer cases of cardiovascular disease, dementia, and cancer, enthusiasm built for a long-term, preventive strategy, as opposed to a short-term, symptom-based strategy for MHT use. Since the primary findings from the Women’s Health Initiative (WHI) hormone trial1, 2 failed to demonstrate any cardiovascular benefit from estrogen, either given alone or in combination with a progestin (to women with a uterus), the early millennium was marked by a movement away from MHT entirely,3 as the absence of this benefit became a perception of excessive or unacceptable risk. However, over the past 5–6 years, we have been able to observe long-term outcomes of the WHI women (which have chiefly shown a cessation of risk when therapy was stopped)4 and a series of clinical trials demonstrating a relative lack of effectiveness of many alternatives to MHT.5 Thus, MHT is repositioned as an effective treatment for menopausal symptoms, appropriate for short term for most women who feel a need for symptom control. MHT remains an option for women whose menopausal symptoms do not abate over time, especially when the symptoms are severe and alternative methods have not been helpful.6 This chapter discusses the available hormonal preparations for treatment and their practical clinical use, along with the medical evidence supporting them, and the principal risks and benefits to be gained, and ends with a series of questions that remain to be answered about the role of MHT.

 

HORMONAL AGENTS AND ROUTE OF ADMINISTRATION

The dose of estrogen that is effective in maintaining the axial and peripheral bone mass in most women is equivalent to 0.625 mg of daily, oral conjugated estrogens.7, 8 However, there is potential for antiresorptive therapy even at very low doses of estrogen. In a randomized trial of 167 women who were aged 65 or older, Prestwood et al. showed that as little as 0.25 mg oral micronized estradiol daily increased bone mineral density at the hip, spine and whole body.9 Transdermal doses as low as 0.14 µg have also been shown to be effective as an antiresorptive.10 This dose equivalent is frequently effective for relief of VMS and is a useful clinical starting point for treatment. The relative potencies of commercially available estrogens are important to keep in mind when prescribing estrogen. Clinicians should be familiar with these relative potencies, which are based on antiresorptive activity in bone, ability to suppress follicle stimulating hormone (FSH) secretion, or to cause induction of liver proteins. These are not precise, but form reasonable guidelines for flexible dosing. Relative potencies are listed in Table 1.

Table 1 Relative effective estrogen doses11, 12, 13

Estrogen

FSH suppression

Liver protein induction

Bone density

Piperazine estrogen sulfate

1.0 mg

1.25 mg

1.25 mg

Micronized estradiol

1.0 mg

1.0 mg

1.0 mg

Conjugated estrogens

1.0 mg

0.625 mg

0.625 mg

Ethinyl estradiol

5.0 µg

2–10 µg

5.0 µg

Estradiol valerate

1.0 mg

Esterified estrogens

0.625 mg

Transdermal estradiol

50 µg

100 µg

50 µg

Estriol

2 mg

FSH, follicle stimulating hormone.

Esterified estrogens are synthetically prepared from plant precursors and are composed mostly of sodium estrone sulfate with a 6–15% component of sodium equilin sulfate. Estradiol valerate is rapidly hydrolyzed to estradiol, and the pharmacology and effects therefore are comparable at similar dosages.14

The only comparative study of oral estriol concluded that 2 mg of oral estriol was as effective as 0.625 mg of conjugated estrogens in maintaining bone density,15 although there was no suppression of FSH observed with oral estriol, presumably because its undergoes conjugation through its first-pass effect on the liver.16

Transdermal administration of estrogens

Estradiol may be administered via an adherent skin patch, changed weekly or twice weekly, a gel applied directly to the skin of the upper arm, or a spray (‘mist’) applied to the inner forearm. The current generation of patches has the hormone dissolved and distributed throughout the adhesive matrix. Patch estrogen is available in doses ranging from 14 µg to 100 µg.

The effect of steroids on the liver is determined by the type of steroid, the dose, and the route of administration. A theoretical obstacle to the use of transdermal hormone therapy has been concern about the lack of benefit on the lipoprotein profile. However, a theoretical benefit of transdermal estradiol administration is its potential to be less effective at inducing liver production of clotting factors. The concentration of estrogen in the portal system after oral administration is four to five times higher than that in the periphery.17 Furthermore, the estradiol:estrone ratio differs in the portal system, with an unphysiologic, relative excess of estrone to estradiol.

Key differences between oral and non-oral estrogen administration

Short-term studies (6 weeks) document increased catabolism of low-density lipoprotein (LDL) and increased production of apoprotein A-I with oral estrogen, but no effect with transdermal estrogen.18, 19 Furthermore, a 2-year study in Los Angeles with a transdermal dose (100 µg) detected no significant change in high-density lipoprotein (HDL) cholesterol levels.20 On the other hand, data from the UK indicate that the transdermal administration 50 µg of estradiol twice a week is equivalent to 0.625 mg of oral conjugated estrogens, on bone density and lipids over a duration of 3 years.21 It may be that transdermal estradiol causes similar changes in lipoproteins over time and with higher doses, but the bulk of the evidence suggests that it is less effective than oral estrogen in inducing favorable lipoprotein changes (increased HDL and decreased LDL). A potential advantage of transdermal estradiol is its null effect on triglycerides. However, the biological significance of the lipid and lipoprotein changes induced by MHT is unclear. MHT-induced lipoproteins are not strong predictors of subsequent cardiovascular disease and thus the salutary changes in HDL (increased) and LDL (decreased) cholesterol associated with oral estrogen use may not be of great clinical import.

There is suggestive evidence that non-oral administration of estrogen is associated with reduced risk of venous thromboembolism (VTE) and deep venous thrombosis (DVT). A recent meta-analysis of observational cohorts indicates an approximate doubling of the risk of VTE with oral estrogen, but no increased risk when transdermal estradiol is used.22 Interestingly, this doubling of VTE risk is seen only in settings where estrogen is given with progestin. In the WHI, women without a uterus taking estrogen (E) alone did not have a statistically significantly increased risk of pulmonary embolism (HR 1.34, 95% CI 0.87–2.06), whereas women taking estrogen plus progestin (E+P) experienced a doubling of risk compared to women randomized to placebo (HR 2.13, 95% CI 1.39–3.25).1, 2

Non-FDA-approved regimens

Estradiol pellets are available in doses of 25, 50, and 75 mg for subcutaneous administration twice yearly. These formulations are not FDA-approved. The 25 mg pellet provides blood levels in the range of 40–60 pg/ml (150–220 pmol/l), levels which are comparable with those obtained with the standard oral dose.23 However, the dosing can be cumulative, and after several years blood levels can rise to two to three times higher. Significant blood levels of estradiol may persist for up to 2 years after the last insertion. Progestational treatment is necessary, and because of the higher blood levels, a minimal duration of 14 days each month is advised. Estradiol pellets confer no advantages over currently available, FDA-approved treatment regimens, have large potential variations in dosing, and a very long half life. Women receiving pellets should be monitored with periodic blood estradiol levels; levels greater than 200 pg/ml (and, preferably, 100 pg/ml) should be avoided and may well exacerbate harms associated with MHT.

Estrogen creams exist as non-FDA-approved, pharmacy compounded estradiol preparations, many admixed with estrone (Bi-Est) and estriol (Tri-Est). These are often advertised to the lay public as safer alternatives to FDA-approved MHT formulations. The American College of Obstetricians and Gynecologists and the American Society for Reproductive Medicine have recently published a Committee Opinion that evaluates these treatments.24 There are insufficient data to recommend that compounded hormones are safe and effective. Individual pharmacokinetics of these compounds are not known. Thus practitioners are encouraged to avail themselves of the large number of FDA approved formulations of MHT that are demonstrated to be safe and effective and to recommend against the use of pharmacy compounded MHT.

MONITORING ESTROGEN DOSAGE WITH ESTRADIOL BLOOD LEVELS

Monitoring the estradiol blood level in postmenopausal women receiving MHT is not straightforward. There are two primary difficulties. First, the clinical assays available differ considerably in their technique and quality (laboratory and antibody variations). Second, the various commercial products represent a diverse collection of estrogenic compounds, ranging from estradiol to unique equine estrogens. Although the body interconverts various estrogens into estrone and estradiol, this process is not consistent within and between individuals. For example, a highly specific and sensitive assay for estradiol might detect very low levels of estradiol in women receiving 0.625 mg of conjugated equine estrogens (<10 pg/ml); however, most clinical assays would be expected to report a level of 40–100 pg/ml (150–370 pmol/l) in the same women. Blood estradiol levels may be useful in selected patients, such as a patient who requests ever-increasing doses of estradiol for the treatment of symptoms. If estradiol is to be measured, the patient should be taking estradiol only containing MHT, and not a multi-estrogen regimen, such as conjugated equine estrogens. The blood level should be drawn at a time that is consistent with the pharmacokinetics of the compound. For example, a women using a weekly estradiol patch at a dose of 100 µg who fails to note improvement in her symptoms can have her estradiol level measured at mid-week, 3.5 days after patch application (or on the second day after application of a twice-weekly patch). An estradiol level below 50 pg/ml is suggestive of inadequate transdermal absorption of the hormone, and higher doses or alternative methods of hormone delivery should be considered. In one study, 50% of women obtained relief from hot flashes with a mean serum estradiol level of 61 pg/ml.25 This study utilized radioimmunoassay to determine circulating estradiol concentration; current methods based on mass spectrometry or equilibrium dialysis might indicate relief at even lower hormone levels.

Remember that because FSH is regulated by factors other than estrogen (i.e., inhibin), FSH levels cannot be used to reliably monitor estrogen dosage. Menopausal hormone therapy produces only a 10–20% decrease in FSH and luteinizing hormone, and there is great individual variability in the responses.26

 

CONTINUOUS AND SEQUENTIAL REGIMENS

Continuous combined MHT

The continuous/combined method of treatment evolved to improve patient satisfaction with hormones and avoid bleeding and other adverse cyclic symptoms. The continuous activity of progestin allows the use of lower doses that, by virtue of daily availability, inhibit endometrial growth. There is evidence that continuous progestin administration is highly protective against the development of endometrial hyperplasia and cancer.27 This approach involves the continuous daily use of the estrogen–progestin combinations listed in Table 2.

Table 2 Systemic MHT regimens

Combined continuous

Sequential

Estrogen

Estrogen

Non-oral

Non-oral

  50 µg transdermal patch

  50 µg transdermal patch

  0.06% estradiol gel

  0.06% estradiol gel 

  Estradiol mist (1.53 mg/spray)

  Estradiol mist (1.53 mg/spray)

  Vaginal estradiol ring (0.05–0.1mg/day)

  Vaginal estradiol ring (0.05–0.1 mg/day)

Oral

Oral

  0.625 mg conjugated estrogens

  0.625 mg conjugated estrogens

  1.25 mg estrone sulfate

  1.25 mg estrone sulfate

  1.0 mg micronized estradiol

  1.0 mg micronized estradiol

Progestin (daily)

Progestin (12–14 days/month)

 100 mg micronized progesterone

200 mg micronized progesterone

 2.5 mg medroxyprogesterone acetate

5–10 mg medroxyprogesterone acetate

 0.35 mg norethindrone

1 mg norethindrone

 Vaginal progesterone gel 4%,27, 28 2x/week

 Vaginal progesterone gel 4%,28, 29 every other day

Combination E+P

 

Combipatch 50 µg estradiol + 0.140–0.250 mg norethindrone acetate

 

The most common sequential method in the US involves estrogen administration with 0.625 mg of conjugated estrogens or 1.0 mg of micronized estradiol daily. A daily dose of 5–10 mg of medroxyprogesterone acetate (MPA) is added for 14 days of every month. One-year randomized trial data indicate that the 5 mg dose protects the endometrium as well as the 10 mg dose.30 Oral estrogen may also be given with micronized progesterone, 200 mg nightly, for 12 days per month. This regimen has demonstrated satisfactory endometrial safety by biopsy criteria in 114 women followed for 3 years.31 Micronized progesterone has been administered vaginally as well, and appears effective in limited numbers of women.32

Progestin withdrawal bleeding can be expected in 80–90% of women on a sequential regimen.33 The sequential regimen also can cause adverse symptoms related to the relatively higher dose of progestin such as breast tenderness, bloating, fluid retention, and depression. Switching from MPA to norethindrone may relieve these complaints. However, for many women, the preferred regimen is continuous because it is likely to result in amenorrhea.

In the sequential regimen (Table 2), the amount of norethindrone equivalent to 10 mg of MPA is 1.0 mg.34 Norethindrone is currently available in a dose of 0.35 mg in the progestin-only minipill oral contraceptive; thus three of these pills can be taken daily for 10–12 days per month. The lowest effective dose of micronized progesterone and the proper dose for continuous daily administration has not been well established; however, a short-term (6-month) study indicates that 100 mg/day was effective.35 Micronized progesterone can be absorbed somewhat irregularly and metabolized rapidly. Peaks in blood levels of progesterone, and active metabolites are associated with sedation, and for some, a decrease in anxiety or insomnia. Rarely, women will report dysphoric mood or excessive sedation from micronized progesterone. Because of its potential to cause sedation, micronized progesterone is best taken at bedtime. Alternative dosing regimens for progesterone described include vaginal capsule placement and the use of vaginal progesterone gel (Crinone 4–8%). These latter regimens are far less well tested than MPA or micronized progesterone and endometrial surveillance should be conducted to avoid long-term complications of therapy. 

For optimal bone protection, MHT should be combined with adequate calcium intake (best achieved without supplementation, see www.nof.org) and vitamin D (600–4000 IU/day of vitamin D3 is sufficient for most women).36

MHT adherence is generally low.37 One of the best predictors of adherence is the presence of significant menopausal symptoms, and thus a clear perception of benefit from therapy.38 The two most common reasons why women discontinue or do not start hormone treatment are fear of cancer and vaginal bleeding.39 The addition of a progestational agent effectively prevents endometrial cancer. However, persistent vaginal bleeding with the traditional sequential regimen is a disadvantage. To go from 80–90% monthly withdrawal bleeding to no bleeding represents a major accomplishment, and thus, the continuous approach has a significant advantage for most women.

VAGINAL THERAPY

Approximately 25–30% of women report vaginal dryness or dyspareunia at menopause. These symptoms do not improve without treatment. Currently, only estrogen is FDA approved as a treatment for symptoms of vaginal atrophy. Dehydroepiandrosterone (DHEA; Prasterone) is currently under investigation, as is a systemic selective estrogen receptor modulator (SERM),40 osfemipine, for this indication.41, 42

Vaginal estradiol can be given as a cream, tablet or silastic ring. Conjugated estrogens can also be administered as a cream. Available regimens are indicated in Table 3. Current dosing of vaginal estrogen is believed to be low enough that progestin is not required for endometrial protection; however, endometrial surveillance with ultrasound should be considered periodically and any unscheduled vaginal bleeding should be investigated diligently.

Table 3 Vaginal estrogen preparations

Vaginal estrogen preparations

Estradiol silastic ring – 3 month ring (2 mg)

Vaginal estradiol (10 µg 2x/week)

CEE cream (1 applicator 2x/week)

Estradiol cream (1 applicator 2x/week)

MANAGING BLEEDING DURING POSTMENOPAUSAL HORMONE THERAPY

What is a normal bleeding pattern on MHT? It is common for women on a sequential regimen to begin bleeding while in the midst of progestin administration. The timing of withdrawal bleeding in women on a sequential estrogen–progestin program has been suggested as a screening method to determine whether further endometrial surveillance is needed. In women taking a variety of progestins for 12 days each month, bleeding on or before day 10 after the addition of the progestin was more likely to be associated with proliferative endometrium. Bleeding beginning on day 11 or later was associated with secretory endometrium, presumably indicating less need for biopsy.43 Although this concept was felt to constitute a practical test to select women who were more likely to benefit from an endometrial biopsy, subsequent study of a relatively large sample of 413 postmenopausal women demonstrated that the day of bleeding does not predict endometrial safety.44 Late, regular withdrawal bleeding on a sequential program does not give 100% assurance that there is no hyperplasia and perhaps endometrial cancer.

With sequential therapy, approximately 80–90% of women can be expected to have monthly withdrawal bleeding. With continuous, combined estrogen–progestin therapy, 40–60% of patients can be expected to experience breakthrough bleeding during the first 6 months of treatment; however, this percentage decreases to 10–20% after 1 year.31, 33 Although amenorrhea with continuous, combined therapy is a gratifying accomplishment, breakthrough bleeding is relatively frequent, and can become a difficult management problem, as it often causes alarm.

Why call it breakthrough bleeding? The bleeding experienced by women on continuous, combined therapy is similar to that seen with oral contraceptives. It originates from an endometrium dominated by progestational influence; hence, the endometrium usually is atrophic and yields little, if anything, to an exploring biopsy instrument. It is helpful to explain to patients that this bleeding represents tissue breakdown as the endometrium adjusts to its new hormonal stimulation. For most patients, the incidence of breakthrough bleeding with oral contraceptives is greatest in the first few months of treatment and usually disappears thereafter. Indeed, this is the same pattern exhibited by postmenopausal women on continuous, combined therapy, and, therefore, the most effective management strategy is patient education and support. The best approach is to gain time, since bleeding will cease in most patients. This means good educational preparation of the patient beforehand and frequent telephone contact to allay anxiety and encourage persistence.

It is believed that the closer a patient is to having been bleeding (either to her premenopausal state or to having been on a sequential method with withdrawal bleeding), the more likely it is that she will experience breakthrough bleeding if switched to a continuous combined regimen. Some clinicians, therefore, recommend starting with a sequential progestin regimen and converting to a continuous progestin method years later. However, it is just as reasonable to start with a continuous method because women who achieve amenorrhea are highly appreciative. For those who develop bothersome breakthrough bleeding, it is always possible to return to the sequential program.

Options to control intractable breakthrough bleeding for a woman who wishes to remain on hormone therapy include endometrial ablation and progestin intrauterine device (IUD) use. With ablation, concern exists regarding the potential for isolated, residual endometrium to progress to carcinoma without recognition. The progestin IUD releases local progestin (levonorgestrel) in doses sufficient to be effective in suppressing endometrial response and often bleeding. Its chief limitations are the relatively large size of the insertion device, which can be problematic for women who are long past menopause, and the long-term potential for persistent spotting.45, 46 Patients also need to be notified that the levonorgestrel IUD is not FDA approved for this use. Finally, in cases where there are no other options, vaginal hysterectomy provides a definitive alternative.

It is not essential to routinely perform endometrial biopsies before initiating hormone therapy. Endometrial abnormalities in asymptomatic postmenopausal women are rare.47, 48 A reasonable economic moderation would be to limit pretreatment biopsies (using the plastic endometrial suction device in the office) to patients at highest risk for endometrial changes: women with conditions associated with chronic estrogen exposure (obesity, dysfunctional uterine bleeding, anovulation and infertility, hirsutism, high alcohol intake, hepatic disease, and metabolic problems such as diabetes mellitus and hypothyroidism) and women in whom irregular bleeding occurs while on estrogen–progestin therapy. In the absence of abnormal bleeding, trusting in the protective effects of the progestin is reasonable, and routine, periodic biopsies are not necessary when well-tested regimens are used. However, women who elect to be treated with unopposed estrogen, as well as those who are using ‘nonstandard’ progestin regimens (such as quarterly progestin, or preparations such as Crinone, which have not been extensively studied) require endometrial surveillance at least once a year.

Endometrial surveillance can be accomplished by aspiration biopsy (pipelle or its equivalent) or using transvaginal ultrasound with or without fluid contrast. For most situations, a transvaginal ultrasound with a sagittal endometrial thickness measurement less than 5 mm is reassuring.49, 50, 51 In one study of 85 women who underwent both ultrasound and biopsy, it was estimated that 75% of biopsies could be avoided by appropriate interpretation of ultrasound.52, 53 It is important that the ultrasonographer be experienced and provide a clear picture of the midplane of the uterine cavity. If there is uncertainty about the quality of the image, an endometrial biopsy should be performed. Common reasons for an unsatisfactory ultrasound image include the presence of fibroids, and an axial uterus that is in the same plane as the ultrasound probe. If bleeding persists despite a normal ultrasound or negative biopsy, office hysteroscopy or fluid contrast ultrasound can be performed to rule out polyps or submucous myomas. Since curettage and aspiration biopsy alone can miss focal endometrial disease,54 hysteroscopy should be performed in concert with curettage procedures. The lack of a thin endometrium in women on MHT should not be a cause for alarm—just an indication for a biopsy or further evaluation.50, 55, 56 Because endometrial thickness by ultrasonography in patients on a sequential regimen can be affected by day in the treatment cycle, ultrasonography assessment should be obtained toward the end of the progestin phase or at the beginning of the cycle.57 Doppler velocimetry does not improve the accuracy of discriminating between normal and abnormal endometrium.58 Most importantly, a clinician should not be satisfied with “normal” findings on ultrasonography if a patient has persistent bleeding. Biopsies and ultrasound are imperfect monitoring tools and have false negative findings.59 Regimens that do not involve at least 12 days of progestin a month, or in which progestin non-adherence occurs, carry a higher risk of endometrial abnormalities and cancers.60, 61 Continuous use of daily progestin carries a risk lower than that of no hormone use.27 Vigilance on the part of the clinician is needed to detect endometrial cancer at the earliest possible stage, when it is amenable to curative treatment.

SIDE-EFFECTS OF PROGESTINS AND THEIR MANAGEMENT

Many women do not tolerate treatment with progestational hormones. Typical side-effects include breast tenderness, bloating, and even depression. These reactions are significant reasons for discontinuation of MHT. While these reactions are likely rare, and do not appear to be major limitations to the conduct of randomized, clinical trials,62, 63 such women are overrepresented in clinical practice, as they present a clinical challenge. Oral, micronized progesterone given in 200 mg nightly as a sequential agent or 100 mg nightly in a continuous, combined regimen has been shown to result in the most favorable lipid profile of any of the progestins, effectively prevented endometrial hyperplasia over 3 years, and was associated with improved mood and few side-effects in the Postmenopausal Estrogens/Progestins Interventions (PEPI) trial31, 64 and other short-term studies.65 Nonetheless, some patients are sensitive to micronized progesterone or to MPA.

Several clinical strategies can be employed to help patients maintain endometrial protection without incurring bothersome side-effects. The first is to reduce the frequency with which progestin is administered. While it is well documented that monthly or continuous exposure to progestin effectively prevents endometrial hyperplasia, experience with other regimens is limited. In one clinical trial, the administration of MPA every 3 months was associated with longer, heavier menses, more unscheduled bleeding and a 1.5% incidence of hyperplasia at 1 year, whereas in another, overall bleeding was less but the incidence of hyperplasia was approximately 4%.66, 67 In a third study, there was no endometrial hyperplasia encountered by 143 women who completed 2 years of treatment; however, the progestin administered every 3 months was of very high dosage, 20 mg of MPA daily for 14 days.68 The Scandinavian LongCycle Study, a clinical trial scheduled to last 5 years, was cancelled after 3 years because of a 12% incidence of endometrial hyperplasia and four cases of endometrial cancer.69 In this trial, the relatively short duration of progestin administration (10 days) may have been related to its ineffectiveness in preventing hyperplasia; however, norethindrone at a dose of 1 mg was used, which is a relatively potent progestin. Therefore, if a patient chooses this regimen as a means of managing progestin intolerance, more intensive endometrial monitoring is required on an ongoing basis. While ultrasound can be used to monitor in these cases, an annual endometrial biopsy should be strongly considered. Indeed, any program that differs from the standard regimen and is untested by clinical studies of sufficient length and subject numbers, requires periodic surveillance of the endometrium.

Alternative progestins can be tried. In a sequential regimen, the dose of norethindrone is 0.7–1 mg (available in the progestin-only minipill oral contraceptive; each pill contains 0.35 mg of norethindrone and thus two pills must be taken daily for 12–14 days each month). In the continuous, combined regimen, the dose of norethindrone is 0.35 mg daily. Vaginal administration of the micronized progesterone capsule may avoid some of these sedative or other untoward side-effects32.

Progesterone can also be administered in a vaginal gel that allows the delivery of low doses that can effectively protect the endometrium with low systemic levels because of a first-pass effect on the uterus.70 The administration of 90 mg every 2 days produces secretory changes in the endometrium.71 However, no long-term studies of sufficient size are available documenting endometrial safety and metabolic effects. Thus, women using this regimen should undergo periodic endometrial monitoring. It should also be noted that the use of vaginal progesterone gel is substantially more expensive than other, oral progestin preparations. Finally, the contraceptive levonorgestrel-releasing IUD can be considered. It is not FDA-approved for this use, but has the advantage of up to a 10-year duration of effectiveness.

THE ADDITION OF ANDROGENS

After menopause, the circulating level of androstenedione is about half that seen before menopause.72 Most of this postmenopausal androstenedione is derived from the adrenal gland. Testosterone levels do not fall appreciably, however, and in most women, the free androgen index is the same as or greater postmenopause than it was premenopausally.73 Adrenal androgen production, which declines steadily with age from young adulthood, is markedly reduced postmenopause74 and thus there is a supposition that postmenopausal women may be relatively androgen deficient. The addition of testosterone to an estrogen therapy program has been reported to provide no additional beneficial impact on bone or on relief from hot flushes and to correlate with circulating estradiol in one study, while two other studies appeared to show a small incremental benefit of estrogen plus androgen.75, 76, 77

Putative benefits of androgen treatment include improvement in psychological well-being and an increase in sexually motivated behavior. The therapeutic role of androgens for female sexual dysfunction remains unclear. To date, testosterone therapy for menopausal women has focused on the common complaint of low desire. Testosterone may influence multiple aspects of the female sexual response, as evidenced by improvements in desire,78 subjective and objective measures of arousal,78, 79, 80 and increased frequency of orgasm.81, 82, 83 These effects, however, follow the administration of relatively large doses of androgen, and cannot be considered physiologic.84 In a well-designed, placebo-controlled study of 40 naturally menopausal women, lower doses of androgen (but still pharmacologic – 5 mg of methyltestosterone) contributed little to actual sexual behavior, although an increase in sexual fantasies and masturbation could be documented.85

Benefits of androgen therapy must be balanced against known and unknown risks, as well as unwanted effects, in particular, hirsutism and a negative impact on the cholesterol lipoprotein profile. In a short-term study comparing a product with estrogen and a relatively low oral dose of testosterone (1.25 mg of methyltestosterone) to estrogen alone, a negative impact on the lipid profile was apparent within 3 months.86 Over a 2-year period, the administration of estrogen (1.25 mg) combined with 2.5 mg of methyltestosterone produced a significant overall adverse impact on the cholesterol lipoprotein profile.76 In addition, 30% of the patients experienced acne, and 36% developed facial hirsutism. A lower dose of this combination (0.625 mg of esterified estrogens and 1.25 mg of methyltestosterone) also significantly lowers HDL cholesterol.87 Transdermal testosterone treatment of women appears to avoid an adverse impact on the lipoprotein profile and does not appear to result in excess hirsutism.77, 82, 83, 88 Long-term safety data are lacking. Patients require a careful benefit to risk assessment and discussion before undertaking the non-FDA-approved use of androgens.

Androgens do not necessarily protect the endometrium. Addition of a progestin is necessary. It is uncertain (and unstudied) how much aromatization of the administered testosterone increases the estrogen impact and whether this might further increase the risk of endometrial or breast cancer.

SELECTIVE ESTROGEN AGONISTS–ANTAGONISTS

Research in estrogen receptor biology has led to the discovery of mixed estrogen agonists. These pharmaceuticals provide promise for selective actions on specific target tissues. It is now theoretically possible to isolate desired actions from unwanted side-effects. The ideal SERM would have antiresorptive properties on the skeleton, would reduce vasomotor symptoms, treat vaginal dryness/dyspareunia, and have no stimulatory effects on the endometrium, breast or liver. Such a compound is not yet clinically available, but a variety of SERMs are in clinical trials or are FDA approved for use in menopausal women.

Raloxifene is an oral SERM that exerts no proliferative effect on the endometrium but produces favorable responses in bone and lipids.89, 90, 91 Raloxifene’s mixed agonist properties make it an estrogen receptor antagonist at the breast. The National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) indicated that both tamoxifen and raloxifene were similarly effective in preventing invasive breast cancer, with raloxifene being less effective in the prevention of noninvasive disease.92 In an updated analysis with a median 81-month follow-up, tamoxifen was significantly superior for preventing invasive cancer, and both tamoxifen and raloxifene were equally protective against the development of noninvasive disease.93 Compared to tamoxifen, raloxifene caused far less endometrial cancer and hyperplasia, 25% fewer thromboembolic events, and fewer cataracts.

SERMs are currently under development for use in combination with estrogen, and as specific treatment for vulvovaginal atrophy accompanying menopause.

DOES HORMONE THERAPY CAUSE FIBROID TUMORS TO GROW?

Uterine leiomyomas are monoclonal tumors that retain sensitivity to both estrogen and progestin, and, therefore, it is appropriate to be concerned over whether leiomyomas will grow in response to postmenopausal hormone therapy. As assessed by vaginal ultrasonography, the number and size of uterine leiomyomas increased in one study of 50 perimenopausal women being treated with an intramuscular depot form of estrogen–progestin therapy.94 However, this study contained women receiving relatively high-dose hormones in depot form every 6–10 weeks, which may have led to increased levels of hormones. Fibroid size increased a mean of 3–3.5 cm over the 1-year study period. No fibroid growth was observed after 1 year in women with small asymptomatic fibroids administered a daily combination of 0.625 mg of conjugated estrogens and 2.5 mg of MPA in another study; in this same cohort, women treated with transdermal estradiol (50 μg) and 5 mg of MPA daily did have an increase in the size of their fibroids (a response that may reflect the effect of a higher progestin dose).95

In a follow-up study of 14 women taking standard doses of hormone therapy, ultrasonography detected no changes in uterine or myoma volume over a mean follow-up of 19.7 months.96

Although the findings to date are reassuring, it is possible that some women will experience growth of pre-existing uterine fibroids after taking hormone therapy. On the other hand, there is no evidence to suggest that malignant disease of the myometrium is associated with menopausal hormone therapy, as suggested by a case–control study.97

RHEUMATIC DISEASE AND MENOPAUSAL HORMONE THERAPY

Exogenous estrogen, either oral contraceptives or postmenopausal therapy, has demonstrated protection,98 a lack of association,99, 100 and a positive association with rheumatoid arthritis in a variety of observational studies. In one randomized clinical trial of 200 women with rheumatoid arthritis, the use of transdermal estrogen had no net effect on disease, although women with higher circulating estradiol seemed to improve the most, implying benefit of menopausal hormone therapy for rheumatoid arthritis.101

In the Nurses' Health Study (NHS), the use of postmenopausal estrogen was associated with an approximately two-fold increase in systemic lupus erythematosus, an observation based on 30 cases in past and current users of estrogen.102 In a 12-month follow-up study of 60 postmenopausal women with stable systemic lupus erythematosus, 30 of whom were taking menopausal hormone therapy, no adverse effects were demonstrated.103

The approach of using the ‘lowest possible dose for the shortest possible time’ for menopausal hormone therapy is a reasonable path to pursue for symptomatic women with rheumatic diseases. Insufficient data exist to guide the clinician about preferred routes of administration in these settings. The course of the disease should be monitored in relation to the initiation of hormone therapy.

ESTROGEN THERAPY AND OSTEOARTHRITIS

Osteoarthritis is the most common form of arthritis in older people, and its prevalence increases rapidly in women after menopause. The impact of estrogen therapy on osteoarthritis is therefore a logical concern. Increasing severity of osteoarthritis of the knee has been associated with increased bone density as well as increased estrogen levels in middle aged women.104 Women with hip arthritis have been shown to have higher hip bone density in a manner that correlates with severity of disease—suggesting that increased bone density may play a pathogenetic role in the disease.105 On the other hand, hormone use was associated with a halving of the risk of hip osteoarthritis and a lesser severity of the disease by these same investigators.106 Thus, for this common condition, menopausal hormone use seems to have a salutary or neutral effect on the course of the disease.

ESTROGEN THERAPY AND THE ORAL CAVITY

Oral complaints are common among postmenopausal women. The administration of estrogen provides significant relief from oral discomfort, burning, bad taste, and dryness.107 Oral alveolar bone loss (which can lead to loss of teeth) is strongly correlated with osteoporosis, and the salutary effect of estrogen on skeletal bone mass also should be manifested on oral bone.108, 109

In the Leisure World Cohort, tooth loss and edentia were significantly reduced in estrogen users compared with nonusers (with a reduced need for dentures), and this beneficial effect was greater with increasing duration of estrogen use.110 An approximately 25% reduced risk of tooth loss in current users of estrogen has been observed in the Nurses' Health Study.111

Professional singers have used hormone therapy to prevent what they view as unwanted voice changes associated with the menopause. In a 1-year study, objective voice analyses documented a more androgenic change in voice in the early postmenopausal years with a lesser change associated with estrogen treatment, slightly attenuated by the addition of a progestin.112

ESTROGEN THERAPY AND VISION

There is some evidence that estrogen therapy improves visual acuity (or lessens the decrease occurring during the early postmenopausal years), perhaps because of a beneficial effect on lacrimal fluid.113 An increased prevalence of keratoconjunctivitis sicca (dry eyes) in menopausal and postmenopausal women is recognized by ophthalmologists, and estrogen therapy offers the potential for symptomatic relief.114 However, one small study of women with and without dry eye who initiated hormone therapy indicated that symptoms were not alleviated in the 20 women who had dry eye at the outset of the study, and 11 of 20 developed dry eye after hormone therapy initiation.115 There is further evidence that postmenopausal estrogen therapy has an effect that protects against lens opacities.116, 117, 118 Estrogen–progestin treatment also lowers intraocular pressure.119

SHOULD WOMEN OVER 65 BE STARTED ON HORMONE THERAPY?

In general, the risks of MHT outweigh its benefits for long-term use. There is therefore little rationale for initiating MHT in women who are more than 10 years postmenopause. However, a positive impact of estradiol on bone clearly takes place in women over 65. Ultra-low doses of 0.25 mg of micronized estradiol daily were found to result in significant increases in bone mineral density confer osteoprotection. Given the panoply of agents available to treat postmenopausal osteoporosis, there are likely only few women appropriate for this treatment.

HOW LONG SHOULD MENOPAUSAL HORMONE THERAPY BE CONTINUED?

Based on the WHI findings1, 2 indicating a lack of cardiovascular benefit for MHT, MHT is currently prescribed for the treatment of menopausal symptoms. Symptoms considered to have a well established evidence base to be attributable to menopause and likely to be treatable with MHT, include:120

o       Hot flushes and night sweats

o       Vaginal dryness and dyspareunia

o       Poor sleep

o       Adverse mood (depressive symptoms or major depression)

Symptoms for which evidence was found to be fair include:

o       Sexual dysfunction

o       Cognitive impairment

o       Urinary incontinence

Symptoms for which evidence was found to be poor include:

o       Body composition

o       Joint aches and pains

It should be noted that the relationships described above apply to women undergoing natural menopause, and not surgical menopause or antiestrogenic therapy (such as aromatase inhibition for breast cancer treatment or prophylaxis).

ALTERNATIVE TREATMENTS FOR HOT FLUSHES

For women who wish to avoid pharmacologic therapy, behavioral methods may be employed. These are usually effective for women with mild to moderate symptoms. Paced respiration has been shown to be effective in one small randomized trial. Other complementary and alternative treatments have been suggested, but none have demonstrated consistent efficacy.5, 121, 122 At best, these treatments have not demonstrated harm in most of the clinical trials performed to date.

Several non-hormonal treatments are accumulating medical evidence for efficacy that is less than estrogen, but acceptable for many patients. These are gabapentin, venlafaxine and escitalopram (as well as other selective serotonin/norepinephrine reuptake inhibitors). None of these medications are FDA approved for this use. A brief summary is provided in the Table 4.


Table 4 Non-hormonal medication options for vasomotor symptoms

Medication

Dose range

Time to efficacy

Limiting side-effects

Gabapentin

300–900 mg, up to TID

Days

Drowsiness, dizziness

May be best given at bedtime

Venlafaxine

37.5–150 mg

daily

Days

Nausea, GI upset, weight loss, loss of libido, anorgasmia

Escitalopram

10–20 mg

daily

Days

Loss of libido, anorgasmia, possible weight gain

Paroxetine

20–40 mg

daily

Days

Loss of libido, anorgasmia, possible weight gain

Concerns have been raised about the use of certain selective serotonin reuptake inhibitor (SSRI) class drugs in women who are being treated with tamoxifen for breast cancer. These agents can interfere with the metabolism of tamoxifen into more active antiestrogens and may thus reduce the effectiveness of tamoxifen. Paroxetine is the most active in this regard, followed by fluoxetine and venlafaxine. Although the data are inconsistent,123 a recent study cited increased mortality in women taking tamoxifen concurrent with paroxetine.124 Paroxetine should probably be avoided in women with breast cancer who are being treated with tamoxifen, and other SSRI drugs should be used with caution based on these findings.

For women who cannot tolerate these alternatives, there are other medications that have demonstrated mixed efficacy in small trials. Transdermal clonidine, at a 100 μg dose once weekly, has been shown to have efficacy.125, 126 Side-effects include postural hypotension. It may be prudent to begin with a lower dosage or even apply a half-sized patch at first. A modest impact can be expected.

Bellergal (a combination of belladonna alkaloids, ergotamine tartrate, and phenobarbital) treatment is slightly better than a placebo, but it also is a potent sedative.127 Veralipride, a dopamine antagonist that is active in the hypothalamus, is relatively effective in inhibiting flushing at a dose of 100 mg daily.128, 129 Mastodynia and galactorrhea are the major side-effects. MPA (10–20 mg daily) and megestrol acetate (20 mg twice daily) also are effective, but concerns regarding exogenous steroids (especially in patients who have had breast cancer) would apply to progestins as well.130, 131 Methyldopa, in doses of 500–1000 mg/day, is approximately twice as effective as a placebo, suggesting a role for adrenoreceptors in the hot flush mechanism.132 Propranolol and similar agents are ineffective, as is vitamin E.

Tibolone is a steroid, related to the 19-nortestosterone family, available in Europe for the treatment of hot flushes in a dose of 2.5 mg/day.133 It has the additional advantages of lacking any endometrial stimulation, improving bone mineral density and reducing vaginal dryness. It is not available in the US due to concerns about breast cancer risk.

“Natural” therapies

The use of “natural” remedies for menopausal symptoms is widely promoted to women.134, 135 Patients should be asked about their use of “natural” therapies. Botanicals and non-oral, compounded hormones are exempted the same oversight applied by the FDA to pharmaceuticals. Such remedies need not provide proof of efficacy, and FDA is only authorized to intervene if safety is an issue. Due to overall underreporting of adverse events in postmarketed drugs, it is difficult to demonstrate that “natural” compounds are unsafe. Several botanicals are purported to contain estrogen-like compounds, including ginseng, agnus castus, red sage, and black cohosh. The dosage and purity of herbal and compounded preparations are not well controlled. In one FDA survey of 29 compounded hormone preparations, seven failed testing of purity and accuracy of the labelled dose.136 Given the wide array of estrogen and progestin preparations, including many that contain pure 17-beta estradiol and natural progesterone, there is little need to deviate from FDA approved drugs to provide patients with “bioidentical” hormones. The use of non-FDA-approved products without rigorous scientific study should be discouraged.

Phytoestrogens

Phytoestrogens, also called isoflavones, are present in many plants, especially legumes, and bind to the estrogen receptor. Soybeans, a rich source of phytoestrogens, contain genistein, daidzein, and glycitein. These phytoestrogens are characterized by mixed estrogenic and antiestrogenic actions, depending on the target tissue. In addition, the soy phytoestrogens have a greater affinity for the estrogen receptor-beta compared with estrogen receptor-alpha.137 In the monkey, soybean phytoestrogens do not maintain bone density but do have favorable effects on atherosclerosis, vasomotor responses, lipids and lipoproteins, carbohydrate metabolism and fat deposition.138, 139 In the monkey, phytoestrogens did not stimulate proliferation of breast and endometrial cells after 6 months of exposure.140

The daily intake of dietary soy reduces the number of hot flushes in postmenopausal women, although there is significant variability in response, and efficacy appears to be less than that of estrogen.141 In normally cycling women, soy consumption increases menstrual cycle length and produces a reduction in the circulating levels of estradiol.142 The replacement of potent estradiol with target-specific phytoestrogens may be beneficial. However, in postmenopausal women, phytoestrogens may act as weak estrogen agonists, and promote some of the problems associated with estrogen use. Appropriate clinical trials are required to determine how phytoestrogens compare with estrogens, and the efficacy, safety, timing of administration, and dosage. In the meantime, clinicians should use phytoestrogens cautiously or avoid them entirely in women with estrogen-dependent cancers.

Dehydroepiandrosterone

The impressive decline (75–85%) in circulating levels of DHEA that occurs with aging (greater in men than in women)74 has stimulated a search for a beneficial impact of DHEA supplementation. Several well performed, randomized clinical trials provide no support for a benefit of DHEA on cognition, mood, physical performance, and sexual function.143, 144, 145 One study demonstrated a detrimental effect of DHEA supplementation on cognition.146

POTENTIAL PROBLEMS WITH ESTROGEN–PROGESTIN THERAPY

Metabolic

Patients with high-risk factors need special attention when estrogen therapy is being considered. Metabolic contraindications to estrogen therapy include chronically impaired liver function, acute vascular thrombosis (with or without emboli), and neurophthalmologic vascular disease. Estrogens may have adverse effects on some patients with seizure disorders, familial hyperlipidemias (high triglycerides), and migraine headaches.

Pancreatitis and severe hypertriglyceridemia can be precipitated by the administration of oral estrogen to women with elevated triglyceride levels.147 In women with triglyceride levels between 250 and 750 mg/dl, estrogen should be provided with great caution, and a non-oral route of administration is preferred. The triglyceride response is rapid, and a repeat level should be obtained in 2–4 weeks. If increased, hormone therapy must be discontinued. A level greater than 750 mg/dl represents an absolute contraindication to estrogen treatment. Although triglyceride levels in the normal range were not affected by progestins in the PEPI trial, an exaggerated triglyceride response to estrogen might be attenuated by a progestin, especially a progestin of the 19-nortestosterone family, and, therefore, the daily combination method of treatment should be considered for women with elevated triglycerides.

Physiologic and epidemiologic evidence indicates that estrogen use increases the risk of gallbladder disease. In the WHI, both E+P and E alone groups had a higher incidence of overall gall bladder events (for E alone: HR 1.67, 95% CI 1.35–2.06; for E + P: HR 1.59, 95% CI 1.28–1.97).148 Cholecystitis, cholelithiasis and cholecystectomy were all more prevalent in women randomized to active hormone compared to placebo; other biliary disease was not more prevalent. The NHS and other observational cohort studies indicate that oral estrogen therapy may carry a 1.5- to 2.0-fold increased risk of gallbladder disease.149, 150 The risk of cholecystectomy increased with dose and duration of use and was seen to persist for 5 or more years after stopping treatment.

The routine, periodic use of blood chemistries is not cost-effective, and careful monitoring for the appearance of the symptoms and signs of biliary tract disease will suffice. This potential problem may be limited to oral therapy because non-oral routes of estrogen administration do not appear to increase biliary cholesterol saturation (a lithogenic response) in contrast to oral treatment.151

Weight gain

The gain in weight that many middle-aged individuals experience is largely the result of lifestyle, specifically the balance of dietary intake and exercise. Weight gain in women at menopause is not necessarily caused by the hormonal changes associated with the menopause.152 The large Rancho Bernardo prospective cohort study and the randomized PEPI clinical trial indicate that hormone therapy with or without progestin is not associated with increased body weight.153, 154 In the PEPI trial, the hormone-treated groups gained less weight than the placebo group. In the multiethnic cohort of the Study of Women’s Health Across the Nation (SWAN), longitudinal evaluation of weight gain indicated that initiating hormones prior to the final menstrual period was associated with a higher likelihood of weight gain (OR 2.94, 95% CI 1.14–7.58), as was surgical menopause (OR 5.07, 95% CI 2.29–11.02).155 Low sex hormone binding globulin (SHBG) and high testosterone or free androgen index have been associated with metabolic syndrome156 and risk of obesity; however, a more recent analysis of SWAN data indicates that the adiposity precedes the hormone changes.157

It has been proposed that estrogen (with or without progestin) prevents the tendency to increase central body fat with aging. There is limited evidence that this is the case. Studies using newer techniques of body fat compartment measurement may help resolve this issue.

Venous thrombosis

Pharmacologic doses of estrogen (oral contraceptives) are associated with an increased risk of venous thrombosis. The impact of the lower doses administered to postmenopausal women with and without progestin appears to depend on the dose, the exact hormonal preparation given, and the route of administration. Given its relative rarity, attributing venous thromboembolism (VTE) to hormone use can only be done reliably in the context of a large, randomized, controlled trial. In the WHI, the use of estrogen plus progestin resulted in a hazard ratio of 2.13 (95% CI 1.39–3.25) for pulmonary embolism.1 The WHI findings are in overall agreement with the Heart and Estrogen/Progestin Replacement Study (HERS) trial158 and with the NHS), in which the risk of pulmonary embolism was increased twofold in the current hormone users.159 The risk drops to a nonsignificant level within a year of hormone discontinuation.4

In the estrogen only arm of the WHI, the risk of pulmonary embolism was not significantly increased.2 This finding implies that the progestin may be playing a role in pulmonary embolism risk. On the other hand, overall VTEs remained high in the women who took estrogen alone, suggesting that women who have had prior hysterectomies share characteristics that put them at higher VTE risk regardless of whether they are treated with estrogen. In settings where estrogen can be given alone, therefore, concern of a drug-related VTE is minimized, but surveillance should always be encouraged for this potentially life-threatening complication. In a large meta-analysis of postmenopausal hormone users, transdermal estradiol did not increase VTE risk, as opposed to oral estrogen which was associated with an approximate doubling of risk.22

What is the final message for clinicians and patients? The personal VTE risk is low because of the low frequency of this event (about 1/10,000 to 1/15,000). If the relative risk is doubled, this would increase the incidence of venous thromboembolism to about one case per 5000 women per year of hormone use. VTE carries with it a 1% risk of mortality, and the background risk of VTE increases with age. It is prudent to stick to the recommendation of ‘lowest possible dose for the shortest possible time’ to minimize a patient’s exposure to this rare but potentially lethal complication of hormone therapy. On the other hand, clinicians can mitigate risk by favoring the prescription of non-oral estradiol when hormone therapy is needed.

If a patient has a family history or a previous episode of idiopathic thromboembolism, an evaluation to search for an underlying abnormality in the coagulation system is warranted. The following measurements are recommended, and abnormal results require consultation with a hematologist regarding prognosis and prophylactic treatment:

 Factor V Leiden mutation
 Antithrombin III
 Protein C
 Protein S
 Activated protein C resistance ratio
 Activated partial thromboplastin time
 Anticardiolipin antibodies
 Prothrombin gene
 Homocysteine
 Complete blood count

Varicose veins are not a risk factor unless they are extensive, and unlike arterial thrombosis, smoking is not a risk factor for VTE.

If a patient has a congenital predisposition for VTE or if she is otherwise considered to be high risk, alternative methods for symptom control should be considered. In cases in which hormones are the only viable option, the clinician and patient can consider the combination of hormone therapy and chronic anticoagulation, in consultation with a hematologist.

There are no studies of VTE after surgical procedures in postmenopausal hormone users. Recommending appropriate prophylactic treatment in hormone users having major surgery is sensible, especially if other risk factors are present. If possible, patients should discontinue hormone treatment several weeks before surgery.

Endometrial neoplasia

Estrogen normally promotes mitotic growth of the endometrium. Abnormal progression of growth through simple hyperplasia, complex hyperplasia, atypia, and early carcinoma has been associated with unopposed estrogen activity, administered either continuously or in cyclic fashion.30 Only 1 year of treatment with unopposed estrogen (0.625 mg of conjugated estrogens or the equivalent) produces a 20% incidence of hyperplasia, largely simple hyperplasia; in the 3-year PEPI trial, 30% of the women on unopposed estrogen developed adenomatous or atypical hyperplasia.31 About 10% of women with complex hyperplasia progress to frank cancer, and complex hyperplasia is observed to antedate adenocarcinoma in 25–30% of cases. If atypia is present, 20–25% of cases will progress to carcinoma within a year.160

Approximately 40 case–control and cohort studies estimate that the risk of endometrial cancer in women on estrogen therapy (unopposed by a progestational agent) is increased by a factor of about 2–10 times the normal incidence of 1 per 1000 postmenopausal women per year.161, 162 The risk increases with the dose of estrogen and with the duration of exposure (reaching a 10-fold increase with 10–15 years of use, and perhaps an incidence of 1 in 10 with long-term use) and lingers for up to 10 years after estrogen is discontinued. Although most endometrial cancer associated with estrogen use has a low grade and stage, and is associated with better survival (probably because of earlier detection), the overall risk of invasive cancer and death is increased. The risk of endometrial hyperplasia and cancer is not reduced by the administration of unopposed estrogen in a cyclic fashion (a period of time each month without treatment).163

A short-term study (2 years) indicates that one half the usual standard dose of estrogen (in this case, 0.3 mg of esterified estrogens) was not associated with an increased incidence of endometrial hyperplasia compared with a placebo group.164 But we have learned that long-term exposure to low levels of estrogen can induce abnormal endometrial growth, and in our view, lower dose estrogen therapy requires either endometrial assessment annually or the addition of a progestin to the treatment regimen. This is supported by a case–control study from Washington that contained 18 cases and nine controls who had exclusively used only 0.3 mg/day of unopposed conjugated estrogens.165 The use of this half-dose estrogen was associated with an overall fivefold increased risk of endometrial cancer, reaching a relative risk of 9.2 in current users for more than 8 years’ duration. Although limited by small numbers of subjects, the conclusion is logical and consistent with our understanding of the importance of duration of exposure to any increased level of endometrial estrogen stimulation. Possible exceptions include the low dose vaginal estradiol ring (2 mg given over 90 days; Estring), and twice weekly 10 µg vaginal tablets (Vagifem).

Risks of unopposed estrogen can be reduced or eliminated by the addition of a progestational agent to the regimen. Whereas estrogen promotes the growth of endometrium, progestins inhibit mitosis and promote differentiation of the endometrial glands. This counter effect is accomplished via a number of cellular signaling pathways activated by progestin. These include reduction in cellular receptors for estrogen and induction of target cell enzymes that convert estradiol to an excreted metabolite, estrone sulfate.

Reports of the clinical impact of adding progestin in sequence with estrogen include both the reversal of hyperplasia and a diminished incidence of endometrial cancer.166, 167, 168, 169, 170 The protective action of progestational agents requires time to reach its maximal effect. For this reason, the duration of exposure to the progestin each month is critical. Whereas one standard method incorporated the addition of a progestational agent for 10 days a month, most have argued in favor of 12 or 14 days. The minimal requirement is a monthly exposure of at least 10 days’ duration.171, 172 About 2–3% of women per year develop endometrial hyperplasia when the progestin is administered for less than 10 days monthly. Continuous daily progestin is associated with the lowest risk of endometrial hyperplasia.173 Alternative regimens of progestin that have not been adequately studied include vaginal gel (Crinone), vaginal use of micronized progesterone capsules, and less-than-monthly courses of progestin. All alternative regimens should include endometrial surveillance because of the uncertainty of their long-term endometrial safety.

The lowest daily dose of progestin that protects the endometrium has not been established. Currently, the sequential program uses 200 mg of micronized progesterone or 5 or 10 mg of MPA, and the combined daily method uses 100 mg micronized progesterone or 2.5 mg MPA. The dose of norethindrone that is comparable with 100 mg micronized progesterone or 2.5 mg of MPA is 0.25 mg.34 Although lower doses of progestational agents are effective in achieving target tissue responses (such as reducing the nuclear concentration of estrogen receptors), the long-term impact on endometrial histologic features has not been firmly established. The question of dose is an issue of clinical importance, because combined E+P treatment seems to carry additional risks for women that are not seen with E alone regimens.1, 2 Moreover, since the need for progestin is solely to protect the endometrium, minimizing systemic progestin exposure is desirable.

Whereas the protective effect of progestin is considerable and predictable, it is unwise to expect that no patients on estrogen–progestin therapy will develop endometrial cancer. Appropriate monitoring of patients cannot be disregarded. Although routine assessments are not cost-effective, interventions directed by the patient’s clinical presentation are prudent and necessary. For women with no prior exposure to hormone therapy, irregular bleeding or spotting within the first 6 months of treatment is common. Thereafter, the vast majority of women on continuous progestin therapy become amenorrheic and most women on sequential progestin have predictable, monthly bleeding. Changes in an established bleeding pattern are cause for concern and should trigger a clinical response, such as ultrasound evaluation or endometrial sampling.

Ovarian cancer

In the WHI, estrogen alone hormone therapy, but not E+P therapy, was associated with an increased risk of ovarian cancer.174 Other epidemiological data support a duration-dependent association, with a relative risk that ranges from approximately 1.5 to 2.0.175, 176, 177 Other case–control studies have not observed an association,178 and a retrospective analysis of prognosis after surgery indicated no detrimental impact for ovarian cancer in patients who received postmenopausal hormone therapy after diagnosis.179

Cervical cancer

The association between postmenopausal hormone therapy and cancer of the uterine cervix has not been extensively studied. Evidence from one cohort study and one case–control study indicates that the postmenopausal use of estrogen does not increase the risk of cervical cancer.180, 181 Indeed, these studies observed protection against cervical cancer in the estrogen users, but this may reflect detection bias (more examinations and Pap smears in estrogen users). In a follow-up report of 120 women treated for stage I and II cervical cancer, no adverse effects of hormone therapy on survival or recurrence were observed.182

Colorectal cancer

Many cohort and case–control studies report a significantly reduced risk of colorectal cancer in past and current users of MHT,183, 184, 185, 186, 187 however, it is possible that women prescribed MHT may be healthier at baseline. Randomized controlled trials evaluating the effect of MHT on colorectal cancer have not shown as impressive a benefit.

The WHI initially found a significantly lower incidence of colorectal cancer among women in the E+P arm (RR 0.62, 95% CI 0.43–0.89), however, this difference was no longer statistically significant at a mean follow-up of 2.4 years after trial completion (RR 0.75, 95% CI 0.57–1.00).4 Of note, women on E+P who developed colorectal cancer had a greater number of positive lymph nodes (mean ±SD, 3.2 ± 4.1 vs. 0.8 ± 1.7; p = 0.002) and were more likely to have regional or metastatic disease (76.2% vs. 48.5%; p = 0.004) than those on placebo.188

Data from the Heart and Estrogen/Progestin Replacement Study follow-up (HERS II) also did not support a significant benefit of E+P with regards to colorectal cancer incidence after a mean follow-up of 6.8 years (relative HR 0.81, 95% CI 0.46–1.45).189 Five additional double blinded, randomized controlled trials did not find a significant risk reduction in colon cancer with at least 1 year of MHT use.190

Given that the largest randomized trial to address this issue found a risk reduction, it is reasonable to assume that combined E+P hormone therapy reduces new cases of colon cancer; however, the effects are short lived and do not persist after hormones are discontinued. The use of MHT to reduce the risk of colorectal cancer is not recommended.191

Malignant melanoma

The possibility of a relationship between exogenous hormones and cutaneous malignant melanoma has been the subject of many observational studies. Only a few studies have found a statistically significant association between MHT and malignant melanoma.180, 192, 193 Others report slight increases in the risk of malignant melanoma associated with the use of exogenous estrogen, but none demonstrated statistical significance.194, 195, 196 Combining data from 36 observational studies published between 1977 and 2009 and including 5626 cases of melanoma, Gandini et al. did not find an increased risk of melanoma with the use of oral contraceptives (RR 1.04, 95% CI 0.92–1.18) or MHT (RR 1.16, 95% CI 0.93–1.44).197 Post hoc analysis of the WHI randomized placebo controlled trial further supports the lack of an association between MHT and melanoma (HR 0.92, 95% CI 0.61–1.37)198 and a study evaluating recurrence of melanoma with MHT suggests that exogenous hormones are safe even in women with a history of melanoma.199

Breast cancer

Breast cancer is the second most common malignancy affecting women and the second most common cause of cancer death among women in the US. Over 230,000 new invasive cancer cases and 39,500 deaths were estimated to occur in 2011.200 Breast cancer is known to be hormonally influenced, and a large number of studies have been performed to evaluate the effects of exogenous hormones on the incidence of breast cancer. Several of the larger studies evaluating the relationship between postmenopausal hormone use and breast cancer are discussed here.

The British Million Women Study (MWS), California Teachers Study (CTS), Nurses’ Health Study (NHS) and WHI, have all reported an association between breast cancer and MHT in current, but not prior users of MHT. In addition to differences in risk related to current vs. prior use, differences in estrogen-only and E+P preparations have been consistently reported in these studies, with E+P conferring risk and estrogen alone treatments having less or no risk. As such, these regimens are discussed separately.

ESTROGEN ONLY

Several cohort studies have shown an association between estrogen only preparations of MHT and breast cancer. In the NHS, a prospective cohort study of 28,835 postmenopausal women who had undergone a hysterectomy, the risk of breast cancer was increased among current users with duration of use of 20 years or more (RR 1.42, 95% CI 1.13–1.77). Risk increased with increasing duration of use; however, the increase in relative risk did not become statistically significant until 20 years or more duration. Among women with estrogen receptor (ER) and progesterone receptor (PR) positive breast cancer, the risk of breast cancer with MHT increased earlier at 15–15.9 years duration of use (RR 1.48, 95% CI 1.05–2.07).201

Among 56,867 perimenopausal and postmenopausal women in California in the prospective cohort CTS, the risk of breast cancer was significantly increased among current users of estrogen only MHT after a mean follow-up of 9.8 years (adjusted RR 1.33, 95% CI 1.17–1.51). This significantly increased risk was seen even in current users with less than 5 years duration of use (RR 1.23, 95% CI 1.02–1.49) compared to never users. Of women diagnosed with breast cancer, those with current E-only MHT use had an increased risk of ER and PR positive breast cancer.202

The MWS, a prospective cohort study involving 1,084,110 British women, found an increase in breast cancer among current users of estrogen-only MHT (RR 1.30, 95% CI 1.21–1.40). When evaluated by duration of use, this increased risk became statistically significant at 1–4 years of use and increased progressively with longer duration. There was no variation in risk related to type of estrogen used (equine estrogen or estradiol).203

In contrast to these cohort studies, the WHI randomized, placebo-controlled trial found a non-significant reduction in breast cancer among the women on estrogen-only MHT.204 This reduction in risk became statistically significant after a mean follow-up of 10.7 years with HR 0.77 (95% CI 0.62–0.95).205 One possible reason for the discrepancy between the WHI and cohort studies may be the timing of initiation of estrogen therapy in relation to menopausal onset. However, when evaluated by age group, the women in the WHI did not differ in the risk reduction seen with estrogen-only therapy. Although apparently counterintuitive, the WHI findings imply that there is an enduring benefit of estrogen alone on breast cancer risk. It is important to recognize that breast cancer cases were higher overall in the women randomized to the E alone arm of the trial—all of whom had had a prior hysterectomy. It is therefore not appropriate to extrapolate these results to women with a uterus.

Although the WHI did not find an increased risk of breast cancer among women on estrogen-only MHT, the findings of the NHS, CTS and MWS should not be utterly disregarded at this time.

ESTROGEN AND PROGESTIN

The WHI E+P arm was stopped prior to completion in part because of an increased risk of breast cancer among women receiving E+P MHT. This trial enrolled 16,608 postmenopausal women who were monitored with yearly mammograms and clinical breast examinations. After a mean follow-up of 5.6 years (maximum 8.6 years), total breast cancer and invasive breast cancer was increased with E+P use compared to placebo (HR 1.24, 95% CI 1.02–1.50 and HR 1.24, 95% CI 1.01–1.54, respectively). The increase in risk was apparent after 4 years of MHT in women with no prior MHT use prior to enrolment, and after 3 years in women with prior MHT use,206 suggesting a possible cumulative effect of MHT on breast cancer risk. Follow-up of 15,730 women in the E+P arm of the WHI showed that the increase in breast cancer was no longer statistically significant after the intervention phase was stopped, suggesting that risk can be reduced to baseline after cessation of MHT.4 An analysis of both the WHI randomized clinical trial E+P arm and the WHI observational cohort further substantiated the risk reduction after cessation of MHT with the risk decreasing to non-significant levels in less than 2 years.206

The MWS also found an increased incidence of breast cancer among current users of E+P MHT with a RR of 2.00 (95% CI 1.88–2.12). This risk increased with increasing duration of hormone use and did not vary significantly by continuous or sequential use of progestin.203

The risk of breast cancer among current users of E+P MHT was increased with a RR of 1.69 (95% CI 1.50–1.90) among women in the CTS. This increase was persistent regardless of sequential or continuous administration of progestin and increased with increasing duration of use. The greatest breast cancer risk was seen among current users of E+P MHT who took continuous progestin for 15 years or more duration (RR 1.92, 95% CI 1.29–2.86). Current users of E+P MHT also had an increased risk of ER/PR positive breast cancer.202 Of note, past users of E or E+P MHT did not have a significantly increased risk of breast cancer, further supporting the WHI findings of reduction of risk after cessation of MHT.  

Taken together, these studies support an increased risk of breast cancer with current E+P MHT use, with further increased risk with duration of use, and a relatively rapid reduction in risk following completion of MHT.

Timing of therapy in relation to menopause

One critique of the WHI randomized controlled trial is that 90% of women in the estrogen-only arm and 83% of women in the E+P arm were more than 5 years removed from their final menstrual period at the time of randomization for MHT. Various authors have explored the variation in risk related to time of initiation of MHT with respect to onset of menopause. An analysis of the WHI interventional and observational trial data for women who initiated MHT within 5 years of menopause found more incident invasive breast cancer among women who started MHT less than 5 years from onset of menopause in both the estrogen-only and E+P arms. The increase in breast cancer compared to placebo was significant in the E+P arm but only among those with MHT initiation within less than 5 years of menopause regardless of history of MHT use prior to randomization (HR 1.77, 95% CI 1.07–2.93 for women with no prior MHT use and HR 2.06, 95% CI 1.30–3.27 for those with prior MHT use).207

An analysis of the MWS evaluating time from menopause to MHT initiation and risk of breast cancer found similar results. Among current users of estrogen-only MHT, there was no significant increase in breast cancer risk if use began 5 years or more after menopause (RR 1.05, 95% CI 0.89–1.24), however, if use began less than 5 years from menopause, risk was increased compared to non-users (RR 1.43, 95% CI 1.35–1.70). Among current users of E+P formulations, risk was also increased with RR 1.53 (95% CI 1.38–1.70) for onset 5 or more years from menopause and 2.04 (95% CI 1.95–2.14) for onset less than 5 years from menopause.208

In the E3N study, a prospective cohort of 98,995 French women, recent users of E+P MHT who had initiated MHT within 3 years of menopause had a 1.61 relative risk of breast cancer (95% CI 1.43–1.81), while those who initiated more than 3 years from menopause had a 1.35 relative risk (95% CI 1.13–1.63) above never users.209

All of these studies suggest that early initiation of MHT may be detrimental with regards to breast cancer risk. Estrogen is a known stimulus for breast cancer growth and anti-estrogenic therapies are now standard of care for ER positive tumors. However, the relationship between estrogen and breast cancer is complex. In addition to its growth-promoting effects, estrogen is an apoptotic trigger for breast cancer cells exhibiting antihormone resistance.210 It is theorized that the response of breast cancer cells to estrogen changes in response to the hormonal environment in which they are growing. In a low-estrogen environment (i.e. postmenopausal or antihormone treated), tumor cells become susceptible to estrogen-induced apoptosis. This and other properties of estrogen may explain the reduction in breast cancer seen in postmenopausal women on estrogen-only MHT in the WHI interventional trial.

Tumor characteristics/prognosis of women who develop breast cancer while taking MHT

A few observational studies have found more favorable breast cancer histology (lobular and tubular tumors)211, 212, 213 among MHT users, while others have observed that MHT-associated breast cancers are smaller and have fewer positive lymph nodes214 suggesting more favorable prognosis. However, this is not consistent with the WHI findings.

A secondary analysis of the E+P arm of the WHI found no difference in type of breast cancer, histology or grade between the MHT and placebo groups. However, in women randomized to MHT, the invasive breast cancers were larger, more likely to be node positive, and of a more advanced stage among than those in women on placebo. There was no difference in the number of ER/PR positive or ER/PR negative breast cancers between the E+P and placebo groups. There was also a higher percentage of women with an abnormal ultrasound requiring medical evaluation among women randomized to E+P compared to those receiving placebo after only 1 year of MHT use.215 These findings do not support the oft-touted notion that breast cancer contracted while taking MHT is a less aggressive disease. E+P use may also impair the ability of mammographic detection of malignancy, resulting in later cancer stage at diagnosis.

MHT use in women with a history of breast cancer

Several observational studies have not identified an increased risk of breast cancer recurrence among current MHT users with a history of prior breast cancer.216, 217 However, given the results discussed above, concern regarding the safety of MHT in breast cancer survivors should be maintained. Only two randomized studies have been published regarding the risk of breast cancer recurrence with MHT: the Hormonal Replacement Therapy after Breast Cancer-Is It Safe? (HABITS) trial and the Stockholm randomized trial.

Both the HABITS trial and the Stockholm trial are Swedish studies that started recruitment in 1997. Because of slow recruitment for both studies, a joint Data Monitoring and Safety Committee (DMSC) was formed in 2002 with plans for joint interim safety analyses and final data analysis. An interim safety analysis of the pooled data in October of 2003 found the combined hazard ratio (HR) for recurrence of breast cancer with MHT to be unacceptably high at 1.8 (95% CI 1.03–3.1). There was significant heterogeneity between the studies, however, with the HABITS study indicating an HR 3.3 (95% CI 1.5–7.4) and the Stockholm trial a non-significant HR of 0.82 (95% CI 0.35–1.9).218 Subsequently, both studies were terminated prior to completion. Because of the conflicting findings of the two studies, they are more closely discussed here.

The HABITS trial was a prospective, randomized, open-label non-inferiority trial in which 447 women with a history of early stage breast cancer were randomized to either MHT or to best symptomatic treatment without hormone. All women had stage 0–2 disease with less than four positive axillary lymph nodes. The MHT regimen was at the discretion of their provider, but most women with an intact uterus received a sequential or continuous combined regimen of estradiol hemihydrate and norethisterone acetate; those who had undergone hysterectomy received estradiol alone. Therapy was to be continued for 2 years; women were followed by a breast cancer specialist for recurrence of disease twice yearly for 3 years then annually for a total of at least 5 years. Mammograms were done with a target interval of 18–24 months.

Baseline characteristics for women in the HT arm and non-HT arm were similar, with the exception being that there were more women with receptor-positive breast cancer in the HT arm (62.3% vs. 54.5%). Women in the HT arm had an increased rate of a new breast cancer event even after adjusting for use of HT prior to diagnosis of the original breast cancer, tamoxifen use, and hormone receptor status (HR 2.2, 95% CI 1.0–5.1). All of the women in HT arm who experienced a recurrence had been exposed to HT; five women in the non-HT arm with recurrence had also been exposed to HT after random assignment. There was no difference in distant metastasis-free survival or overall mortality between the two arms.219

The Stockholm trial randomized 378 postmenopausal women with a history of treated breast cancer to HT or no HT for 5 years. Randomization was stratified by tamoxifen use, type of HT and time since primary diagnosis (less than 2 years vs. more than 2 years). For those randomized to HT, women under age 55 received cyclic combined therapy with estradiol for 21 days with medroxyprogesterone acetate added the last 10 days followed by 7 hormone free days, while those 55 years or older received a “long cycle” regimen composed of estradiol for 84 days with medroxyprogesterone acetate for the last 14 days followed by 7 days with no treatment. Women without a uterus received estradiol valerate continuously. Women were followed twice yearly for the first 5 years after the primary diagnosis and annually for the next 5 years. Mammograms were performed annually.

Baseline characteristics for the two groups were similar. Of women in the control group, 10% took some form of HT after randomization. At a median follow up of 10.8 years, the HR for breast cancer recurrence or death in the HR group compared to the non-HR group was non-significantly increased at 1.3 (95% CI 0.9–1.9). However, when analysed specifically for contralateral breast cancer, a significantly increased risk was observed with HT with 14 (of 23 total recurrences) reported among HT users compared to four (of 27 total recurrences) among non-users (HR 3.6, 95% CI 1.2–10.9);220 a hazard ratio that comes very close to that of the HABITS trial. Of the 18 total contralateral breast cancer cases, 11 were diagnosed in women on concomitant tamoxifen and eight (seven in the HT group) were of different histology than the primary tumor. In comparison, five of the 26 recurrent breast cancers in the HT arm of the HABITS trial were found in the contralateral breast.218 Also of note, in the Stockholm trial, women who were diagnosed with breast cancer less than 2 years before randomization had an increased risk of contralateral breast cancer (HR 4.8, 95% CI 1.0–22).220 The occurrence of contralateral breast cancer is notable and may represent a new primary breast cancer occurrence or growth of non-excised multifocal primary disease. Similar to the HABITS trial, no difference in mortality was seen between the HT and non-HT groups in the Stockholm trial.220

The apparently contradictory findings between the HABITS trial and Stockholm trial may be due to chance, however several authors have suggested that differences in the patient populations, primary and secondary end points and interventions may explain their different outcomes. The Stockholm trial focused on minimizing progestin exposure. This reduction in progestin exposure in the Stockholm trial may explain the lack of an association between HT and breast cancer recurrence. In addition, the HABITS trial contained a higher proportion of women with positive lymph nodes (26% vs. 16%) and fewer women treated with concurrent tamoxifen (21% vs. 52%).221  

A recent systematic review of 20 studies, including the HABITS and Stockholm trials, concluded that the available data are insufficient to exclude a harmful effect of HT on breast cancer recurrence.222 The possibility that regimens minimizing progestin exposure may reduce risk of recurrence with MHT and the effect of length of time between cancer diagnosis and initiation of MHT on breast cancer recurrence are two areas in particular that warrant further evaluation.

Review of the data regarding breast cancer and MHT as discussed in the sections above can lead to a few general conclusions. E+P MHT increases the risk of breast cancer in current users, with increased risk seen with increasing duration of use and reduction of risk to baseline relatively quickly after cessation. Observational data suggests E-only MHT may increase risk in current users, however, the largest randomized controlled trial found a risk reduction among current users. Women who start MHT shortly after menopausal onset are at increased risk for breast cancer compared to those who start years after menopause which may explain the conflicting data seen in the WHI E-only arm compared to observational studies. Although observational studies have found more favorable features of breast cancer in MHT users, the WHI showed a worse prognosis for those women on MHT who developed breast cancer. The use of MHT in women with a history of breast cancer is still controversial, but regimens that minimize progestin exposure may confer less risk of recurrence than others.

Endometrial cancer, endometrioid tumors, and endometriosis

The question of safety of MHT in women with a history of endometrial cancer is still not adequately answered. Retrospective studies looking at MHT after treatment for early stage endometrial adenocarcinoma have not shown an increase in recurrence or cancer related mortality.223, 224, 225 However, the concern that MHT may stimulate residual cancer cells remains. The Gynecologic Oncology Group (GOG) attempted to answer this question more definitively with a prospective randomized placebo-controlled trial, however, patient recruitment decreased significantly after the initial WHI findings were released and the trial was terminated prematurely. Women with stage I or II endometrial cancer who had undergone surgical treatment with indications for MHT were randomized to receive either estrogen-only or placebo for 3 years. A total of 618 women were randomized to each arm and had a median follow up of 35.7 months. In all 1.3% of women in the placebo arm and 1.5% of women in the estrogen arm experienced disease recurrence (RR 1.27, 80% CI 0.92–1.77), while 0.6% and 0.8%, respectively, died from their endometrial cancer.226 Although this study did not find a statistically significant difference in disease recurrence with MHT, sample size was significantly short of their goal of 2108 patients.

Although lacking a well powered randomized, controlled trial, existing data suggest that MHT in women with early stage endometrial cancer may not increase recurrence risk. Little is known about the risk in patients with more advanced disease. If a high-risk tumor is estrogen and progesterone receptor negative, it seems reasonable to allow immediate hormone therapy if symptoms are severe and cannot be controlled with non-hormonal alternatives. Because the latent period of endometrial cancer is relatively short, a period of time (5 years) without evidence of recurrence would increase the likelihood of safety on an estrogen program. The combination of estrogen–progestin is recommended in view of the potential protective action of the progestational agent. A similar approach makes sense for patients previously treated for endometrioid tumors of the ovary. In view of the fact that adenocarcinoma has been reported in patients with pelvic endometriosis on unopposed estrogen, a combined estrogen–progestin program is also advised in patients with a history of endometriosis who require MHT.227, 228

OTHER CONDITIONS

A nonoral route of administration should be considered for women at high risk of gallbladder disease because the nonoral route avoids the production of a more lithogenic bile (which is associated with oral treatment).229 Close surveillance is indicated for some patients with seizure disorders, familial hyperlipidemias (elevated triglycerides), and migraine headaches. Patients with migraine headaches often improve if a daily, continuous method of treatment is used, eliminating hormone fluctuations that can trigger headaches.

Conditions that do not represent contraindications include controlled hypertension, diabetes mellitus, smoking, and varicose veins. The belief that estrogen is potentially harmful with each of these clinical situations is derived from old studies of high-dose oral contraceptives. Estrogen in appropriate doses is acceptable in the presence of these conditions.

No other cancers (besides those mentioned earlier) are known to be adversely affected by hormone therapy. Postmenopausal hormone therapy can be administered to all patients with cervical, ovarian, or vulvar malignancies.

REFERENCES

1

Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 2002;288:321-33.

2

Anderson GL, Limacher M, Assaf AR, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 2004;291:1701-12.

3

Ettinger B, Wang SM, Leslie RS, et al. Evolution of postmenopausal hormone therapy between 2002 and 2009. Menopause 2012;19:610-5.

4

Heiss G, Wallace R, Anderson GL, et al. Health risks and benefits 3 years after stopping randomized treatment with estrogen and progestin. JAMA 2008;299:1036-45.

5

Nedrow A, Miller J, Walker M, Nygren P, Huffman LH, Nelson HD. Complementary and alternative therapies for the management of menopause-related symptoms: a systematic evidence review. Arch Intern Med 2006;166:1453-65.

6

http://www.endo-society.org/advocacy/policy/upload/Joint-Statement-The-Experts-Do-Agree-About-Hormone-Therapy.pdf.

7

Lindsay R, Hart DM, Clark DM. The minimum effective dose of estrogen for prevention of postmenopausal bone loss. Obstet Gynecol 1984;63:759-63.

8

Genant HK, Cann CE, Ettinger B, Gordan GS. Quantitative computed tomography of vertebral spongiosa: a sensitive method for detecting early bone loss after oophorectomy. Ann Intern Med 1982;97:699-705.

9

Prestwood KM, Kenny AM, Kleppinger A, Kulldorff M. Ultralow-dose micronized 17beta-estradiol and bone density and bone metabolism in older women: a randomized controlled trial. JAMA 2003;290:1042-8.

10

Menostar: a low-dose estrogen patch for osteoporosis. Obstet Gynecol 2005;105:432-3.

11

Mashchak CA, Lobo RA, Dozono-Takano R, et al. Comparison of pharmacodynamic properties of various estrogen formulations. Am J Obstet Gynecol 1982;144:511-8.

12

Horsman A, Jones M, Francis R, Nordin C. The effect of estrogen dose on postmenopausal bone loss. N Engl J Med 1983;309:1405-7.

13

Field CS, Ory SJ, Wahner HW, Herrmann RR, Judd HL, Riggs BL. Preventive effects of transdermal 17 beta-estradiol on osteoporotic changes after surgical menopause: a two-year placebo-controlled trial. Am J Obstet Gynecol 1993;168:114-21.

14

Dusterberg B, Nishino Y. Pharmacokinetic and pharmacological features of oestradiol valerate. Maturitas 1982;4:315-24.

15

Itoi H, Minakami H, Sato I. Comparison of the long-term effects of oral estriol with the effects of conjugated estrogen, 1-alpha-hydroxyvitamin D3 and calcium lactate on vertebral bone loss in early menopausal women. Maturitas 1997;28:11-7.

16

Schiff I, Tulchinsky D, Ryan KJ, Kadner S, Levitz M. Plasma estriol and its conjugates following oral and vaginal administration of estriol to postmenopausal women: correlations with gonadotropin levels. Am J Obstet Gynecol 1980;138:1137-41.

17

Kuhl H. Pharmacokinetics of oestrogens and progestogens. Maturitas 1990;12:171-97.

18

Colvin PL, Jr., Auerbach BJ, Koritnik DR, Hazzard WR, Applebaum-Bowden D. Differential effects of oral estrone versus 17 beta-estradiol on lipoproteins in postmenopausal women. J Clin Endocrinol Metab 1990;70:1568-73.

19

Walsh BW, Li H, Sacks FM. Effects of postmenopausal hormone replacement with oral and transdermal estrogen on high density lipoprotein metabolism. J Lipid Res 1994;35:2083-93.

20

Pang SC, Greendale GA, Cedars MI, et al. Long-term effects of transdermal estradiol with and without medroxyprogesterone acetate. Fertil Steril 1993;59:76-82.

21

Hillard TC, Whitcroft SJ, Marsh MS, et al. Long-term effects of transdermal and oral hormone replacement therapy on postmenopausal bone loss. Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 1994;4:341-8.

22

Olie V, Canonico M, Scarabin PY. Risk of venous thrombosis with oral versus transdermal estrogen therapy among postmenopausal women. Current opinion in hematology 2010;17:457-63.

23

Lobo RA, March CM, Goebelsmann U, Krauss RM, Mishell DR, Jr. Subdermal estradiol pellets following hysterectomy and oophorectomy. Effect upon serum estrone, estradiol, luteinizing hormone, follicle-stimulating hormone, corticosteroid binding globulin-binding capacity, testosterone-estradiol binding globulin-binding capacity, lipids, and hot flushes. Am J Obstet Gynecol 1980;138:714-9.

24

Committee CoGPatASfRMP. Committee Opinion 532: compounded bioidentical menopausal hormone therapy. Obstet Gynecol 2012;120:411-5.

25

Steingold KA, Laufer L, Chetkowski RJ, et al. Treatment of hot flashes with transdermal estradiol administration. J Clin Endocrinol Metab 1985;61:627-32.

26

Castelo-Branco C, Martinez de Osaba MJ, Vanrezc JA, Fortuny A, Gonzalez-Merlo J. Effects of oophorectomy and hormone replacement therapy on pituitary-gonadal function. Maturitas 1993;17:101-11.

27

Weiderpass E, Adami HO, Baron JA, et al. Risk of endometrial cancer following estrogen replacement with and without progestins. J Natl Cancer Inst 1999;91:1131-7.

28

de Ziegler D, Ferriani R, Moraes LA, Bulletti C. Vaginal progesterone in menopause: Crinone 4% in cyclical and constant combined regimens. Hum Reprod 2000;15 Suppl 1:149-58.

29

Cicinelli E, de Ziegler D, Galantino P, et al. Twice-weekly transdermal estradiol and vaginal progesterone as continuous combined hormone replacement therapy in postmenopausal women: a 1-year prospective study. Am J Obstet Gynecol 2002;187:556-60.

30

Woodruff JD, Pickar JH. Incidence of endometrial hyperplasia in postmenopausal women taking conjugated estrogens (Premarin) with medroxyprogesterone acetate or conjugated estrogens alone. The Menopause Study Group. Am J Obstet Gynecol 1994;170:1213-23.

31

Effects of hormone replacement therapy on endometrial histology in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA 1996;275:370-5.

32

Cicinelli E, de Ziegler D, Alfonso R, Nicoletti R, Bellavia M, Colafiglio G. Endometrial effects, bleeding control, and compliance with a new postmenopausal hormone therapy regimen based on transdermal estradiol gel and every-other-day vaginal progesterone in capsules: a 3-year pilot study. Fertil Steril 2005;83:1859-63.

33

Archer DF, Pickar JH, Bottiglioni F. Bleeding patterns in postmenopausal women taking continuous combined or sequential regimens of conjugated estrogens with medroxyprogesterone acetate. Menopause Study Group. Obstet Gynecol 1994;83:686-92.

34

King RJ, Whitehead MI. Assessment of the potency of orally administered progestins in women. Fertil Steril 1986;46:1062-6.

35

Gillet JY, Andre G, Faguer B, et al. Induction of amenorrhea during hormone replacement therapy: optimal micronized progesterone dose. A multicenter study. Maturitas 1994;19:103-15.

36

http://www.iom.edu/Reports/2010/Dietary-Reference-Intakes-for-Calcium-and-Vitamin-D/DRI-Values.aspx.

37

Speroff T, Dawson NV, Speroff L, Haber RJ. A risk-benefit analysis of elective bilateral oophorectomy: effect of changes in compliance with estrogen therapy on outcome. Am J Obstet Gynecol 1991;164:165-74.

38

Berman RS, Epstein RS, Lydick E. Risk factors associated with women's compliance with estrogen replacement therapy. Journal of women's health / the official publication of the Society for the Advancement of Women's Health Research 1997;6:219-26.

39

Ravnikar VA. Compliance with hormone therapy. Am J Obstet Gynecol 1987;156:1332-4.

40

Burich RA, Mehta NR, DeGregorio MW. Ospemifine and 4-hydroxyospemifine effectively prevent and traet breast cancer in the MTag.Tg transgenic mouse model. Menopause 2012;19:96-103.

41

http://www.clinicaltrials.gov/ct2/show/NCT00729469.

42

Simon J, Nachtigall L, Gut R, Lang E, Archer DF, Utian W. Effective treatment of vaginal atrophy with an ultra-low-dose estradiol vaginal tablet. Obstet Gynecol 2008;112:1053-60.

43

Padwick ML, Pryse-Davies J, Whitehead MI. A simple method for determining the optimal dosage of progestin in postmenopausal women receiving estrogens. N Engl J Med 1986;315:930-4.

44

Sturdee DW, Barlow DH, Ulrich LG, et al. Is the timing of withdrawal bleeding a guide to endometrial safety during sequential oestrogen-progestagen replacement therapy? UK Continuous Combined HRT Study Investigators. Lancet 1994;344:979-82.

45

Andersson JK, Rybo G. Levonorgestrel-releasing intrauterine device in the treatment of menorrhagia. British journal of obstetrics and gynaecology 1990;97:690-4.

46

Raudaskoski TH, Lahti EI, Kauppila AJ, Apaja-Sarkkinen MA, Laatikainen TJ. Transdermal estrogen with a levonorgestrel-releasing intrauterine device for climacteric complaints: clinical and endometrial responses. Am J Obstet Gynecol 1995;172:114-9.

47

Archer DF, McIntyre-Seltman K, Wilborn WW, Jr., et al. Endometrial morphology in asymptomatic postmenopausal women. Am J Obstet Gynecol 1991;165:317-20; discussion 20-2.

48

Korhonen MO, Symons JP, Hyde BM, Rowan JP, Wilborn WH. Histologic classification and pathologic findings for endometrial biopsy specimens obtained from 2964 perimenopausal and postmenopausal women undergoing screening for continuous hormones as replacement therapy (CHART 2 Study). Am J Obstet Gynecol 1997;176:377-80.

49

Langer RD, Pierce JJ, O'Hanlan KA, et al. Transvaginal ultrasonography compared with endometrial biopsy for the detection of endometrial disease. Postmenopausal Estrogen/Progestin Interventions Trial. N Engl J Med 1997;337:1792-8.

50

Karlsson B, Granberg S, Wikland M, et al. Transvaginal ultrasonography of the endometrium in women with postmenopausal bleeding--a Nordic multicenter study. Am J Obstet Gynecol 1995;172:1488-94.

51

Botsis D, Kassanos D, Pyrgiotis E, Zourlas PA. Vaginal sonography of the endometrium in postmenopausal women. Clinical and experimental obstetrics & gynecology 1992;19:189-92.

52

Hanggi W, Bersinger N, Altermatt HJ, Birkhauser MH. Comparison of transvaginal ultrasonography and endometrial biopsy in endometrial surveillance in postmenopausal HRT users. Maturitas 1997;27:133-43.

53

Goldstein SR. Sonography in postmenopausal bleeding. J Ultrasound Med 2012;31:333-6.

54

Mortakis AE, Mavrelos K. Transvaginal ultrasonography and hysteroscopy in the diagnosis of endometrial abnormalities. J Am Assoc Gynecol Laparosc 1997;4:449-52.

55

Granberg S, Ylostalo P, Wikland M, Karlsson B. Endometrial sonographic and histologic findings in women with and without hormonal replacement therapy suffering from postmenopausal bleeding. Maturitas 1997;27:35-40.

56

Bakos O, Smith P, Heimer G. Transvaginal ultrasonography for identifying endometrial pathology in postmenopausal women. Maturitas 1994;20:181-9.

57

Levine D, Gosink BB, Johnson LA. Change in endometrial thickness in postmenopausal women undergoing hormone replacement therapy. Radiology 1995;197:603-8.

58

Sladkevicius P, Valentin L, Marsal K. Endometrial thickness and Doppler velocimetry of the uterine arteries as discriminators of endometrial status in women with postmenopausal bleeding: a comparative study. Am J Obstet Gynecol 1994;171:722-8.

59

Feldman S, Shapter A, Welch WR, Berkowitz RS. Two-year follow-up of 263 patients with post/perimenopausal vaginal bleeding and negative initial biopsy. Gynecol Oncol 1994;55:56-9.

60

McGonigle KF, Karlan BY, Barbuto DA, Leuchter RS, Lagasse LD, Judd HL. Development of endometrial cancer in women on estrogen and progestin hormone replacement therapy. Gynecol Oncol 1994;55:126-32.

61

Beresford SA, Weiss NS, Voigt LF, McKnight B. Risk of endometrial cancer in relation to use of oestrogen combined with cyclic progestagen therapy in postmenopausal women. Lancet 1997;349:458-61.

62

Kirkham C, Hahn PM, Van Vugt DA, Carmichael JA, Reid RL. A randomized, double-blind, placebo-controlled, cross-over trial to assess the side-effects of medroxyprogesterone acetate in hormone replacement therapy. Obstet Gynecol 1991;78:93-7.

63

Prior JC, Alojado N, McKay DW, Vigna YM. No adverse effects of medroxyprogesterone treatment without estrogen in postmenopausal women: double-blind, placebo-controlled, crossover trial. Obstet Gynecol 1994;83:24-8.

64

Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) Trial. The Writing Group for the PEPI Trial. JAMA 1995;273:199-208.

65

Fitzpatrick LA, Pace C, Wiita B. Comparison of regimens containing oral micronized progesterone or medroxyprogesterone acetate on quality of life in postmenopausal women: a cross-sectional survey. Journal of women's health & gender-based medicine 2000;9:381-7.

66

Williams DB, Voigt BJ, Fu YS, Schoenfeld MJ, Judd HL. Assessment of less than monthly progestin therapy in postmenopausal women given estrogen replacement. Obstet Gynecol 1994;84:787-93.

67

Ettinger B, Selby J, Citron JT, Vangessel A, Ettinger VM, Hendrickson MR. Cyclic Hormone Replacement Therapy Using Quarterly Progestin. Obstetrics and Gynecology 1994;83:693-700.

68

Hirvonen E, Salmi T, Puolakka J, et al. Can progestin be limited to every third month only in postmenopausal women taking estrogen? Maturitas 1995;21:39-44.

69

Cerin A, Heldaas K, Moeller B. Adverse endometrial effects of long-cycle estrogen and progestogen replacement therapy. The Scandinavian LongCycle Study Group. N Engl J Med 1996;334:668-9.

70

Miles RA, Paulson RJ, Lobo RA, Press MF, Dahmoush L, Sauer MV. Pharmacokinetics and endometrial tissue levels of progesterone after administration by intramuscular and vaginal routes: a comparative study. Fertil Steril 1994;62:485-90.

71

Ross D, Cooper AJ, Pryse-Davies J, Bergeron C, Collins WP, Whitehead MI. Randomized, double-blind, dose-ranging study of the endometrial effects of a vaginal progesterone gel in estrogen-treated postmenopausal women. Am J Obstet Gynecol 1997;177:937-41.

72

Meldrum DR, Davidson BJ, Tataryn IV, Judd HL. Changes in circulating steroids with aging in postmenopausal women. Obstet Gynecol 1981;57:624-8

73

Burger HG, Dudley EC, Cui J, Dennerstein L, Hopper JL. A prospective longitudinal study of serum testosterone, dehydroepiandrosterone sulfate, and sex hormone-binding globulin levels through the menopause transition. J Clin Endocrinol Metab 2000;85:2832-8.

74

Labrie F, Belanger A, Cusan L, Gomez JL, Candas B. Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. J Clin Endocrinol Metab 1997;82:2396-402.

75

Garnett T, Studd J, Watson N, Savvas M, Leather A. The effects of plasma estradiol levels on increases in vertebral and femoral bone density following therapy with estradiol and estradiol with testosterone implants. Obstet Gynecol 1992;79:968-72.

76

Watts NB, Notelovitz M, Timmons MC, Addison WA, Wiita B, Downey LJ. Comparison of oral estrogens and estrogens plus androgen on bone mineral density, menopausal symptoms, and lipid-lipoprotein profiles in surgical menopause. Obstet Gynecol 1995;85:529-37.

77

Davis SR, McCloud P, Strauss BJ, Burger H. Testosterone enhances estradiol's effects on postmenopausal bone density and sexuality. Maturitas 1995;21:227-36.

78

Somboonporn W, Davis SR, Seif M, Bell R. Testosterone for peri- and postmenopausal women. Cochrane Database Syst Rev 2005, updated 2009;CD004509.

79

Heard-Davison A, Heiman JR, Kuffel S. Genital and subjective measurement of the time course effects of an acute dose of testosterone vs. placebo in postmenopausal women. J Sex Med 2007;4:209-17.

80

Tuiten A, Von Honk J, Koppeschaar H, Bernaards C, Thijssen J, Verbaten R. Time course of effects of testosterone administration on sexual arousal in women. Archives of general psychiatry 2000;57:149.

81

Davis SR, Moreau M, Kroll R, et al. Testosterone for Low Libido in Menopausal Women Not Taking Estrogen Therapy. N Eng J Med 2008;359:2005-17.

82

Shifren JL, Braunstein GD, Simon JA, et al. Transdermal testosterone treatment in women with impaired sexual function after oophorectomy. N Engl J Med 2000;343:682-8.

83

Braunstein GD, Sundwall DA, Katz M, et al. Safety and efficacy of a testosterone patch for the treatment of hypoactive sexual desire disorder in surgically menopausal women: a randomized, placebo-controlled trial. Arch Intern Med 2005;165:1582-9.

84

Sherwin BB, Gelfand MM. The role of androgen in the maintenance of sexual functioning in oophorectomized women. Psychosom Med 1987;49:397-409.

85

Myers LS, Dixen J, Morrissette D, Carmichael M, Davidson JM. Effects of estrogen, androgen, and progestin on sexual psychophysiology and behavior in postmenopausal women. J Clin Endocrinol Metab 1990;70:1124-31.

86

Hickok LR, Toomey C, Speroff L. A comparison of esterified estrogens with and without methyltestosterone: effects on endometrial histology and serum lipoproteins in postmenopausal women. Obstet Gynecol 1993;82:919-24.

87

Barrett-Connor E, Young R, Notelovitz M, et al. A two-year, double-blind comparison of estrogen-androgen and conjugated estrogens in surgically menopausal women. Effects on bone mineral density, symptoms and lipid profiles. The Journal of reproductive medicine 1999;44:1012-20.

88

Davis SR, Moreau M, Kroll R, et al. Testosterone for low libido in postmenopausal women not taking estrogen. N Engl J Med 2008;359:2005-17.

89

Draper MW, Flowers DE, Huster WJ, Neild JA, Harper KD, Arnaud C. A controlled trial of raloxifene (LY139481) HCl: impact on bone turnover and serum lipid profile in healthy postmenopausal women. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 1996;11:835-42.

90

Boss SM, Huster WJ, Neild JA, Glant MD, Eisenhut CC, Draper MW. Effects of raloxifene hydrochloride on the endometrium of postmenopausal women. Am J Obstet Gynecol 1997;177:1458-64.

91

Delmas PD, Bjarnason NH, Mitlak BH, et al. Effects of raloxifene on bone mineral density, serum cholesterol concentrations, and uterine endometrium in postmenopausal women. N Engl J Med 1997;337:1641-7.

92

Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006;295:2727-41.

93

Vogel VG, Costantino JP, Wickerham DL, et al. Update of the National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial: Preventing breast cancer. Cancer Prev Res (Phila) 2010;3:696-706.

94

Frigo P, Eppel W, Asseryanis E, et al. The effects of hormone substitution in depot form on the uterus in a group of 50 perimenopausal women--a vaginosonographic study. Maturitas 1995;21:221-5.

95

Sener AB, Seckin NC, Ozmen S, Gokmen O, Dogu N, Ekici E. The effects of hormone replacement therapy on uterine fibroids in postmenopausal women. Fertil Steril 1996;65:354-7.

96

Schwartz LB, Lazer S, Mark M, Nachtigall LE, Horan C, Goldstein SR. Does the use of postmenopausal hormone replacement therapy influence the size of uterine leiomyomata? A preliminary report. Menopause-the Journal of the North American Menopause Society 1996;3:38-43.

97

Schwartz SM, Weiss NS, Daling JR, et al. Exogenous sex hormone use, correlates of endogenous hormone levels, and the incidence of histologic types of sarcoma of the uterus. Cancer 1996;77:717-24.

98

Vandenbroucke JP, Witteman JC, Valkenburg HA, et al. Noncontraceptive hormones and rheumatoid arthritis in perimenopausal and postmenopausal women. JAMA 1986;255:1299-303.

99

Koepsell TD, Dugowson CE, Nelson JL, Voigt LF, Daling JR. Non-contraceptive hormones and the risk of rheumatoid arthritis in menopausal women. International journal of epidemiology 1994;23:1248-55.

100

Spector TD, Brennan P, Harris P, Studd JW, Silman AJ. Does estrogen replacement therapy protect against rheumatoid arthritis? J Rheumatol 1991;18:1473-6.

101

Hall GM, Daniels M, Huskisson EC, Spector TD. A randomised controlled trial of the effect of hormone replacement therapy on disease activity in postmenopausal rheumatoid arthritis. Ann Rheum Dis 1994;53:112-6.

102

Sanchez-Guerrero J, Liang MH, Karlson EW, Hunter DJ, Colditz GA. Postmenopausal estrogen therapy and the risk for developing systemic lupus erythematosus. Ann Intern Med 1995;122:430-3.

103

Arden NK, Lloyd ME, Spector TD, Hughes GR. Safety of hormone replacement therapy (HRT) in systemic lupus erythematosus (SLE). Lupus 1994;3:11-3.

104

Sowers MF, Hochberg M, Crabbe JP, Muhich A, Crutchfield M, Updike S. Association of bone mineral density and sex hormone levels with osteoarthritis of the hand and knee in premenopausal women. Am J Epidemiol 1996;143:38-47.

105

Nevitt MC, Lane NE, Scott JC, et al. Radiographic osteoarthritis of the hip and bone mineral density. The Study of Osteoporotic Fractures Research Group. Arthritis Rheum 1995;38:907-16.

106

Nevitt MC, Cummings SR, Lane NE, et al. Association of estrogen replacement therapy with the risk of osteoarthritis of the hip in elderly white women. Study of Osteoporotic Fractures Research Group. Arch Intern Med 1996;156:2073-80.

107

Volpe A, Lucenti V, Forabosco A, et al. Oral discomfort and hormone replacement therapy in the post-menopause. Maturitas 1991;13:1-5.

108

Krall EA, Dawson-Hughes B, Papas A, Garcia RI. Tooth loss and skeletal bone density in healthy postmenopausal women. Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 1994;4:104-9.

109

Daniell HW. Postmenopausal tooth loss. Contributions to edentulism by osteoporosis and cigarette smoking. Arch Intern Med 1983;143:1678-82.

110

Paganini-Hill A. The benefits of estrogen replacement therapy on oral health. The Leisure World cohort. Arch Intern Med 1995;155:2325-9.

111

Grodstein F, Colditz GA, Stampfer MJ. Post-menopausal hormone use and tooth loss: a prospective study. J Am Dent Assoc 1996;127:370-7, quiz 92.

112

Harris TM. The pharmacological treatment of voice disorders. Folia phoniatrica 1992;44:143-54.

113

Metka M, Enzelsberger H, Knogler W, Schurz B, Aichmair H. Ophthalmic complaints as a climacteric symptom. Maturitas 1991;14:3-8.

114

Kramer P, Lubkin V, Potter W, Jacobs M, Labay G, Silverman P. Cyclic changes in conjunctival smears from menstruating females. Ophthalmology 1990;97:303-7.

115

Erdem U, Ozdegirmenci O, Sobaci E, Sobaci G, Goktolga U, Dagli S. Dry eye in post-menopausal women using hormone replacement therapy. Maturitas 2007;56:257-62.

116

Klein BE, Klein R, Ritter LL. Is there evidence of an estrogen effect on age-related lens opacities? The Beaver Dam Eye Study. Arch Ophthalmol 1994;112:85-91.

117

Benitez del Castillo JM, del Rio T, Garcia-Sanchez J. Effects of estrogen use on lens transmittance in postmenopausal women. Ophthalmology 1997;104:970-3.

118

Cumming RG, Mitchell P. Hormone replacement therapy, reproductive factors, and cataract. The Blue Mountains Eye Study. Am J Epidemiol 1997;145:242-9.

119

Tint NL, Alexander P, Tint KM, Vasileiadis GT, Yeung AM, Azuara-Blanco A. Hormone therapy and intraocular pressure in nonglaucomatous eyes. Menopause 2010;17:157-60.

120

Multiple A. Management of menopausal symptoms. Am J Med 2005;118

121

Nelson HD, Vesco KK, Haney E, et al. Nonhormonal therapies for menopausal hot flashes: systematic review and meta-analysis. JAMA 2006;295:2057-71.

122

Newton KM, Reed SD, LaCroix AZ, Grothaus LC, Ehrlich K, Guiltinan J. Treatment of vasomotor symptoms of menopause with black cohosh, multibotanicals, soy, hormone therapy, or placebo: a randomized trial. Ann Intern Med 2006;145:869-79.

123

Dezentje VO, van Blijderveen NJ, Gelderblom H, et al. Effect of concomitant CYP2D6 inhibitor use and tamoxifen adherence on breast cancer recurrence in early-stage breast cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2010;28:2423-9.

124

Kelly CM, Juurlink DN, Gomes T, et al. Selective serotonin reuptake inhibitors and breast cancer mortality in women receiving tamoxifen: a population based cohort study. BMJ 2010;340:c693.

125

Nagamani M, Kelver ME, Smith ER. Treatment of menopausal hot flashes with transdermal administration of clonidine. Am J Obstet Gynecol 1987;156:561-5.

126

Goldberg RM, Loprinzi CL, O'Fallon JR, et al. Transdermal clonidine for ameliorating tamoxifen-induced hot flashes. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 1994;12:155-8.

127

Lebherz TB, French L. Nonhormonal treatment of the menopausal syndrome. A double-blind evaluation of an autonomic system stabilizer. Obstet Gynecol 1969;33:795-9.

128

Melis GB, Gambacciani M, Cagnacci A, Paoletti AM, Mais V, Fioretti P. Effects of the dopamine antagonist veralipride on hot flushes and luteinizing hormone secretion in postmenopausal women. Obstet Gynecol 1988;72:688-92.

129

David A, Don R, Tajchner G, Weissglas L. Veralipride: alternative antidopaminergic treatment for menopausal symptoms. Am J Obstet Gynecol 1988;158:1107-15.

130

Lobo RA, McCormick W, Singer F, Roy S. Depo-medroxyprogesterone acetate compared with conjugated estrogens for the treatment of postmenopausal women. Obstet Gynecol 1984;63:1-5.

131

Loprinzi CL, Michalak JC, Quella SK, et al. Megestrol acetate for the prevention of hot flashes. N Engl J Med 1994;331:347-52.

132

Nesheim BI, Saetre T. Reduction of menopausal hot flushes by methyldopa. A double blind crossover trial. Eur J Clin Pharmacol 1981;20:413-6.

133

Ross LA, Alder EM. Tibolone and climacteric symptoms. Maturitas 1995;21:127-36.

134

Somers S. Ageless: the naked truth about bioidentical hormones. Random House, NY 2006.

135

Somers S. The sexy years: the secret to fabulous sex, great health and vitality, for women and men. Random House, NY 2004.

136

http://www.fda.gov/ForConsumers/ConsumerUpdates/ucm049311.htm. Bioidenticals: sorting myths from facts. Accessed October 5, 2009 2008.

137

Kuiper GG, Carlsson B, Grandien K, et al. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997;138:863-70.

138

Anthony MS, Clarkson TB, Hughes CL, Jr., Morgan TM, Burke GL. Soybean isoflavones improve cardiovascular risk factors without affecting the reproductive system of peripubertal rhesus monkeys. The Journal of nutrition 1996;126:43-50.

139

Anthony MS, Clarkson TB, Bullock BC, Wagner JD. Soy protein versus soy phytoestrogens in the prevention of diet-induced coronary artery atherosclerosis of male cynomolgus monkeys. Arterioscler Thromb Vasc Biol 1997;17:2524-31.

140

Foth D, Cline JM. Effects of mammalian and plant estrogens on mammary glands and uteri of macaques. The American journal of clinical nutrition 1998;68:1413S-7S.

141

Albertazzi P, Pansini F, Bonaccorsi G, Zanotti L, Forini E, De Aloysio D. The effect of dietary soy supplementation on hot flushes. Obstet Gynecol 1998;91:6-11.

142

Lu LJ, Anderson KE, Grady JJ, Nagamani M. Effects of soya consumption for one month on steroid hormones in premenopausal women: implications for breast cancer risk reduction. Cancer Epidemiol Biomarkers Prev 1996;5:63-70.

143

Dayal M, Sammel MD, Zhao J, Hummel AC, Vandenbourne K, Barnhart KT. Supplementation with DHEA: effect on muscle size, strength, quality of life, and lipids. J Womens Health (Larchmt) 2005;14:391-400.

144

Kritz-Silverstein D, von Muhlen D, Laughlin GA, Bettencourt R. Effects of dehydroepiandrosterone supplementation on cognitive function and quality of life: the DHEA and Well-Ness (DAWN) Trial. Journal of the American Geriatrics Society 2008;56:1292-8.

145

Nair KS, Rizza RA, O'Brien P, et al. DHEA in elderly women and DHEA or testosterone in elderly men. N Engl J Med 2006;355:1647-59.

146

Parsons TD, Kratz KM, Thompson E, Stanczyk FZ, Buckwalter JG. Dhea supplementation and cognition in postmenopausal women. The International journal of neuroscience 2006;116:141-55.

147

Glueck CJ, Lang J, Hamer T, Tracy T. Severe hypertriglyceridemia and pancreatitis when estrogen replacement therapy is given to hypertriglyceridemic women. J Lab Clin Med 1994;123:59-64.

148

Cirillo DJ, Wallace RB, Rodabough RJ, et al. Effect of estrogen therapy on gallbladder disease. JAMA 2005;293:330-9.

149

Grodstein F, Colditz GA, Stampfer MJ. Postmenopausal hormone use and cholecystectomy in a large prospective study. Obstet Gynecol 1994;83:5-11.

150

La Vecchia C, Negri E, D'Avanzo B, Parazzini F, Gentile A, Franceschi S. Oral contraceptives and non-contraceptive oestrogens in the risk of gallstone disease requiring surgery. Journal of epidemiology and community health 1992;46:234-6

151

!!! INVALID CITATION !!!

152

Wing RR, Matthews KA, Kuller L, Meilhan EH, Pantinga PL. Weight gain at the time of menopause. Arch Intern Med 1990;151:97-102

153

Kritz-Silverstein D, Barrett-Connor E. Long-term postmenopausal hormone use, obesity, and fat distribution in older women. JAMA 1996;275:46-9.

154

Espeland MA, Stefanick ML, Kritz-Silverstein D, et al. Effect of postmenopausal hormone therapy on body weight and waist and hip girths. Postmenopausal Estrogen-Progestin Interventions Study Investigators. J Clin Endocrinol Metab 1997;82:1549-56.

155

Sutton-Tyrrell K, Zhao X, Santoro N, et al. Reproductive hormones and obesity: 9 years of observation from the Study of Women's Health Across the Nation. Am J Epidemiol 2010;171:1203-13

156

Torrens JI, Sutton-Tyrrell K, Zhao X, et al. Relative androgen excess during the menopausal transition predicts incident metabolic syndrome in midlife women: study of Women's Health Across the Nation. Menopause 2009;16:257-64.

157

Wildman RP, Tepper PG, Crawford S, et al. Do Changes in Sex Steroid Hormones Precede or Follow Increases in Body Weight during the Menopause Transition? Results from The Study of Women's Health Across the Nation. J Clin Endocrinol Metab 2012;97:E1695-704.

158

Grady D, Hulley SB, Furberg C. Venous thromboembolic events associated with hormone replacement therapy. JAMA 1997;278:477.

159

Grodstein F, Stampfer MJ, Goldhaber SZ, et al. Prospective study of exogenous hormones and risk of pulmonary embolism in women. Lancet 1996;348:983-7.

160

Kurman RJ, Kaminski PF, Norris HJ. The behavior of endometrial hyperplasia. A long-term study of "untreated" hyperplasia in 170 patients. Cancer 1985;56:403-12.

161

Grady D, Gebretsadik T, Kerlikowske K, Ernster V, Petitti D. Hormone replacement therapy and endometrial cancer risk: a meta-analysis. Obstet Gynecol 1995;85:304-13.

162

Weiss NS, Hill DA. Postmenopausal estrogens and progestogens and the incidence of gynecologic cancer. Maturitas 1996;23:235-9.

163

Schiff I, Sela HK, Cramer D, Tulchinsky D, Ryan KJ. Endometrial hyperplasia in women on cyclic or continuous estrogen regimens. Fertil Steril 1982;37:79-82.

164

Notelovitz M, Varner RE, Rebar RW, et al. Minimal endometrial proliferation over a two-year period in postmenopausal women taking 0.3 mg of unopposed esterified estrogens. Menopause-the Journal of the North American Menopause Society 1997;4:80-8.

165

Cushing KL, Weiss NS, Voigt LF, McKnight B, Beresford SA. Risk of endometrial cancer in relation to use of low-dose, unopposed estrogens. Obstet Gynecol 1998;91:35-9.

166

Thom MH, White PJ, Williams RM, et al. Prevention and treatment of endometrial disease in climacteric women receiving oestrogen therapy. Lancet 1979;2:455-7.

167

Whitehead MI, Townsend PT, Pryse-Davies J, Ryder TA, King RJ. Effects of estrogens and progestins on the biochemistry and morphology of the postmenopausal endometrium. N Engl J Med 1981;305:1599-605.

168

Gambrell RD, Jr., Bagnell CA, Greenblatt RB. Role of estrogens and progesterone in the etiology and prevention of endometrial cancer: review. Am J Obstet Gynecol 1983;146:696-707.

169

Persson I, Adami HO, Bergkvist L, et al. Risk of endometrial cancer after treatment with oestrogens alone or in conjunction with progestogens: results of a prospective study. BMJ 1989;298:147-51.

170

Voigt LF, Weiss NS, Chu J, Daling JR, McKnight B, van Belle G. Progestagen supplementation of exogenous oestrogens and risk of endometrial cancer. Lancet 1991;338:274-7.

171

Varma TR. Effect of long-term therapy with estrogen and progesterone on the endometrium of post-menopausal women. Acta Obstet Gynecol Scand 1985;64:41-6.

172

Pike MC, Peters RK, Cozen W, et al. Estrogen-progestin replacement therapy and endometrial cancer. J Natl Cancer Inst 1997;89:1110-6.

173

Weiderpass E, Adami H-O, Baron J, et al. Risk fo endometrial cancer following estrogen replacement with and without progestins. JNCI 1999;91:1131-7.

174

Anderson GL, Judd HL, Kaunitz AM, et al. Effects of estrogen plus progestin on gynecologic cancers and associated diagnostic procedures: the Women's Health Initiative randomized trial. JAMA 2003;290:1739-48.

175

Rodriguez C, Calle EE, Coates RJ, Miracle-McMahill HL, Thun MJ, Heath CW, Jr. Estrogen replacement therapy and fatal ovarian cancer. Am J Epidemiol 1995;141:828-35.

176

Risch HA. Estrogen replacement therapy and risk of epithelial ovarian cancer. Gynecol Oncol 1996;63:254-7.

177

Lacey JV, Jr., Mink PJ, Lubin JH, et al. Menopausal hormone replacement therapy and risk of ovarian cancer. JAMA 2002;288:334-41.

178

Hempling RE, Wong C, Piver MS, Natarajan N, Mettlin CJ. Hormone replacement therapy as a risk factor for epithelial ovarian cancer: results of a case-control study. Obstet Gynecol 1997;89:1012-6.

179

Eeles RA, Tan S, Wiltshaw E, et al. Hormone replacement therapy and survival after surgery for ovarian cancer. BMJ 1991;302:259-62.

180

Adami HO, Persson I, Hoover R, Schairer C, Bergkvist L. Risk of cancer in women receiving hormone replacement therapy. Int J Cancer 1989;44:833-9.

181

Parazzini F, La Vecchia C, Negri E, et al. Case-control study of oestrogen replacement therapy and risk of cervical cancer. BMJ 1997;315:85-8.

182

Ploch E. Hormonal replacement therapy in patients after cervical cancer treatment. Gynecol Oncol 1987;26:169-77.

183

Chute C, Willett W, Colditz G, Stampfer M, Rosner B, Speizer F. A prospective study of reproductive history and exogenous estrogens on the risk of colorectal cancer in women. Epidemiology 1991;2:201-7.

184

Jacobs E, White E, Weiss N. Exogenous hormones, reproductive history, and colon cancer. Cancer Causes Control 1994;5:359-66.

185

Calle E, Miracle-McMahill H, Thun M, Heath CJ. Estrogen replacement therapy and risk of fatal colon cancer in a prospective cohort of postmenopausal women. J Natl Cancer Inst 1995;87:517-23.

186

Kampman E, Potter J, Slattery M, Caan B, Edwards S. Hormone replacement therapy, reproductive history, and colon cancer: a multicenter, case-control study in the United States. Cancer Causes Control 1997;8:146-58.

187

Troisi R, Schairer C, Chow W, Schatzkin A, Brinton L, Fraumeni JJ. A prospective study of menopausal hormones and risk of colorectal cancer (United States). Cancer Causes Control 1997;8:130-8.

188

Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, et al. Estrogen plus progestin and colorectal cancer in postmenopausal women. New Engl J Med 2004;350:991-1004.

189

Hulley S, Furberg C, Barrett-Connor E, et al. Noncardiovascular disease outcomes during 6.8 years of hormone therapy. Heart and estrogen/progestin replacement study follow-up (HERS II). JAMA 2002;288:58-66.

190

Marjoribanks J, Farquhar C, Roberts H, Lethaby A. Long term hormone therapy for perimenopausal and postmenopausal women. Cochrane Databaes of Systematic Reviews 2012;7:CD004143.

191

Solimando R, Bazzoli F, Ricciardiello L. Chemoprevention of colorectal cancer: a role for ursodeoxycholic acid, folate and hormone replacement treatment? Best Practice & Research Clinical Gastroenterology 2011;25:555-68.

192

Holly EA, Cress RD, Ahn DK. Cutaneous melanoma in women: ovulatory life, menopause, and use of exogenous estrogens. Cancer Epidemiol Biomarkers Prev 1994;3:661-8.

193

Koomen E, Joosse A, Herings R, Casparie M, Guchelaar H, Nijsten T. Estrogens, oral contraceptives and hormonal replacement therapy increase the incidence of cutaneous melanoma: a population-based case-control study. Annals of Oncology 2009;20:358-64.

194

Adami H, Persson I, Hoover R, Schairer C, Bergkvist L. Risk of cancer in women receiving hormone replacement therapy. Int J Cancer 1989;44:833-9.

195

Holman C, Armstrong B, Heenan P. Cutaneous malignant melanoma in women: exogenous sex hormones and reproductive factors. Br J Cancer 1984;50:673-80.

196

Osterlind A, Tucker M, Stone B, Jensen O. The Danish case-control study of cutaneous malignant melanoma. III. Hormonal and reproductive factors in women. Int J Cancer 1988;42:821-4.

197

Gandini S, Iodice S, Koomen E, Di Pietro A, Sera F, Caini S. Hormonal and reproductive factors in relation to melanoma in women: current review and meta-analysis. European Journal of Cancer 2011;47:2607-17.

198

Tang J, Spaunhurst K, Chlebowski R, et al. Menopausal hormone therapy and risks of melanoma and nonmelanoma skin cancers: Women's Health Initiative randomized trials. J Natl Cancer Inst 2011;103.

199

MacKie R, Bray C. Hormone replacement therapy after surgery for stage 1 or 2 cutaneous melanoma. British Journal of Cancer 2004;90:770-2.

200

DeSantis C, Siegel R, Bandi P, Jemal A. Breast cancer statistics, 2011. CA Cancer J Clin 2011;61:409-18.

201

Chen W, Manson J, Hankinson S, et al. Unopposed estrogen therapy and the risk of invasive breast cancer. Arch Intern Med 2006;166:1027-32.

202

Saxena T, Lee E, Henderson K, et al. Menopausal hormone therapy and subsequent risk of specific invasive breast cancer subtypes in the California Teachers Study. Cancer Epidemiol Biomarkers Prev 2010;19:2366-78.

203

Breast cancer and hormone-replacement therapy in the Million Women Study. The Million Women Study Collaborators. Lancet 2003;362:419-27.

204

Effects of conjugated equine estrogen in postmenopausal women with hysterectomy. The Women's Health Initiative Steering Committee. JAMA 2004;291:1701-12.

205

LaCroix A, Chlebowski R, Manson J, et al. Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy. A randomized controlled trial. JAMA 2011;305:1305-14.

206

Chlebowski RT, Kuller LH, Prentice RL, et al. Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med 2009;360:573-87.

207

Prentice RL, Chlebowski RT, Stefanick ML, et al. Estrogen plus progestin therapy and breast cancer in recently postmenopausal women. Am J Epidemiol 2008;167:1207-16.

208

Beral V, Reeves G, Bull D, Green J. Breast cancer risk in relation to the interval between menopause and starting hormone therapy. J Natl Cancer Inst 2011;103:296-305.

209

Fournier A, Mesrine S, Boutron-Ruault M, Clavel-Chapelon F. Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks? J Clin Oncol 2009;27:5138-43.

210

Jordan V, Ford L. Paradoxical clinical effect of estrogen on breast cancer risk: a "new" biology of estrogen-induced apoptosis. Cancer Prev Res 2011;4:633-7.

211

Flesch-Janys D, Slanger T, Mutschelknauss E, et al. Risk of different histological types of postmenopausal breast cancer by type and regimen of menopausal hormone therapy. Int J Cancer 2008;123:933-41.

212

Reeves G, Beral V, Green J, Gathani T, Bull D. Hormonal therapy for menopause and breast-cancer risk by histological type: a cohort study and meta-analysis. Lancet Oncol 2006;11:910-8.

213

Panjari M, Bell R, Lijovic M, et al. The relationship between hormone therapy use at the time of diagnosis of breast cancer and tumor characteristics. Horm Cancer 2010;1:93-9.

214

Cerne J, Frkovic-Grazio S, Gersak K. Breast tumor characteristics in hormone replacement therapy users. Pathol Oncol Res 2011;17:917-23.

215

Chlebowski R, Hendrix S, Langer R, et al. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women. The Women's Health Initiative Randomized Trial. JAMA 2003;289:3243-53.

216

Durna E, Wren B, Heller G, Leader L, Sjoblom P, Eden J. Hormone replacement therapy after a diagnosis of breast cancer: cancer recurrence and mortality. MJA 2002;177:347-51.

217

O'Meara E, Rossing M, Daling J, Elmore J, Barlow W, Weiss N. Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality. J Natl Cancer Inst 2001;93:754-62.

218

Holmberg L, Anderson H. HABITS (hormonal replacement therapy after breast cancer-is it safe?), a randomized comparison: trial stopped. Lancet 2004;363:453-5.

219

Holmberg L, Iversen O, Rudenstam C, et al. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors. J Natl Cancer Inst 2008;100:475-82.

220

Fahlen M, Fornander T, Johansson H, et al. Hormone replacement therapy after breast cancer: 10 year follow up of the Stockholm randomized trial. Eur J Cancer 2012.

221

von Schoultz E, Rutqvist L. Menopausal hormone therapy after breast cancer: the Stockholm randomized trial. J Natl Cancer Inst 2005;97:533-5.

222

Antoine C, Liebens F, Carly B, Pastijn A, Neusy S, Rozenberg S. Safety of hormone therapy after breast cancer: a qualitative systematic review. Human Reproduction 2007;22:616-22.

223

Creasman W, Henderson D, Hinshaw W, Clarke-Pearson D. Estrogen replacement therapy in the patient treated for endometrial cancer. Obstet Gynecol 1986;67:326-30.

224

Lee R, Burke T, Park R. Estrogen replacement therapy following treatment for stage I endometrial carcinoma. Gynecol Oncol 1990;36:189-91.

225

Chapman J, DiSaia P, Osann K, Roth P, Gillotte D, Berman M. Estrogen replacement in surgical stage I and II endometrial cancer survivors. Am J Obstet Gynecol 1996;175:1195-200.

226

Barakat R, Bundy B, Spirtos N, Bell J, Mannel R. Randomized double-blind trial of estrogen replacement therapy versus placebo in stage I or II endometrial cancer: a Gynecologic Oncology Group study. J Clin Oncol 2006;24:587-92.

227

Heaps JM, Nieberg RK, Berek JS. Malignant neoplasms arising in endometriosis. Obstet Gynecol 1990;75:1023-8.

228

Reimnitz C, Brand E, Nieberg RK, Hacker NF. Malignancy arising in endometriosis associated with unopposed estrogen replacement. Obstet Gynecol 1988;71:444-7.

229

Van Erpecum K, Van Berge Henegouwen G, Verschoor L, Stoelwinder B, Willekens F. Different hepatobiliary effects of oral and transdermal estradiol in postmenopausal women. Gastroenterology 1991;100:482-8