14,408
Views
15
CrossRef citations to date
0
Altmetric
Review

Evidence on the use of progesterone in menopausal hormone therapy

Pages 346-354 | Received 15 Jan 2018, Accepted 17 Mar 2018, Published online: 09 Apr 2018

Abstract

A need exists for a regulatory agency-approved hormone therapy (HT) with naturally occurring hormones combining progesterone (P4) and estradiol (E2), since no single product contains both endogenous hormones. Many women choose HT with P4 and millions of women around the world are using unapproved, poorly regulated compounded HT. The use of natural P4 in HT results, for the most part, in favorable outcomes without deleterious effects, as shown in clinical studies of postmenopausal women. Importantly, P4 used in HT prevents endometrial hyperplasia from estrogens while helping relieve vasomotor symptoms and improving quality-of-life measures. Additionally, risk of venous thromboembolism and breast cancer does not appear to increase with use of P4 plus estrogens as shown with synthetic progestins plus estrogens in large observations studies, and no detrimental effects of P4 in HT have been found on outcomes related to cardiovascular disease or cognition. A regulatory agency-approved HT with naturally occurring E2/P4 could be an option for the millions of women who desire a bioidentical product and/or are exposed to potential risks of inadequately studied and under-regulated compounded HT.

Introduction

The goal of hormone therapy (HT) is to relieve menopausal women of symptoms such as hot flushes, night sweats, dyspareunia, and vaginal dryness. When prescribing HT, either natural progesterone (P4) or a synthetic progestogen (progestin) is required for endometrial protectionCitation1. Observational data suggest a safer profile of P4 versus progestins with respect to cardiovascular and breast cancer outcomesCitation2–6.

A high demand for HT containing P4 is shown by the estimated annual prescriptions of up to 21 million products containing natural P4, representing the most prescribed form of HT in the USCitation7. Since no single FDA-approved formulation contains E2 and P4 combined, most of these (up to 18 million prescriptions) are for non-FDA-approved compounded hormone therapyCitation7. Given that compounded HT is not adequately studied and under-regulated, such high exposure of menopausal women to compounded HT is concerning since certain risks may be associated with compounded HT. Recent evidence suggests an increased risk of endometrial hyperplasia and cancer in users of compounded HTCitation8–10.

A regulatory agency-approved menopausal HT with E2 and P4 could help fill the need for women to be safely treated with a natural or bioidentical product for their bothersome vasomotor symptoms (VMS)Citation11,Citation12, shown to negatively impact their quality of life, sleep, and work productivityCitation11,Citation13. This paper reviews historical and more recent clinical data supporting the use of P4, by various routes of administration, in combination with estrogens (E2 or conjugated equine estrogens (CEE)) as menopausal HT.

Methodology

PubMed was searched since its inception for clinical studies on HT containing natural P4. Relevant studies, including randomized, controlled trials of estrogens with P4, and large observational studies comparing different progestins with P4 were the focus of this article. Studies on transdermal P4 were not included since no adequately designed trials have shown it can deliver adequate P4 for endometrial protectionCitation14.

Keywords such as bone, breast, breast cancer, cardiovascular (CVD), cognition, endometrium, hot flush, lipids, mammographic breast density, myocardial infarction, quality of life, sleep, stroke, vasomotor symptoms, and venous thromboembolism (VTE) were used in conjunction with P4 and menopause. Limits were the English language, keywords within the title or abstract, clinical trials, and review articles. Bibliographies of relevant review articles were also reviewed.

Endometrial safety

Progesterone and progestins have a pronounced effect on endometrial morphology, with different potencies and dosages inducing qualitative and quantitative morphologic changes. Randomized, controlled trials show progestogens at the right dosage prevent endometrial hyperplasiaCitation15,Citation16 and endometrial cancerCitation17; however, oral P4 has not been thought to protect the endometrium as well as progestins, based on European observational data (almost exclusively from France)Citation18,Citation19. Although an increased risk of endometrial cancer was found with current useCitation18,Citation19 or >5 years useCitation19 of estrogens plus P4, but not with use of estrogens plus synthetic progestins, in the Etude Epidémiologique auprès de femmes de l’Education Nationale (E3N) and European Prospective Investigation into Cancer and Nutrition (EPIC) trials, definitive conclusions are difficult to make from these reports since risk was not analyzed by dose or timing (i.e. cyclic vs. continuous) of progestogen use.

Effectiveness of P4 in protecting the endometrium was first supported by proof-of-concept studies showing a dose-dependent inhibition of endometrial proliferation in women taking various doses of cyclic P4 with estrogensCitation20,Citation21. In the Postmenopausal Estrogen/Progestin Interventions (PEPI) trial (n = 118–120 in each group), cyclic 200 mg P4 (12 days) plus 0.625 mg CEE provided endometrial hyperplasia rates similar to those of placeboCitation15. Based on these dataCitation15, the only FDA-approved P4 regimen for endometrial protection is cyclic 200 mg with CEECitation22. However, the PEPI study was not powered adequately to detect endometrial hyperplasia or cancer risk. To demonstrate adequate endometrial protection (≤1% with <4% upper bound of 95% confidence interval), regulatory agencies world-wide currently recommend a 1-year clinical trial with at least 300 subjectsCitation23,Citation24.

A preliminary report of the phase-3 REPLENISH trial showed multiple doses of the investigational, oral, continuous combined, solubilized E2/P4 (TX-001HR, TherapeuticsMD, Boca Raton, FL, USA: 1 mg/100 mg, 0.5 mg/100 mg, 0.5 mg/50 mg, 0.25 mg/50 mg) resulting in no cases of endometrial hyperplasia or cancer (n = 274–306 per group) after 12 monthsCitation25. This was the first sufficiently powered trial to identify oral P4 doses that antagonized the endometrial effects of different E2 doses (1 mg, 0.5 mg, or 0.25 mg)Citation25.

Vaginal P4 has also been given for endometrial protection, as it has been hypothesized to have better access to endometrial tissues through the ‘first uterine pass effect’ and possibly fewer adverse effectsCitation26,Citation27. While a significantly higher endometrial-to-serum P4 ratio with vaginal P4 compared with intramuscular P4 injection has been shown in a small studyCitation27, no studies large enough adequately to evaluate endometrial protection with vaginal P4 have been conducted. The Early versus Late Intervention Trial with Estradiol (ELITE), the only 5-year, randomized trial comparing 1 mg/day oral E2 or placebo with 45 mg/day of cyclical vaginal P4 gel (10 days/cycle)Citation28, found no significant difference in E2/P4 vs. placebo for endometrial cancer (assessed as an adverse event; reported by Stute and colleaguesCitation14). Some smaller studies have shown vaginal P4 to prevent endometrial hyperplasia when given cyclically or continuously as a gel with oral or transdermal estrogensCitation29–31, or as a vaginal ringCitation32, but not as a vaginal P4 creamCitation33.

In conclusion, the REPLENISH trial is the first and only adequately designed trial showing oral, continuous P4 to prevent endometrial hyperplasia when given at the right ratio.

Uterine bleeding

Uterine bleeding has been related to an imbalance between angiogenic and antiangiogenic factors in the endometrial vasculatureCitation34. Such alterations could induce proangiogenic activity in the endometrial capillary plexus, with consequent aberrant vasculogenesis that may result in irregular endometrial bleedingCitation34. In vitro studies have shown that the angiogenic imbalance is less pronounced with P4 than with synthetic progestogensCitation35,Citation36. In addition, clinical studies demonstrated that the ratio of estrogen to progestogen may play a role in determining the risk of vaginal bleedingCitation37. However, few head-to-head studies have evaluated the bleeding profiles of various HT regimens of estrogens with progesterone compared with HT containing other progestogens.

In a 9-month study of women taking CEE randomized to either micronized 200 mg/day P4 (n = 89) or 5 mg/day medroxyprogesterone acetate (MPA; n = 93), more women taking P4 versus MPA had amenorrhea (p = 0.001), and women taking P4 had fewer bleeding days (p = 0.01) and less blood flow (p < 0.001)Citation38. Significantly less vaginal bleeding was seen with oral P4 than MPA (p < 0.01) in a small study (n = 23) of alternating 2-week cycles of CEE alone (0.625 mg) or CEE plus progestogen (cyclic MPA 5 mg/day or P4 200 mg/day)Citation39. P4 also achieved a high rate of amenorrhea (91.6%) in a study of 98 women treated for 6 months with transdermal E2 1.5 mg/day plus oral P4 100 mg/day on days 1–21 or 25Citation40. Preliminary data from the REPLENISH trial showed that several continuous combined doses of E2/P4 (TX-001HR) resulted in high rates of amenorrhea that improved over time (56–73% from cycles 1–13)Citation41. In contrast, Di Carlo and colleaguesCitation42 reported that sequential HT containing progesterone had more episodes of irregular bleeding than with sequential HT containing synthetic progestins. The same investigators reported that users of HT with sequential vaginal progesterone had more regular bleeding episodes, fewer irregular bleeding episodes and more amenorrhea than users of HT with sequential oral progesteroneCitation43.

Menopausal symptoms

Although progestogens are included in HT to provide endometrial protection, some progestogens, including P4 at high dosage, have been shown to provide relief of VMS when given without an estrogen. A 12-week, randomized study in postmenopausal women (n = 133) found that daily VMS frequency improved significantly more with oral 300 mg/day P4 alone than with placeboCitation44.

Data from the REPLENISH trial show that, for the most part, the frequency of moderate-to-severe VMS significantly improved from week 3 to 12 for E2/P4 doses of 1 mg/100 mg, 0.5 mg/100 mg, and 0.25 mg/50 mg, and at weeks 6–12 for 0.5 mg/50 mg compared with placebo (p < 0.05)Citation25. Significant reductions in VMS severity were observed at weeks 3–12 for 1 mg E2/100 mg P4 and 0.5 mg E2/100 mg P4, and at most weeks between 6 and 12 for 0.5 mg E2/50 mg P4 and 0.25 mg E2/50 mg P4Citation25.

Head-to-head studies, including the PEPI trial, show similar relief of hot flushes with CEE plus cyclic or continuous P4 or MPACitation45–47.

Quality of life

One study suggests positive effects on quality-of-life outcomes with HT regimens containing 0.625 mg CEE with MPA 5 mg/day or micronized P4 200 mg/day (n = 182) for 9 monthsCitation38. In the REPLENISH trial, E2/P4 (1 mg/100 mg, 0.5 mg/100 mg, 0.5 mg/50 mg) significantly improved quality of life as measured by MENQOL at 12 weeks and 12 monthsCitation48.

Sleep

Progesterone is converted to allopregnanolone in the mammalian brain, which has been shown to induce GABAergic effects and promote sleep. Sleep parameters improved in several trials of P4-containing HT, with some showing better improvements when P4 vs. MPA was used. A polysomnography study of 21 menopausal women found that 0.625 mg CEE with cyclic 200 mg/day P4, but not with cyclic 5 mg/day MPA, significantly improved sleep efficiency and decreased time spent awake after sleep onset from baseline, with similar improvements in other subjective sleep indices in both groupsCitation46. In another small, cross-over, polysomnography study (n = 10), 300 mg P4 alone for 21 days significantly reduced time spent awake and increased rapid eye movement sleep in the first third of the night vs. placeboCitation49. Continuous CEE 0.3 mg/day with P4 100 mg also provided significantly better sleep outcomes than CEE/MPA 2.5 mgCitation47. The REPLENISH trial showed improved sleep parameters (Medical Outcomes Study – Sleep Scale) with various continuous E2/P4 doses at week 12 and months 6 and 12Citation50, consistent with a reduction in hot flushes.

Bone

Effects of HT containing P4 or MPA appear to be similar on bone mineral density (BMD). This was shown for the spine and hip in the PEPI trial with CEE plus cyclic MPA or cyclic P4 vs. placebo at 3 yearsCitation51, and in a smaller, 3-year, randomized, open-label study (n = 60) of transdermal E2 gel 0.75 or 1.5 mg/day with P4 100 mg/day or MPA 2 mg/day (25 days per month)Citation52. Other studies show BMD improvements with estrogens plus cyclic or intermittent P4 useCitation53–55. In a KEEPS substudy (n = 76), oral CEE or transdermal E2 patch with cyclic oral micronized P4 for 4 years prevented the cortical volumetric BMD decline and cortical porosity increase at the distal radius seen with placeboCitation53.

Breast

Effects of HT on breast cell proliferation in premenopausal and postmenopausal women have been evaluated. PremenopausalCitation56 and postmenopausalCitation57 women scheduled for breast surgery, for presumably benign lesions, were randomized to transdermal gel containing 1.5 mg E2, 25 mg P4, combined E2/P4, or placebo applied daily to the breast for approximately 14 days before surgery. For the most part, in both studies, proliferation markers (mitotic index and proliferating cell nuclear antigen index) increased with E2 vs. placebo, decreased with P4 vs. placebo, and were comparable between E2/P4 and P4 or placeboCitation56,Citation57. In another study of postmenopausal women (n = 77) taking two cycles of 0.625 mg CEE plus cyclic 5 mg MPA or 1.5 mg E2 gel plus cyclic oral 200 mg P4, breast cell proliferation significantly increased from baseline with CEE/MPA, but not with E2/P4Citation58,Citation59.

New-onset breast tenderness after initiation of HT containing a synthetic progestin has been associated with increased mammographic density and breast cancer riskCitation60,Citation61. A KEEPS substudy (n = 116) that evaluated breast pain over 4 years reported no significant increases in pain over time and no differences in ‘worst’ pain or ‘average’ pain at any time point with oral 0.45 mg/day CEE or transdermal 50 μg/day E2 (both given with cyclic 200 mg/day P4) or placeboCitation62. In a small study (n = 23), women were given placebo, then cyclic 5 mg/day MPA or 200 mg/day P4 alone followed by 2 months of 0.625 mg/day CEE with progestogen in the last 2 weeks/monthCitation39. While neither progestogen was associated with breast tenderness when given without CEE, breast tenderness was greater with CEE/MPA than with CEE/P4Citation39.

When mammographic breast density was evaluated in the PEPI trial, women randomized to CEE alone, CEE/cyclic MPA, CEE/continuous MPA, or CEE/cyclic P4 had 0.9%, 4.6%, 4.4%, and 3.1% increases in breast density, respectively, compared with placebo (−0.4%; p < 0.001 for combined progestogen group vs. placebo, with no difference among progestogen groups)Citation63. A smaller Swedish study showed 18.9% of patients taking oral CEE 0.625 mg plus cyclic MPA 5 mg (n = 37) had an increase in BI-RADS category of at least one level from baseline (p = 0.01 vs. baseline) compared with 6.3% of women taking percutaneous E2 gel 1.5 mg plus oral P4 200 mg (n = 32; p = NS)Citation59.

Large observational studies have compared effects of HT containing progestins or P4 on breast cancer risk. In two publications from European studies (n = 54 548 (E3N cohort) and 80 377 (E3N-EPIC cohort)), an increase in breast cancer risk was found in women who currently took estrogen plus synthetic progestogens but not estrogens plus P4 (risk ratios reported in ; one studyCitation64 combined P4 with dydrogesterone)Citation64–66. Breast cancer risk by duration of HT use is summarized in . In the most recent analysis of E3N, increased risk of breast cancer was found with current use of estrogen plus P4/dydrogesterone for >5 years, and current use of estrogen plus synthetic progestogens for ≤5 years and >5 years with a greater magnitude than with P4Citation64. In addition, current use of estrogen plus P4/dydrogesterone for ≤5 or >5 years in women with a body mass index (BMI) < 25 kg/m2, or estrogen plus synthetic progestogens for ≤5 or >5 years independent of BMI, had an increased breast cancer risk, but with a greater magnitude in women taking synthetic progestinsCitation64. Since P4 and dydrogesterone were combined in the analysis, making definitive conclusions about P4 is not possibleCitation64. Finally, while risk with estrogens plus P4 disappeared in past users, it remained in users of estrogens plus synthetic progestinsCitation64. In the initial report of the E3N cohortCitation66, no significantly increased risk was found at 2-year increments for estrogens plus progesterone or dydrogesterone, but increased risk was consistently seen with estrogens plus synthetic progestins over the same time periods. Analysis by duration of use in the initial combined E3N–EPIC cohort found an increase in risk with transdermal estrogens plus synthetic progestins vs. P4 for the first 4 years of useCitation65. While an increased risk was shown with 2–4 years or >4 years of oral estrogens plus synthetic progestin use, risk with oral estrogens plus P4 was not analyzed due to the small number of women taking itCitation65.

Table 1. Risk of breast cancer and venous thromboembolism with hormone therapy containing progesterone or progestins in large observational studies.

Table 2. Risk of breast cancer with hormone therapy containing progesterone or progestins by duration of use in large observational studies.

In a French case–control study (n = 1232), current use of estrogen plus P4 did not increase breast cancer risk, while estrogen plus synthetic progestogens significantly elevated the risk, particularly testosterone derivatives ()Citation2. A systematic review and meta-analysis of studies (n = 14 studies) of women using E2 with or without progestogen showed no association between E2 only and risk of breast cancer; however, risk was significantly elevated when E2 was combined with MPA, norethisterone acetate, or levonorgestrel, but not with P4 or dydrogesteroneCitation67.

Cardiovascular disease

Venous thromboembolism

Micronized P4 does not appear to increase the risk of VTE in postmenopausal women. Both the French case–control ESTHER (Estrogen and Thromboembolism Risk) and French E3N cohort studies reported that concomitant use of P4 or pregnane derivatives with estrogens did not increase the VTE risk in postmenopausal women, while concomitant use of norpregnane derivatives did ()Citation3,Citation4. Similarly, the Menopause, Estrogen, and Veins cohort study suggested no additional risk of P4 on recurrent VTE in women taking transdermal estrogens, whereas norpregnane derivatives with transdermal estrogens were associated with significantly increased risk ()Citation5. In the French Study of NorpregnAnes on Coagulation, concentrations of activated protein C (APC) sensitivity and prothrombin fragment 1 + 2 were similar with transdermal estrogens plus P4 vs. non-users, while women who used concomitant norpregnane derivatives were less sensitive to APC and had higher prothrombin fragment 1 + 2 than non-usersCitation68.

Cardiovascular disease outcomes

Only two randomized, controlled studies examined the effects of HT with P4 on cardiovascular outcomes. ELITE tested the hormone-timing hypothesis, that early but not later initiation of HT would prevent subclinical atherosclerosis progressionCitation28. Healthy postmenopausal women (n = 643) stratified by time since menopause (<6 years or ≥10 years) were randomized to 1 mg/day oral E2 or placebo; women with a uterus received 45 mg/day vaginal P4 gel or placebo applied cyclically for 10 days/cycleCitation28. At a median 5-year follow-up, carotid artery intima-media thickness (CIMT) progression was significantly lower with E2 than with placebo only in early postmenopause (p = 0.008) but not later in menopause (p = 0.29), and the effects of E2 alone and E2/P4 were similarCitation28. In KEEPS, women (n = 727) in early menopause (within 6 and 36 months) were randomized to continuous oral CEE 0.45 mg/day or transdermal E2 50 μg/day patch (plus oral P4 200 mg/day for 12 days/month)Citation69. Vascular imaging showed comparable increases in both CIMT and coronary artery calcium scores over 4-year follow-up between groupsCitation69.

Progesterone also appears to be beneficial in women who have myocardial ischemia. In a cross-over trial of women with stable exertional angina, myocardial ischemia, coronary artery disease and/or previous myocardial infarction using a treadmill, 4 weeks of E2 (2 mg/day) improved their exercise time to myocardial ischemia, which was further improved after receiving 12 days transvaginal P4 gel (90 mg every other day) but not oral MPA (10 mg/day)Citation70.

Lastly, a French case–control study found an increased risk of ischemic stroke in women who used oral but not transdermal estradiol, and an increased risk with concomitant use of norpregnane derivatives, but not P4, pregnane derivatives, or nortestosterone derivatives ()Citation6.

Lipids

Well-established effects of oral estrogens on lipids are increases in high density lipoprotein (HDL) cholesterol decreases in total cholesterol and low density lipoprotein (LDL) cholesterol, and increase in triglycerides, while these effects are not apparent with transdermal estrogens. Addition of P4 has been shown to enhance or have neutral effects on the beneficial changes of oral estrogens on lipids.

In the randomized, controlled PEPI trial, all active treatment groups increased HDL cholesterol relative to a decline in the placebo group after 3 years, and HDL cholesterol improvements were significantly greater in the CEE and CEE/P4 groups vs. the CEE/MPA groupsCitation71. All active regimens decreased LDL cholesterol and increased triglycerides vs. placeboCitation71. Similarly in another study, 0.3 mg CEE plus P4 100 mg/day resulted in higher HDL cholesterol and triglycerides, lower LDL cholesterol and non-HDL cholesterol, and lower total cholesterol/HDL cholesterol ratio compared with baseline (all, p < 0.05)Citation72. In a study of transdermal E2 gel alone or with cyclic oral P4 (200 mg), or 0.625 mg CEE alone or with cyclic oral P4, adding P4 to CEE enhanced the elevated HDL cholesterol level, preserved the decrease in LDL cholesterol, and lowered the elevation in triglycerides, while P4 added to the transdermal E2 regimen prevented an increase in HDL cholesterolCitation73.

In other studies, neutral or beneficial effects of P4 were observed. In an open-label, randomized trial (n = 107) of 12 cycles, CEE/dydrogesterone was associated with a significant increase in triglycerides, while CEE/P4 had a neutral effect; both therapies significantly elevated HDL cholesterol and lowered LDL cholesterol levelsCitation74. Two other randomized, cross-over trials showed a neutral or beneficial effect of P4 in non-oral estrogen regimens in early postmenopausal womenCitation75,Citation76 and a transient decrease in HDL cholesterol was seen with a levonorgestrel-containing intrauterine device, but not with oral or vaginal P4Citation77.

Endothelial function

Several trials have confirmed that P4 has no adverse vascular effects. In a KEEPS substudy, recently postmenopausal women without CVD at baseline who received oral CEE 0.45 mg/day or transdermal E2 50 μg/day combined with cyclic oral P4 (200 mg/day) had no significant change in reactive hyperemia index (used to assess endothelial function) with treatmentCitation78. A study of forearm blood flow in response to brachial artery infusions of vasodilators showed that administration of P4 either alone or with E2 had no detrimental effects in postmenopausal women without cardiovascular risk factorsCitation79. In a 12-week, cross-over study of women receiving oral CEE (0.625 mg) or transdermal E2 (50 μg) combined with continuous oral P4 (100 mg), CEE/P4 significantly decreased E-selectin, P-selectin, and intercellular adhesion molecule (ICAM-1) and increased C-reactive protein, while transdermal E2/P4 only decreased ICAM-1 levelsCitation80. Furthermore, endothelial function was improved and inflammation markers decreased in recently menopausal women who received 3 months of transdermal E2 (50 μg) plus cyclic oral P4 (200 mg) compared with non-usersCitation81.

Expression of vascular cell adhesion molecule (VCAM-1) has been shown to be one of the earliest events that occur in the atherogenic processCitation82. In human umbilical vein endothelial cells, pretreatment with P4, but not MPA, inhibited TNFα-induced increases in VCAM-1 protein expression and mRNA accumulationCitation82. Similarly, significant reductions in IL-1β-stimulated VCAM-1 and ICAM-1 expression were observed when female human iliac artery endothelial cells were pretreated with E2/P4 (but not either alone)Citation83.

Cognition

Progesterone combined with estrogens does not appear to have any deleterious or beneficial effects on cognition in postmenopausal women, as shown by the limited available evidence. The timing effect of oral E2 (1 mg) vs. placebo on cognition was evaluated in the ELITE-Cog trial (n = 567); women with a uterus also received 4% vaginal P4 gel (45 mg) or placebo 10 days/cycleCitation84. Compared with placebo, E2/P4 had no negative effects on verbal memory, executive functions, or global cognition after 2.5 years of treatment in women who initiated treatment within 6 years or ≥10 years after menopauseCitation84. Similarly, the 4-year KEEPS-Cognitive and Affective Study found no significant difference in cognitive variables between recently menopausal women who received oral CEE (0.45 mg) or transdermal E2 (50 μg), both combined with cyclic oral P4 (200 mg), or placeboCitation85. In a small, 12-week trial of recently menopausal women who received 0.625 mg CEE plus cyclic 10 mg MPA (n = 9), 200 mg P4 (n = 8), or placebo (n = 7), only CEE/P4 provided significant improvements from baseline in delayed verbal memory and working memory testsCitation86.

Summary

A need exists for regulatory agency-approved HT with naturally occurring hormones such as P4 and E2, as use of compounded HT is on the rise, exposing millions of women to potential risk. Existing evidence from clinical studies on the use of natural P4 in HT, for the most part, shows favorable outcomes, without deleterious effects. Both E2 and CEE combined with P4 relieve vasomotor symptoms and improve measures of quality of life. Additionally, HT with P4 has been shown to prevent endometrial hyperplasia from estrogens, and large observational studies show risk of VTE and breast cancer does not increase with use of P4 with estrogens. Further, no detrimental effects of P4 on outcomes related to cardiovascular disease or cognition have been reported. A recent randomized, controlled study, REPLENISH, showed that an oral, combined E2/P4 capsule at various doses improved hot flushes without endometrial hyperplasia in menopausal women. A regulatory agency-approved HT with naturally occurring P4 and E2 could be an option for the millions of women exposed to potential risks with inadequately studied compounded HT.

Conflict of interest

Dr Mirkin is an employee of TherapeuticsMD.

Acknowledgements

The medical writing assistance provided by Kathleen Ohleth, PhD, CMPP and Dominique Verlaan, PhD, CMPP of Precise Publications, LLC is greatly appreciated.

Additional information

Funding

Medical writing assistance provided by Kathleen Ohleth, PhD, CMPP and Dominique Verlaan, PhD, CMPP of Precise Publications, LLC was supported by TherapeuticsMD.

References

  • The NAMS 2017 Hormone Therapy Position Statement Advisory Panel. The 2017 hormone therapy position statement of the North American menopause society. Menopause 2017;24:728–53
  • Cordina-Duverger E, Truong T, Anger A, et al. Risk of breast cancer by type of menopausal hormone therapy: a case-control study among post-menopausal women in France. PLoS One 2013;8:e78016
  • Canonico M, Fournier A, Carcaillon L, et al. Postmenopausal hormone therapy and risk of idiopathic venous thromboembolism: results from the E3N cohort study. Arterioscler Thromb Vasc Biol 2010;30:340–5
  • Canonico M, Oger E, Plu-Bureau G, et al. Hormone therapy and venous thromboembolism among postmenopausal women: impact of the route of estrogen administration and progestogens: the ESTHER study. Circulation 2007;115:840–5
  • Olie V, Plu-Bureau G, Conard J, Horellou MH, Canonico M, Scarabin PY. Hormone therapy and recurrence of venous thromboembolism among postmenopausal women. Menopause 2011;18:488–93
  • Canonico M, Carcaillon L, Plu-Bureau G, et al. Postmenopausal hormone therapy and risk of stroke: impact of the route of estrogen administration and type of progestogen. Stroke 2016;47:1734–41
  • TherapeuticsMD. TXMD Overview November 2017. Available from: https://ir.therapeuticsmd.com/static-files/c2e7f9d1-d150-4b4d-8703-e4f5c362169c [last accessed 7 Jan 2018]
  • Eden JA, Hacker NF, Fortune M. Three cases of endometrial cancer associated with bioidentical hormone replacement therapy. Med J Aust 2007;187:244–5
  • Davis R, Batur P, Thacker HL. Risks and effectiveness of compounded bioidentical hormone therapy: a case series. J Womens Health (Larchmt) 2014;23:642–8
  • Dezman VL, Gersak MZ, Gersak K. Two case of atypical endometrial hyperplasia associated with “bioidentical” hormone replacement therapy: IGCS-0084 uterine cancer, including sarcoma. Int J Gynecol Cancer 2015;25(Suppl 1):71
  • Blumel JE, Chedraui P, Baron G, et al. A large multinational study of vasomotor symptom prevalence, duration, and impact on quality of life in middle-aged women. Menopause 2011;18:778–85
  • Hunter MS, Gentry-Maharaj A, Ryan A, et al. Prevalence, frequency and problem rating of hot flushes persist in older postmenopausal women: impact of age, body mass index, hysterectomy, hormone therapy use, lifestyle and mood in a cross-sectional cohort study of 10,418 British women aged 54-65. BJOG 2012;119:40–50
  • Whiteley J, Wagner JS, Bushmakin A, Kopenhafer L, Dibonaventura M, Racketa J. Impact of the severity of vasomotor symptoms on health status, resource use, and productivity. Menopause 2013;20:518–24
  • Stute P, Neulen J, Wildt L. The impact of micronized progesterone on the endometrium: a systematic review. Climacteric 2016;19:316–28
  • Writing Group for the PEPI Trial. Effects of hormone replacement therapy on endometrial histology in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) trial. JAMA 1996;275:370–5
  • Pickar JH, Yeh IT, Wheeler JE, Cunnane MF, Speroff L. Endometrial effects of lower doses of conjugated equine estrogens and medroxyprogesterone acetate: two-year substudy results. Fertil Steril 2003;80:1234–40
  • Chlebowski RT, Anderson GL, Sarto GE, et al. Continuous combined estrogen plus progestin and endometrial cancer: the women’s health initiative randomized trial. J Natl Cancer Inst 2016;108:dvj350
  • Allen NE, Tsilidis KK, Key TJ, et al. Menopausal hormone therapy and risk of endometrial carcinoma among postmenopausal women in the European prospective investigation into cancer and nutrition. Am J Epidemiol 2010;172:1394–403
  • Fournier A, Dossus L, Mesrine S, et al. Risks of endometrial cancer associated with different hormone replacement therapies in the E3N cohort, 1992-2008. Am J Epidemiol 2014;180:508–17
  • Moyer DL, Felix JC, Kurman RJ, Cuffie CA. Micronized progesterone regulation of the endometrial glandular cycling pool. Int J Gynecol Pathol 2001;20:374–9
  • Darj E, Nilsson S, Axelsson O, Hellberg D. Clinical and endometrial effects of oestradiol and progesterone in post-menopausal women. Maturitas 1991;13:109–15
  • Prometrium® (Progesterone, USP) Prescribing Information. Catalent Pharma Solutions. St. Petersburg, FL. 2011
  • US Department of Health and Human Services (FDA). Guidance for Industry: Estrogen and estrogen/progestin drug products to treat vasomotor symptoms and vulvar and vaginal atrophy symptoms–recommendations for clinical evaluation. January 2003. Available from: http://www.fda.gov/downloads/Drugs/DrugSafety/informa-tionbyDrugClass/UCM135338.pdf [last accessed Aug 2015]
  • European Medicines Agency. Guideline on clinical investigation of medicinal products for hormone replacement therapy of oestrogen deficiency symptoms in postmenopausal women. October 2005. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003348.pdf [last accessed Dec 2016]
  • Lobo RA, Archer DF, Kagan R, et al. A 17β-estradiol-progesterone oral capsule for vasomotor symptoms in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2018, In press
  • Cicinelli E, de Ziegler D, Bulletti C, Matteo MG, Schonauer LM, Galantino P. Direct transport of progesterone from vagina to uterus. Obstet Gynecol 2000;95:403–6
  • Ficicioglu C, Gurbuz B, Tasdemir S, Yalti S, Canova H. High local endometrial effect of vaginal progesterone gel. Gynecol Endocrinol 2004;18:240–3
  • Hodis HN, Mack WJ, Henderson VW, et al. Vascular effects of early versus late postmenopausal treatment with estradiol. N Engl J Med 2016;374:1221–31
  • 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
  • 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
  • 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
  • Noe G, Sitruk-Ware R, Zegers-Hochschild F, et al. Endometrial effect of progesterone delivered by vaginal rings in estrogen-treated postmenopausal women. Climacteric 2010;13:433–41
  • Ferrero S, Gerbaldo D, Fulcheri E, Cristoforoni P. Vaginal micronized progesterone in continuous hormone replacement therapy. A prospective randomized study. Minerva Ginecol 2002;54:519–30
  • Mirkin S, Navarro F, Archer DF. Hormone therapy and endometrial angiogenesis. Climacteric 2003;6:273–7
  • Mirkin S, Archer DF. Effects of levonorgestrel, medroxyprogesterone acetate, norethindrone, progesterone, and 17beta-estradiol on thrombospondin-1 mRNA in Ishikawa cells. Fertil Steril 2004;82:220–2
  • Mirkin S, Wong BC, Archer DF. Effect of 17 beta-estradiol, progesterone, synthetic progestins, tibolone, and tibolone metabolites on vascular endothelial growth factor mRNA in breast cancer cells. Fertil Steril 2005;84:485–91
  • Mirkin S, Archer DF, Taylor HS, Pickar JH, Komm BS. Differential effects of menopausal therapies on the endometrium. Menopause 2014;21:899–908
  • Ryan N, Rosner A. Quality of life and costs associated with micronized progesterone and medroxyprogesterone acetate in hormone replacement therapy for nonhysterectomized, postmenopausal women. Clin Ther 2001;23:1099–115
  • Cummings JA, Brizendine L. Comparison of physical and emotional side effects of progesterone or medroxyprogesterone in early postmenopausal women. Menopause 2002;9:253–63
  • 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
  • Goldstein SR, Constantine G, Archer DF, et al. Effects of TX-001HR on uterine bleeding rates in menopausal women with vasomotor symptoms (S-18). 2017 NAMS Annual Meeting (Oct 11–14, 2017). Philadelphia, PA. 2017. Available from: http://www.menopause.org/docs/default-source/2017/2017-nams-scientific-and-poster-abs-tracts.pdf [last accessed 8 Dec 2017]
  • Di Carlo C, Sammartino A, Di Spiezio SA, et al. Bleeding patterns during continuous estradiol with different sequential progestogens therapy. Menopause 2005;12:520–5
  • Di Carlo C, Tommaselli GA, Gargano V, Savoia F, Bifulco G, Nappi C. Transdermal estradiol and oral or vaginal natural progesterone: bleeding patterns. Climacteric 2010;13:442–6
  • Hitchcock CL, Prior JC. Oral micronized progesterone for vasomotor symptoms–a placebo-controlled randomized trial in healthy postmenopausal women. Menopause 2012;19:886–93
  • Greendale GA, Reboussin BA, Hogan P, et al. Symptom relief and side effects of postmenopausal hormones: results from the Postmenopausal Estrogen/Progestin Interventions Trial. Obstet Gynecol 1998;92:982–8
  • Montplaisir J, Lorrain J, Denesle R, Petit D. Sleep in menopause: differential effects of two forms of hormone replacement therapy. Menopause 2001;8:10–16
  • Gambacciani M, Ciaponi M, Cappagli B, et al. Effects of low-dose, continuous combined hormone replacement therapy on sleep in symptomatic postmenopausal women. Maturitas 2005;50:91–7
  • Simon J, Kaunitz A, Kroll R, Graham S, Bernick B, Mirkin S. TX-001HR improved quality of life in postmenopausal women with vasomotor symptoms (S-19). Menopause 2017;24:1432
  • Schussler P, Kluge M, Yassouridis A, et al. Progesterone reduces wakefulness in sleep EEG and has no effect on cognition in healthy postmenopausal women. Psychoneuroendocrinology 2008;33:1124–31
  • Kagan R, Constantine G, Kaunitz A, Gasper G, Bernick B, Mirkin S. TX-001HR improved the Medical Outcomes Study-Sleep (MOS-Sleep) questionnaire in menopausal women with vasomotor symptoms (S-17). Menopause 2017;24:1431
  • Writing Group for the Pepi Trial. Effects of hormone therapy on bone mineral density: results from the postmenopausal estrogen/progestin interventions (PEPI) trial. JAMA 1996;276:1389–96
  • Sun A, Lin S, Yu W, et al. Percutaneous estrogen in prevention of early postmenopausal bone loss in Chinese women. Chin Med J (Engl) 2002;115:1790–5
  • Farr JN, Khosla S, Miyabara Y, Miller VM, Kearns AE. Effects of estrogen with micronized progesterone on cortical and trabecular bone mass and microstructure in recently postmenopausal women. J Clin Endocrinol Metab 2013;98:E249–57
  • 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
  • Riis BJ, Thomsen K, Strom V, Christiansen C. The effect of percutaneous estradiol and natural progesterone on postmenopausal bone loss. Am J Obstet Gynecol 1987;156:61–5
  • Chang KJ, Lee TT, Linares-Cruz G, Fournier S, de Lignieres B. Influences of percutaneous administration of estradiol and progesterone on human breast epithelial cell cycle in vivo. Fertil Steril 1995;63:785–91
  • Foidart JM, Colin C, Denoo X, et al. Estradiol and progesterone regulate the proliferation of human breast epithelial cells. Fertil Steril 1998;69:963–9
  • Murkes D, Conner P, Leifland K, et al. Effects of percutaneous estradiol-oral progesterone versus oral conjugated equine estrogens-medroxyprogesterone acetate on breast cell proliferation and bcl-2 protein in healthy women. Fertil Steril 2011;95:1188–91
  • Murkes D, Lalitkumar PG, Leifland K, Lundstrom E, Soderqvist G. Percutaneous estradiol/oral micronized progesterone has less-adverse effects and different gene regulations than oral conjugated equine estrogens/medroxyprogesterone acetate in the breasts of healthy women in vivo. Gynecol Endocrinol 2012;28(Suppl 2):12–15
  • Crandall CJ, Aragaki AK, Cauley JA, et al. Breast tenderness and breast cancer risk in the estrogen plus progestin and estrogen-alone women’s health initiative clinical trials. Breast Cancer Res Treat 2012;132:275–85
  • Crandall CJ, Aragaki AK, Cauley JA, et al. Breast tenderness after initiation of conjugated equine estrogens and mammographic density change. Breast Cancer Res Treat 2012;131:969–79
  • Files JA, Miller VM, Cha SS, Pruthi S. Effects of different hormone therapies on breast pain in recently postmenopausal women: findings from the Mayo Clinic KEEPS breast pain ancillary study. J Womens Health (Larchmt) 2014;23:801–5
  • Crandall CJ, Karlamangla A, Huang MH, Ursin G, Guan M, Greendale GA. Association of new-onset breast discomfort with an increase in mammographic density during hormone therapy. Arch Intern Med 2006;166:1578–84
  • Fournier A, Mesrine S, Dossus L, Boutron-Ruault MC, Clavel-Chapelon F, Chabbert-Buffet N. Risk of breast cancer after stopping menopausal hormone therapy in the E3N cohort. Breast Cancer Res Treat 2014;145:535–43
  • Fournier A, Berrino F, Riboli E, Avenel V, Clavel-Chapelon F. Breast cancer risk in relation to different types of hormone replacement therapy in the E3N-EPIC cohort. Int J Cancer 2005;114:448–54
  • Fournier A, Berrino F, Clavel-Chapelon F. Unequal risks for breast cancer associated with different hormone replacement therapies: results from the E3N cohort study. Breast Cancer Res Treat 2008;107:103–11
  • Yang Z, Hu Y, Zhang J, Xu L, Zeng R, Kang D. Estradiol therapy and breast cancer risk in perimenopausal and postmenopausal women: a systematic review and meta-analysis. Gynecol Endocrinol 2017;33:87–92
  • Canonico M, Alhenc-Gelas M, Plu-Bureau G, Olie V, Scarabin PY. Activated protein C resistance among postmenopausal women using transdermal estrogens: importance of progestogen. Menopause 2010;17:1122–7
  • Harman SM, Black DM, Naftolin F, et al. Arterial imaging outcomes and cardiovascular risk factors in recently menopausal women. A Randomized Trial. Ann Intern Med 2014;161:249–60
  • Rosano GM, Webb CM, Chierchia S, et al. Natural progesterone, but not medroxyprogesterone acetate, enhances the beneficial effect of estrogen on exercise-induced myocardial ischemia in postmenopausal women. J Am Coll Cardiol 2000;36:215–19
  • Writing Group for the PEPI Trial. Effects of estrogen or estrogen/progestin regimens on heart disease risk factors in postmenopausal women. The Postmenopausal Estrogen/Progestin Interventions (PEPI) trial. JAMA 1995;273:199–208
  • Koh KK, Han SH, Shin MS, Ahn JY, Lee Y, Shin EK. Significant differential effects of lower doses of hormone therapy or tibolone on markers of cardiovascular disease in post-menopausal women: a randomized, double-blind, crossover study. Eur Heart J 2005;26:1362–8
  • Moorjani S, Dupont A, Labrie F, et al. Changes in plasma lipoprotein and apolipoprotein composition in relation to oral versus percutaneous administration of estrogen alone or in cyclic association with utrogestan in menopausal women. J Clin Endocrinol Metab 1991;73:373–9
  • Xue W, Deng Y, Wang YF, Sun AJ. Effect of half-dose and standard-dose conjugated equine estrogens combined with natural progesterone or dydrogesterone on components of metabolic syndrome in healthy postmenopausal women: a randomized controlled trial. Chin Med J (Engl) 2016;129:2773–9
  • Casanova G, Spritzer PM. Effects of micronized progesterone added to non-oral estradiol on lipids and cardiovascular risk factors in early postmenopause: a clinical trial. Lipids Health Dis 2012;11:133
  • Casanova G, dos Reis AM, Spritzer PM. Low-dose oral or non-oral hormone therapy: effects on C-reactive protein and atrial natriuretic peptide in menopause. Climacteric 2015;18:86–93
  • Suvanto-Luukkonen E, Sundstrom H, Penttinen J, Kauppila A. Lipid effects of an intrauterine levonorgestrel device or oral vs. vaginal natural progesterone in post-menopausal women treated with percutaneous estradiol. Arch Gynecol Obstet 1998;261:201–8
  • Kling JM, Lahr BA, Bailey KR, Harman SM, Miller VM, Mulvagh SL. Endothelial function in women of the kronos early estrogen prevention study. Climacteric 2015;18:187–97
  • Mather KJ, Norman EG, Prior JC, Elliott TG. Preserved forearm endothelial responses with acute exposure to progesterone: A randomized cross-over trial of 17-beta estradiol, progesterone, and 17-beta estradiol with progesterone in healthy menopausal women. J Clin Endocrinol Metab 2000;85:4644–9
  • Shifren JL, Rifai N, Desindes S, McIlwain M, Doros G, Mazer NA. A comparison of the short-term effects of oral conjugated equine estrogens versus transdermal estradiol on C-reactive protein, other serum markers of inflammation, and other hepatic proteins in naturally menopausal women. J Clin Endocrinol Metab 2008;93:1702–10
  • Bechlioulis A, Naka KK, Kalantaridou SN, et al. Short-term hormone therapy improves sCD40L and endothelial function in early menopausal women: potential role of estrogen receptor polymorphisms. Maturitas 2012;71:389–95
  • Otsuki M, Saito H, Xu X, et al. Progesterone, but not medroxyprogesterone, inhibits vascular cell adhesion molecule-1 expression in human vascular endothelial cells. Arterioscler Thromb Vasc Biol 2001;21:243–8
  • Piercy KT, Donnell RL, Kirkpatrick SS, et al. Effects of estrogen, progesterone, and combination exposure on interleukin-1 beta-induced expression of VCAM-1, ICAM-1, PECAM, and E-selectin by human female iliac artery endothelial cells. J Surg Res 2002;105:215–19
  • Henderson VW, St John JA, Hodis HN, et al. Cognitive effects of estradiol after menopause: a randomized trial of the timing hypothesis. Neurology 2016;87:699–708
  • Gleason CE, Dowling NM, Wharton W, et al. Effects of hormone therapy on cognition and mood in recently postmenopausal women: findings from the randomized, controlled KEEPS-cognitive and affective study. PLoS Med 2015;12:e1001833
  • Sherwin BB, Grigorova M. Differential effects of estrogen and micronized progesterone or medroxyprogesterone acetate on cognition in postmenopausal women. Fertil Steril 2011;96:399–403