18,752
Views
47
CrossRef citations to date
0
Altmetric
Review

The impact of micronized progesterone on breast cancer risk: a systematic review

, &
Pages 111-122 | Received 01 Nov 2017, Accepted 21 Dec 2017, Published online: 31 Jan 2018

Abstract

Postmenopausal women with an intact uterus using estrogen therapy should receive a progestogen for endometrial protection. The debate on bioidentical hormones including micronized progesterone has increased in recent years. Based on a systematic literature review on the impact of menopausal hormone therapy (MHT) containing micronized progesterone on the mammary gland, an international expert panel’s recommendations are as follows: (1) estrogens combined with oral (approved) or vaginal (off-label use) micronized progesterone do not increase breast cancer risk for up to 5 years of treatment duration; (2) there is limited evidence that estrogens combined with oral micronized progesterone applied for more than 5 years are associated with an increased breast cancer risk; and (3) counseling on combined MHT should cover breast cancer risk – regardless of the progestogen chosen. Yet, women should also be counseled on other modifiable and non-modifiable breast cancer risk factors in order to balance the impact of combined MHT on the breast.

Introduction

The steroid hormone progesterone (P) plays a key role in female reproductionCitation1. For therapeutic reasons, micronized progesterone (MP) can be used, for example, for endometrial protection when estrogens are applied in menopausal women with an intact uterusCitation2. To discuss various topics on MP, regular international expert meetings of three gynecological endocrinologists from the German-speaking countries, Austria, Germany and Switzerland, have been held since 2015 aiming to provide scientifically proven statements on MP treatment in peri- and postmenopausal women, based on a systematic literature search and discussion of the results. The impact of estrogens combined with MP on the mammary gland, especially on breast density, biopsies (benign breast tissue) and cancer risk is the second topic of this seriesCitation3.

Material and methods

In May 2016, a systematic literature search was performed by an independent agency (gwd consult) using the databases Medline (Pubmed) and Embase. Only articles in English were included. There was no time restriction applied. For each topic (impact of MP on (1) breast biopsy, (2) breast histology and (3) breast cancer risk), individual searches were performed using multiple combinations of keywords, Mesh-terms and text words related to the respective topic. For the first topic, included keywords were ‘progesterone’, ‘breast’, ‘density’, ‘treatment’, ‘micronized’, ‘mammography’, ‘exogenous’, ‘hormone’, ‘proliferation’, ‘HRT’, ‘bio-identical’, while ‘MPA’, ‘norethisterone’, ‘progestin’, ‘medroxyprogesterone’ and ‘receptor’ were excluded keywords. The search yielded 60 relevant articles. For the second topic, included keywords were ‘progesterone’, ‘histologic’, ‘treatment’, ‘breast’, ‘hormone’, ‘biopsy’, ‘parenchymal’, ‘bio-identical’ and ‘histology’ and excluded keywords were ‘progestin’, ‘medroxyprogesterone’, ‘norethisterone’ and ‘receptor”. The search yielded 30 relevant articles. For the third topic, included keywords were ‘progesterone’, ‘breast’, ‘cancer’, ‘risk’, ‘treatment’, ‘micronized’, ‘bio-identical’ while excluded keywords included ‘receptor’ and ‘progestin”. The search yielded 83 relevant articles. After exclusion of duplicates, the final list of relevant articles comprised 141 out of all relevant 173 articles. After May 2016, five additional articles have been identified and included into the reviewCitation4–8. The final eligibility assessment and evaluation of the studies’ quality were performed by the expert group (PS, JN, LW).

Results

Of 143 hits, 19 studiesCitation4–22 were selected for the systematic review and expert panel’s discussion. The other publications were excluded as they, for example, did not use MP but synthetic progestins although stated otherwise in the title, focused on infertility treatment or were not original articles, respectively. In the following, the term ‘progestogen’ was used as an umbrella term for MP and synthetic progestins.

Breast density

Of 60 hits, only six articles were suitable for this reviewCitation9–14 (). Of those, four were subgroup analyses of the placebo-controlled, randomized-controlled trial (PC-RCT) Postmenopausal Estrogen/Progestin Intervention (PEPI) trialCitation9–12, one was a post-hoc analysis of two PC-RCTsCitation13 and another one a head-to-head RCTCitation14. Sample size ranged from 77Citation14 to 571Citation10 postmenopausal women. Treatment duration ranged from 2 monthsCitation14 to 3 yearsCitation9. Within the PEPI trial, four menopausal hormone therapy (MHT) regimens were compared to placeboCitation9–12. MHT regimens comprised oral conjugated equine estrogens (CEE) at 0.625 mg/day (o-CEE), o-CEE at 0.625 mg/day combined with oral medroxyprogesterone acetate (o-MPA) at 10 mg/day for 12 days per month (o-CEE + o-seqMPA), o-CEE at 0.625 mg/day combined with o-MPA at 2.5 mg/day (o-CEE + o-contMPA), and o-CEE at 0.625 mg/day combined with oral MP (o-MP) at 200 mg/day for 12 days per month (o-CEE + o-contMP). The post-hoc analysis combined two Danish RCTsCitation13 comparing placebo to either an oral MHT or nasal–oral MHT regimen. The oral MHT regimen contained oral 17β-estradiol (o-E2) at 1 mg/day combined with trimegestone at 0.125 mg/day (o-E2 + o-contTrimegestone), whereas the nasal–oral MHT contained either nasal E2 at 150 or 300 µg/day, respectively, combined with o-MP at 200 mg/day for 14 days per month in women with an intact uterus (n-E2 ± o-seqMP). The head-to-head RCTCitation14 used two different MHT regimes containing either o-CEE at 0.625 mg/day sequentially combined with o-MPA at 5 mg/day (o-CEE + o-seqMPA) or transdermal E2 (t-E2) gel at 1.5 mg/day sequentially combined with o-MP at 200 mg/day (t-E2 + o-seqMP).

Table 1. Overview of trials investigating menopausal hormone therapy (MHT) containing micronized progesterone (MP) and breast density.

Mammographic density was assessed either categorically, e.g. by Breast Imaging Reporting and Data System (BI-RADS) gradesCitation9,Citation13,Citation14, or continuously, e.g. by computer-based mammographic percent densityCitation10–13. After 1 year of MHT within the PEPI trial, mammographic density was significantly increased by all estrogen–progestogen regimens but not by o-CEE or placeboCitation9–11. There were no group differences between combined MHT regimensCitation9–11. All mammographic density increases observed comprised only one category and mostly appeared during the first year of MHT useCitation9. Similarly, mammographic density was significantly increased by oral estrogens combined with trimegestoneCitation13 or MPACitation14. In contrast, mammographic density remained unchanged after treatment with oral or nasal estrogens combined with o-MPCitation13,Citation14. Furthermore, the associations between mammographic density and new-onset breast discomfortCitation11, change in serum progestogen levels or progesterone receptor genotypeCitation12 were analyzed. Women with new-onset breast discomfort had a 3.9% increase in mammographic density regardless of MHT typeCitation11. Increases of serum progestogen in the highest quartile were associated with 3.5% higher mammographic density compared to increases in the lowest quartile. However, there was no indication that genetic variations in the progesterone receptor had an impact on mammographic density or modified the impact of serum progestogen levels on mammographic densityCitation12.

Breast biopsy

Of 30 hits, only three studies were prospective randomized intervention trialsCitation14–17, of which one study used o-MPCitation14,Citation15 and two topical (applied directly on the breast) MPCitation16,Citation17, respectively (). The latter two trialsCitation16,Citation17 were both placebo-controlled with three active comparator arms: topical MP 25 mg/day, topical E2 gel 1.5 mg/day, and the combination of both (E2 + MP). Study duration was short and comprised 11–14 days prior to a scheduled surgery for the removal of a breast lump. The cohorts included either 33 premenopausalCitation16 or 40 postmenopausal womenCitation17. The study endpoints were similar, namely serum steroid levels (E2, P), tissue steroid concentration (E2, P), mammary epithelial mitotic index and cell proliferation marker (PCNA) expression. While serum E2 levels were significantly higher in women applying topical E2 compared to those applying MP or placebo, significant group differences for serum P levels were only found in postmenopausalCitation17 but not in premenopausal topical MP usersCitation16. Tissue E2 concentration was significantly higher in women applying topical E2 compared to those applying placeboCitation16,Citation17 or MPCitation17. Tissue P concentration was significantly higher in women applying topical MP compared to placeboCitation16 or did not reveal any group differencesCitation17. Mammary epithelial mitotic index was significantly increased in those women applying topical E2 when compared to those using topical MPCitation16,Citation17, E2 + MP or placeboCitation17. Similarly, PCNA expression was highest in topical E2 usersCitation16,Citation17 but still significantly higher in women applying topical E2 + MP compared to women applying MPCitation16 and placeboCitation17. Both authors came to the conclusion that topical MP for up to 14 days reduced E2-induced mammary epithelial proliferation.

Table 2. Overview of trials investigating menopausal hormone therapy (MHT) containing micronized progesterone (MP) and breast biopsies.

The impact of a 2-month systemic MHT containing MP on the mammary gland in 77 healthy postmenopausal women was investigated by one RCT yielding three publicationsCitation14,Citation15. In this RCT, head-to-head comparisons were performed using two different MHT regimes containing either o-CEE at 0.625 mg/day sequentially combined with o-MPA at 5 mg/day (o-CEE + o-seqMPA) or t-E2 gel at 1.5 mg/day sequentially combined with o-MP at 200 mg/day (t-E2 + o-seqMP). Core needle biopsy of the upper outer quadrant of the left breast was performed at baseline and study end. Study endpoints were breast cell proliferation (Ki-67/MIB-1) and apoptosis (bcl-2) assessed by immunohistochemistryCitation14,Citation15, single gene expression analysis assessed by reverse transcription polymerase chain reaction (rtPCR)Citation14 and whole genome expression analysis by microarrayCitation14. Assessable breast samples at both time points were available for 10%Citation14 to 49%Citation14,Citation15 of subjects. After 2 months of treatment, breast cell proliferation and Ki-67 gene expression were significantly increased by o-CEE + o-MPA but not by t-E2 + o-MPCitation14,Citation15. In contrast, breast cell apoptosis and bcl-2 gene expression were either decreased by t-E2 + o-MP or did not reveal group differencesCitation14,Citation15. Induction of progesterone receptor B expression was slightly but not significantly lower after t-E2 + o-MP than o-CEE + o-MPA treatmentCitation14. Microarray analysis revealed an altered gene expression profile (fold change ≥1.5) for 2500 genes within the o-CEE + o-MPA arm and 300 genes within the t-E2 + o-MP armCitation14. A total of 225 genes were involved in mammary tumor development of which 198 were attributable to o-CEE + o-MPA and 34 to t-E2 + o-MP. The different aspects of the study came to the conclusion that, in comparison to ‘conventional’ MHT, transdermal E2 combined with oral MP induced less proliferation and adverse expression of important genes regulating proliferation, apoptosis and tumor inclination in vivo.

Breast cancer risk

Breast cancer risk in respect to MHT containing MP was assessed by two systematic reviews and meta-analysisCitation4,Citation7, one retrospective cohort studyCitation18, two prospective cohort studies (the Etude Epidémiologique de femmes e la Mutuelle Générale de l’Education Nationale (E3N), and Menopause: Risk of breast cancer, morbidity and prevalence (MISSION))Citation8,Citation19–21,Citation23,Citation24, one case–control study (CECILE, a population-based case-control study in Cote d’Or and Ille-et-Vilaine)Citation25 and two PC-RCT (Kronos Early Estrogen Prevention Study (KEEPS)Citation22, and Early versus Late Intervention Trial with Estradiol (ELITE)Citation5) (). The first meta-analysisCitation7 included two cohort studiesCitation8,Citation24 and reported that breast cancer risk was lower for estrogens combined with MP than with synthetic progestins (relative risk (RR) 0.67; 95% confidence interval (CI) 0.55–0.81). Mean MHT duration was 7.0 yearsCitation24 and 8.3 yearsCitation8, respectively. According to the second meta-analysis covering 14 trials, breast cancer risk was increased when estrogens were combined with MPA, norethisterone (NET) or levonorgestrel (LNG) but not when combined with dydrogesterone (DYD) or MPCitation4, respectively. However, the duration of MHT use was not considered.

Table 3. Overview of trials investigating menopausal hormone therapy (MHT) containing micronized progesterone (MP) and breast cancer risk.

Except for the two US-American PC-RCTs, all other studies were performed in France. The primary endpoints were breast cancer riskCitation8,Citation18–21,Citation23–25 or changes in carotid artery intima-media thicknessCitation5,Citation22. In the latter, breast cancer was assessed as a serious adverse eventCitation5,Citation22. The sample size ranged from 643Citation5 to 80 391Citation23 postmenopausal women, and mean follow-up from 4.0Citation22 to 11.2Citation21 years. At study entry, women were in their fifties in allCitation5,Citation18–20,Citation22–24 but two studiesCitation5,Citation8 that also recruited women during late postmenopause. Only CECILE did not report on participants’ ageCitation25. Information on MHT use was obtained from medical recordsCitation8,Citation18, self-administered questionnaires at baselineCitation26 and then every 2 yearsCitation19–21,Citation23,Citation24, in-person interviewsCitation25, and scheduled visits at 2-monthCitation5 or 12-monthCitation22 intervals. Mean duration of MHT use ranged from 2.8 yearsCitation19 to ≥10 yearsCitation18. Adherence to medication was high in KEEPS (>94%)Citation22 and ELITE (98%)Citation5 but not reported in the other studies included.

Both PC-RCTs, KEEPSCitation22 and ELITECitation5 used a sequentially combined MHT. In KEEPS, o-CEE at 0.45 mg/day or t-E2 patch at 50 µg/day was combined with o-MP 200 mg/day on days 1–12 of each month (o-CEE + o-seqMP, t-E2 + o-seqMP)Citation22. In ELITE, o-E2 at 1 mg/day was combined with vaginal MP at 45 mg/day (4% gel) on 10 days during each 30-day cycle (o-E2 + vag-seqMP)Citation5. The observational cohort and case–control studies differentiated between progestogen types such as MPCitation8,Citation18–21,Citation23–25, DYDCitation18,Citation20,Citation21,Citation23,Citation24 and synthetic progestins (progesterone- and testosterone derivatives)Citation8,Citation18,Citation19,Citation21,Citation23–25 but not between estrogen types, MHT dosages and MHT regimen (sequentially or continuously combined MHT). The definition of current and past MHT use differed between studies. For some, current MHT use corresponded to systemic estrogen therapy for ≥1 yearCitation18–20,Citation24. For others, current systemic MHT also comprised women that had stopped MHT use ≤1 yearCitation25 or even ≤5 yearsCitation8 before the reference date, which in contrast was defined as past use in another studyCitation21. One study grouped current and past MHT use togetherCitation23.

The first study to report on the impact of different progestogen types within combined MHT on breast cancer risk did not find a significant difference between combined MHT use and non-use (adjusted RR 1.10; 95% CI 0.73–1.66)Citation18. This finding was not altered when differentiating between time since last MHT use (<5 years vs. ≥5 years) and duration of MHT use (<5 years vs. ≥ 5 years). Unfortunately, subgroup analysis for progestogen types was not performed. However, the majority of combined MHT contained MP (58%) or DYD (10%) and only <3% MPA. The MISSION trial did not find a significant difference for breast cancer risk when comparing MHT users with non-users (non-adjusted RR 0.91; 95% CI 0.45–1.86)Citation8. Similarly, breast cancer risk in MHT users did not differ between MHT types and duration of use (≤5 years vs. >5 years). In contrast, when compared to MHT non-use, the first E3N report from 2005 found a significant increased breast cancer risk for any combined MHT use (multivariate adjusted RR 1.3; 95% CI 1.1–1.5)Citation19. Breast cancer risk was not altered by duration of MHT use (<2 years vs. 2–4 years vs. ≥4 years; p for trend = 0.7). However, when differentiating for progestogen type, estrogens combined with oral MP were not associated with an increased breast cancer risk (multivariate adjusted RR 0.9; 95% CI 0.7–1.2), while estrogens combined with synthetic progestins were (multivariate adjusted RR 1.4; 95% CI 1.2–1.7). Duration of MHT use (<2 years vs. 2–4 years vs. ≥4 years) only had a slight impact when oral estrogens were combined with synthetic progestins (p for trend = 0.07) but not when combined with MP (p for trend = 0.9). Similarly, the second E3N report published in 2008 did not find an increased breast cancer risk for combined MHT containing either MP or DYD regardless of MHT duration (<2 years vs. 2– <4 years vs. ≥4–<6 years vs. ≥6 years), although a significant time trend was observed in women using estrogens combined with MP (p for trend = 0.04)Citation24. In addition, combined MHT containing MP was not found to be associated with any breast cancer subtypeCitation23. These findings were supported by CECILECitation25. However, the numbers of cases and controls were very small in subgroups and the authors did not differentiate between invasive and in situ breast cancer. In contrast, the third E3N report from 2014 found a significant increased breast cancer risk for mean 6.1 years of use of combined MHT containing MP or DYD (multivariate adjusted RR 1.22; 95% CI 1.11–1.35). When differentiating between short-term (≤5 years) and long-term use (>5 years), a significant increased breast cancer risk was only found for long-term use (multivariate adjusted RR 1.31; 95% CI 1.15–1.48). In comparison, use of combined MHT containing synthetic progestogens for more than 5 years was associated with an increased breast cancer risk (multivariate adjusted RR 1.98; 95% CI 1.73–2.26). Importantly, after stopping MHT containing MP or DYD after >5 years of use, breast cancer risk dissolved immediately (3 months to 5 years since last use: multivariate adjusted RR 1.15; 95% CI 0.93–1.42). In contrast, breast cancer risk was still elevated even 5–10 years after stopping MHT containing synthetic progestins when use was at least 5 years (multivariate adjusted RR 1.34; 95% CI 1.04–1.73). The time gap between menopause and MHT initiation did not have an impact on breast cancer risk in women using estrogens combined with MPCitation20,Citation21. In the two PC-RCTs, breast cancer was newly diagnosed in eight women in KEEPSCitation22 (n = 3 o-CEE, n = 3 t-E2, n = 2 placebo), and in 18 women in ELITECitation5 (n = 10 o-E2, n = 8 placebo), respectively. The difference between MHT and placebo groups was not significant in both studies.

Discussion

Current international guidelines on MHT recommend to combine a progestogen when using estrogen therapy in peri- and postmenopausal women with an intact uterus for endometrial protectionCitation2,Citation27–29. However, long-term combined estrogen–progestogen therapy has been shown to be associated with an increased breast cancer risk. During the last years, the debate about (compounded) bioidentical hormones has increased tremendouslyCitation30–32. Specifically, the question has been raised whether bioidentical hormone therapy including MP has a different or even beneficial impact on the mammary gland. Internationally, systemic MP is available at different dosages and routes of application. Also, indication and approval by regulatory authorities may differ from country to country. In Europe, systemic MP is available as a capsule (100 mg, 200 mg) for vaginal or oral application or as a vaginal gel (8% corresponding to 90 mg).

Our systematic review on the impact of estrogens combined with MP on the postmenopausal mammary gland showed that (1) mammographic density may either increase or remain unchanged, (2) proliferation induction was less pronounced compared to ‘conventional’ MHT, and (3) breast cancer risk was not affected for up to 5 years of treatment. However, (4) estrogens combined with MP or dydrogesterone were associated with a slight but significant increase in breast cancer risk after an average of 6 years of treatment duration.

Breast density is a mammographic finding based on differing proportions of fat, connective and epithelial tissue. Mammographic density can be assessed either by the BI-RADS classification (almost entirely fatty, scattered areas of fibroglandular density, heterogeneously dense, extremely dense)Citation33 or by more objective, but not widely implemented computer-based breast density assessmentsCitation34–36. Mammographically dense breast tissue both decreases the sensitivity of mammograms and increases breast cancer riskCitation37 but not breast cancer mortalityCitation38. There are multiple factors contributing to mammographic density such as age, genetics, body habitus, parity, and MHT use. Our systematic review revealed contradicting results for MHT containing MP, with two studies showing no changeCitation13,Citation14 and three substudies showing a significant increase in mammographic densityCitation9–11. The latter are in line with another longitudinal study showing that the age-related change from dense to fatty breast tissue was slowed down more in women taking combined MHT than in those taking estrogen aloneCitation39. The differing results may also be due to the method itself and differences between the US-AmericanCitation4–6 and EuropeanCitation13,Citation14 cohorts. US-American women were in their late fifties, overweight and had mostly used MHT beforeCitation4–6, while European women were younger at least in one substudyCitation13 and had a normal body mass indexCitation8,Citation9. Baseline mammographic density was reported by all but one studyCitation14. Most women (approximately 60%) fell into the BI-RADS categories 1 and 2Citation9–11,Citation13. However, mammographic density interpretation is subjective to some degree as moderate interobserver and intraobserver variabilities, especially between the BI-RADS categories of heterogeneously dense and scattered areas of fibroglandular density, have been reportedCitation40–42. Accordingly, a striking but non-significant interobserver variability was reported by one studyCitation9.

Due to heterogeneous study designs, reports on the impact of estrogens combined with MP on breast tissue were not comparable. Outcome markers differed, ranging from tissue sex steroid concentrations, immunohistochemistry, rtPCR to microarray gene expression analysis. Study duration was short and 2 months at maximum. Furthermore, two studies used topical MPCitation16,Citation17 which is not thought to have a systemic impactCitation43. The observed differences in tissue E2 and P concentrations may be due to pharmacological interference and different reproductive stages. Thus, there is only some weak evidence from breast biopsies in healthy women showing that estrogens combined with oral MP are more ‘breast friendly’ than estrogens combined with oral MPA, a finding supported by studies in non-human primatesCitation44,Citation45.

In respect to breast cancer risk, all studies confirmed that estrogens combined with MP did not increase breast cancer risk when treatment duration was 5 years or less. The only two studies assessing breast cancer risk in women using MHT containing MP for more than 5 years are the prospective cohort studies E3N and MISSION. Yet, compliance, dosage and route of application of MP were not exactly known. In addition, the E3N report from 2014 did not differentiate between MP and dydrogesterone. Another limitation of E3N was the high rates of MHT changes over time: of those who ever used estrogens combined with MP or dydrogesterone, 57% also used estrogens combined with synthetic progestogensCitation21. The majority of studies used oral MP, which is the approved way of application for MHT. Thus, breast safety data on vaginal MP is scarceCitation5 or completely lacking for transdermal MP. Despite the limited evidence, women should be counseled that, if using combined MHT for more than 5 years, the risk of being diagnosed with breast cancer increases – regardless of the progestogen type chosen. However, in order to balance the impact of non-modifiable (e.g. genetics, breast density, parity) and modifiable breast cancer risk factors (e.g. alcohol, smoking, overweight/obesity, physical inactivity, MHT), women should also be counseled that the possible increased breast cancer risk with combined MHT is small (<1 per 1000 women per year of use) and lower than the increased risks associated with common lifestyle factors such as reduced physical activity, obesity and alcohol consumptionCitation46.

Conclusion

Postmenopausal women with an intact uterus using estrogen therapy should receive a progestogen for endometrial protection. Based on a systematic literature review on the impact of micronized progesterone on the mammary gland, an international expert panel’s recommendations on MHT containing micronized progesterone are as follows: (1) estrogens combined with oral (approved) or vaginal (off-label use) micronized progesterone do not increase breast cancer risk for up to 5 years of treatment duration; (2) there is limited evidence that estrogens combined with oral micronized progesterone applied for more than 5 years are associated with an increased breast cancer risk; and (3) counseling on combined MHT should cover breast cancer risk – regardless of the progestogen chosen. Yet, women should also be counseled on other modifiable and non-modifiable breast cancer risk factors in order to balance the impact of combined MHT on the breast.

Conflict of interest

The authors have been part of a German-speaking expert board funded by DR. KADE/BESINS Pharma GmbH. The authors alone are responsible for the content and writing of the paper.

Acknowledgements

The authors are grateful for the support of DR. KADE/BESINS Pharma GmbH for helping with ordering the identified publications. Petra Stute, Ludwig Wildt and Joseph Neulen were involved in the scientific discussion. Petra Stute prepared the manuscript.

Additional information

Funding

This publication was developed by an expert board from Austria, Germany and Switzerland. The board meeting and the independent agency (gwd consult) for the literature search were funded by DR. KADE/BESINS Pharma GmbH without influence on the content.

References

  • Csapo AI, Pulkkinen M. Indispensability of the human corpus luteum in the maintenance of early pregnancy. Luteectomy evidence. Obstet Gynecol Surv 1978;33:69–81
  • de Villiers TJ, Pines A, Panay N, et al. Updated 2013 International Menopause Society recommendations on menopausal hormone therapy and preventive strategies for midlife health. Climacteric 2013;16:316–37
  • Stute P, Neulen J, Wildt L. The impact of micronized progesterone on the endometrium: a systematic review. Climacteric 2016;19:316–28
  • 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
  • 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
  • So¨Derqvist G, Murkes D, Lalitkumar P. Percutaneous estradiol/oral micronized progesterone gives considerably less mammary tumor increasing gene expressions than oral conjugated equine estrogens/medroxyprogesterone acetate in the breasts of healthy women in vivo: results from micro-array and PCR data from core needle biopsies. Climacteric 2016;19:34
  • Asi N, Mohammed K, Haydour Q, et al. Progesterone vs. synthetic progestins and the risk of breast cancer: a systematic review and meta-analysis. Syst Rev 2016;5:121
  • Espie M, Daures JP, Chevallier T, Mares P, Micheletti MC, De Reilhac P. Breast cancer incidence and hormone replacement therapy: results from the MISSION study, prospective phase. Gynecol Endocrinol 2007;23:391–7
  • Greendale GA, Reboussin BA, Sie A, et al. Effects of estrogen and estrogen-progestin on mammographic parenchymal density. Postmenopausal Estrogen/Progestin Interventions (PEPI) Investigators. Ann Intern Med 1999;130:262–9
  • Greendale GA, Reboussin BA, Slone S, Wasilauskas C, Pike MC, Ursin G. Postmenopausal hormone therapy and change in mammographic density. J Natl Cancer Inst 2003;95:30–7
  • 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
  • Lee E, Ingles SA, Van Den Berg D, et al. Progestogen levels, progesterone receptor gene polymorphisms, and mammographic density changes: results from the Postmenopausal Estrogen/Progestin Interventions Mammographic Density Study. Menopause 2012;19:302–10
  • Pettersen PC, Raundahl J, Loog M, Nielsen M, Tanko LB, Christiansen C. Parallel assessment of the impact of different hormone replacement therapies on breast density by radiologist- and computer-based analyses of mammograms. Climacteric 2008;11:135–43
  • 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;28:12–15
  • 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
  • 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
  • de Lignieres B, de Vathaire F, Fournier S, et al. Combined hormone replacement therapy and risk of breast cancer in a French cohort study of 3175 women. Climacteric 2002;5:332–40
  • 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, Mesrine S, Boutron-Ruault MC, 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
  • 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
  • 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
  • Fournier A, Fabre A, Mesrine S, Boutron-Ruault MC, Berrino F, Clavel-Chapelon F. Use of different postmenopausal hormone therapies and risk of histology- and hormone receptor-defined invasive breast cancer. J Clin Oncol 2008;26:1260–8
  • 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
  • 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
  • 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
  • The 2017 hormone therapy position statement of The North American Menopause Society. Menopause 2017;24:728–53
  • Armeni E, Lambrinoudaki I, Ceausu I, et al. Maintaining postreproductive health: a care pathway from the European Menopause and Andropause Society (EMAS). Maturitas 2016;89:63–72
  • Sarri G, Davies M, Lumsden MA. Guideline Development Group. Diagnosis and management of menopause: summary of NICE guidance. BMJ 2015;351:h5746
  • Pinkerton JV, Santoro N. Compounded bioidentical hormone therapy: identifying. Use trends and knowledge gaps among US women. Menopause 2015;22:926–36
  • Pinkerton JV. Think twice before prescribing custom-compounded bioidentical hormone therapy. J Womens Health (Larchmt) 2014;23:631–3
  • Pinkerton JV. What are the concerns about custom-compounded ‘bioidentical’ hormone therapy? Menopause 2014;21:1298–300
  • Sickles E, D'Orsi CJ, Bassett LW, et al. ACR BI-RADS mammography. In D’Orsi CJ, Sickles EA, Mendelson E, et al., eds. ACR BI-RADS Atlas, Breast Imaging Reporting and Data System. Reston, VA: American College of Radiology; 2013
  • Byng JW, Yaffe MJ, Jong RA, et al. Analysis of mammographic density and breast cancer risk from digitized mammograms. Radiographics 1998;18:1587–98
  • Byng JW, Boyd NF, Fishell E, Jong RA, Yaffe MJ. The quantitative analysis of mammographic densities. Phys Med Biol 1994;39:1629–38
  • Yaffe MJ. Mammographic density. Measurement of mammographic density. Breast Cancer Res 2008;10:209
  • Vachon CM, van Gils CH, Sellers TA, et al. Mammographic density, breast cancer risk and risk prediction. Breast Cancer Res 2007;9:217
  • Gierach GL, Ichikawa L, Kerlikowske K, et al. Relationship between mammographic density and breast cancer death in the Breast Cancer Surveillance Consortium. J Natl Cancer Inst 2012;104:1218–27
  • van Duijnhoven FJ, Peeters PH, Warren RM, et al. Postmenopausal hormone therapy and changes in mammographic density. J Clin Oncol 2007;25:1323–28
  • Nicholson BT, LoRusso AP, Smolkin M, Bovbjerg VE, Petroni GR, Harvey JA. Accuracy of assigned BI-RADS breast density category definitions. Acad Radiol 2006;13:1143–9
  • Kerlikowske K, Grady D, Barclay J, et al. Variability and accuracy in mammographic interpretation using the American College of Radiology Breast Imaging Reporting and Data System. J Natl Cancer Inst 1998;90:1801–9
  • Ciatto S, Houssami N, Apruzzese A, et al. Categorizing breast mammographic density: intra- and interobserver reproducibility of BI-RADS density categories. Breast 2005;14:269–75
  • Ruan X, Mueck AO. Systemic progesterone therapy–Oral, vaginal, injections and even transdermal? Maturitas 2014;79:248–55
  • Wood CE, Register TC, Lees CJ, Chen H, Kimrey S, Cline JM. Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys. Breast Cancer Res Treat 2007;101:125–34
  • Wood CE, Register TC, Cline JM. Transcriptional profiles of progestogen effects in the postmenopausal breast. Breast Cancer Res Treat 2009;114:233–42
  • Baber RJ, Panay N, Fenton A. IMS Writing Group. 2016 IMS Recommendations on women’s midlife health and menopause hormone therapy. Climacteric 2016;19:109–50