Effects of menopausal hormone therapy-based on the role of estrogens, progestogens, and their metabolites in proliferation of breast cancer cells ================================================================================================================================================= * Yu Deng * Hongyan Jin ## Abstract Menopausal hormone therapy (MHT) has been widely used for the clinical treatment of symptoms associated with menopause in women. However, the exact nature of the relationship between MHT and the increased risk of breast cancer has not been fully elucidated. The results of the Women’s Health Initiative’s randomized controlled clinical studies showed that estrogen monotherapy was associated with a lower incidence of breast cancer as compared to estrogen-progesterone combined therapy, with an elevated risk of breast cancer. The evidence currently available from randomized trials and observational studies is based on data from different populations, drug formulations, and routes of administration. Even though the risks of MHT and breast cancer have received a great deal of attention, information regarding the unpredictable toxicological risks of estrogen and progestogen metabolism needs to be further analyzed. Furthermore, the diversity and complexity of the metabolic pathways of estrogen and different progestogens as well as the association of the different estrogen and progestogen metabolites with the increased risk of breast cancer need to be adequately studied. Therefore, this review aimed to describe the biological effects of estrogen, progesterone, and their metabolites on the proliferation of breast cancer cells, based on relevant basic research and clinical trials, to improve our understanding of the biological functions of estrogen and progestogen as well as the safety of MHT. * Menopausal hormone therapy * estrogen * progestogens * breast cancer ## Introduction The increasing number of menopausal women in our population has made it necessary to carefully monitor menopausal women’s health care. Menopausal hormone therapy (MHT) is a generic term recommended by The Endocrine Society. It refers to any type of hormone therapy that is administered to menopausal women. This therapy is usually prescribed to alleviate certain menopause-associated symptoms, such as hot flashes, sweating, anxiety, and depression1–4. The most frequently used MHTs presently include the estrogen plus progestogen treatment (EPT) and estrogen treatment (ET) based on uterine integrity. Estrogens are classified into natural and synthetic estrogens (**Table 1**). Natural estrogens mainly include estradiol valerate, 17β-estradiol (E2), and conjugated equine estrogen (CEE), whereas synthetic estrogens include nylestriol and ethinylestradiol. Similarly, progestogens comprise natural progestogens and synthetic progestins (**Table 1**). Natural progestogen is represented by progesterone (P4), whereas synthetic progestins mainly include dydrogesterone, medroxyprogesterone acetate (MPA), norethindrone (NET), and drospirenone6,7. In addition, estrogen and P4 play critical roles in the development of the mammary gland during puberty and pregnancy8. The Women’s Health Initiative (WHI) trial and the Million Women Study (MWS)9,10 reported that the use of EPT increased the risk of breast cancer in women; in contrast, results of a French E3N cohort study11,12 suggested that estrogen plus P4 did not increase this risk. Therefore, the role of estrogens combined with different progestogens in the development of breast cancer in menopausal women is not determined. View this table: [Table 1](http://www.cancerbiomed.org/content/19/4/432/T1) Table 1 Classification of estrogens and progestogens Santen et al.13,14 used growth kinetic models to show that occult tumors required approximately 16 years and 30 rounds of doubling times before they could be detected by clinical testing; EPT mainly stimulated the growth of pre-existing occult breast cancer and decreased the doubling time from an average of 200 days to 150 days. Hence, estrogen and progesterone may promote the progression of pre-existing occult breast cancer under certain conditions. Estrogen and progesterone metabolisms are highly diverse and unstable, thereby resulting in unpredictable toxicological risks in women. Previous studies15–18 have reported that estrogen metabolites (EMs) and progesterone metabolites may be associated with the risk of breast cancer; however, this aspect has not been adequately investigated. It is therefore essential to review the metabolic pathways of estrogens and different progestogens, to obtain an increased awareness about the role of estrogen and progestogen metabolites in breast cancer. Because the roles of estrogens and progestogens are highly diverse, yet not clearly known with respect to the progression of breast cancer, our aim was to summarize the epidemiological studies of MHT and breast cancer, the signaling pathways related to E2 and different progestogens, and the pharmacological differences among the various progestogens. This will help to understand the safety of administering MHT as well as its association with the increased risk of developing breast cancer. ## Association of MHT with the risk of breast cancer The WHI study, which began in 1993, was a long-term, multicenter, randomized, double-blind, and controlled clinical trial that enrolled 27,347 postmenopausal women aged 50–79 years at 40 clinical research centers in the USA. The WHI trial was divided into 2 categories; the WHI EPT Trial and the WHI ET Trial, based on the presence or absence of a uterus, respectively. In the WHI EPT Trial9, 16,608 postmenopausal women who had not undergone hysterectomy were randomly selected to receive a daily dose of placebo or a combination of 0.625 mg CEE/2.5 mg MPA. In the WHI ET Trial19, 10,739 postmenopausal women who had undergone hysterectomy were randomly selected to receive a daily dose of placebo or 0.625 mg CEE. Compared with women who received the placebo, women who received EPT (CEE + MPA) had a significantly increased risk of breast cancer [hazard ratio (HR): 1.26; 95% confidence interval (CI): 1.00–1.59]9. In contrast, women who were administered ET (CEE alone) did not exhibit an increased risk of breast cancer (HR: 0.77; 95% CI: 0.59–1.01)19 when compared with women who received the placebo. The WHI study raised concerns about the association of MHT with the risk of breast cancer. The most used drugs in MHT are CEE, E2, P4, and MPA; however, the most troubling question is their relationships with increased risks of breast cancer. From the data collected in the clinical trials (**Table 2**), it could be inferred that ET, which was only indicated for women who had undergone hysterectomies, had a lower risk of stimulating breast cancer development than EPT (E2 + P4 or E2 + MPA). Moreover, administration of P4 and dydrogesterone appeared to result in a lower risk of inducing breast cancer development than the use of synthetic progestogen, such as MPA28. However, a better understanding of the risks of MHT requires a combined knowledge of the molecular mechanisms as well as the biological functions of estrogens and progestogens. View this table: [Table 2](http://www.cancerbiomed.org/content/19/4/432/T2) Table 2 Results of the clinical studies of menopausal hormone therapy and the risks of breast cancer ## Effect of E2 on breast cancer proliferation ### E2 signaling pathways in breast cancer cells The major endogenous estrogens are E2, estrone (E1), and estriol (E3), among which E2 is the predominant estrogen used by women prior to menopause29. Moreover, E1 and E2 are interchangeable through the action of 17β-hydroxysteroid dehydrogenases 1 and 2 (17β-HSD1 and 2)30. Estrogen plays a wide range of biological roles in mammary glands, uterine tissues, cardiovascular, musculoskeletal, immune, and central nervous systems through the estrogen receptor (ER)31. The ERs are categorized into 2 types, namely ERα and Erβ; ERα is the main receptor for estrogen action in the mammary glands32. Estrogens have been shown to stimulate the proliferation of breast cancer cells through genomic and non-genomic pathways. In the genomic pathway, estrogens and their receptors bind directly to the estrogen response elements (EREs) present in the nucleus of breast cancer cells and recruit cofactors to form transcription initiation complexes; these complexes then activate the transcriptions and expressions of proliferation-related target genes, where time is specified in hours or is even delayed for days33–35. Non-genomic effects have a faster onset (from seconds to a few minutes) and might be related to interactions with structures in the plasma membrane, with the effects frequently associated with the activation of various protein-kinase cascades36. In these pathways33–35, E2 can induce rapid cellular effects by binding to the estrogen membrane receptor (mER) or G protein-coupled estrogen receptor (GPER) localized at the plasma membrane, which is required for rapid downstream signaling in the phosphoinositide 3-kinase (PI3K)/Akt kinase pathway as well as the mitogen-activated protein kinase (MAPK) pathway37–39. The main non-genomic signaling pathways of estrogens through mER are as follows (**Figure 1**). With respect to the MAPK/ERK pathway, Raffo et al.40 reported that the MAPK/ERK pathway was activated within 10 min of administering 10 nM E2 in 2 breast cancer cell lines, namely MCF-7 and LM05-E cells (ER+, PR+). Second, with respect to the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway, Aronica et al.41 reported that very low estradiol concentrations (a half-maximal dose of 10 pM) caused an increase in intracellular cAMP by increasing membrane adenylate cyclase activity, thereby activating genes that contained the cAMP response element. Third, with respect to the PI3K/AkT pathway, Garrido et al.42 reported that within 5, 15, and 25 min of treating MCF-7 cells with 10 nM E2, the PI3K/AkT signaling pathway was activated, thereby stimulating glucose uptake by cells. ![Figure 1](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/19/4/432/F1.medium.gif) [Figure 1](http://www.cancerbiomed.org/content/19/4/432/F1) Figure 1 Signaling pathways of estrogen-mediated breast cancer cell proliferation. Estrogen treatment leads to breast cancer growth through genomic and non-genomic pathways. In the genomic pathway, estrogen and its receptors bind directly to specific DNA sequences called estrogen response elements (EREs) and activate gene expressions; additionally, in non-genomic pathways, estrogen signals can occur by plasma membrane localization of ER, or by GPER, to induce rapid cellular effects by activating several kinase cascades, such as cAMP-PKA, PI3K/AKT, and MEK/ERK 1/2. ER, estrogen receptor; GPER, G protein-coupled estrogen receptor; TF, transcription factor. Adapted from Wilkenfeld et al.33. The GPER (also known as GPR30) is a seven-transmembrane G-protein-coupled receptor (GPCR) embedded into cellular membranes43. Recently published research44 highlighted that E2-induced GPER expression promoted proliferation, invasion, and migration of MCF-7 breast cancer cells through the miR-124/CD151 pathway. Filardo et al.45 found that stimulation of SkBr3 cells that expressed neither ERα nor ERβ, in the presence of 1 nM 17β-estradiol for 5 min, induced a 6-fold increase of Erk phosphorylation. Moreover, Vivacqua et al.46 reported that E2 transactivated the early growth response-1 (Egr-1) promoter sequence and induced Egr-1 expression through the GPER/ERK pathway in SkBr3 breast cancer cells. Additionally, E2 might play an important role in the *in situ* transition of ductal carcinoma in the breast by the GPER signaling pathway47. Deng et al.47 reported that E2 induced basement membrane disruption in breast glandular ducts by promoting matrix metalloproteinase 3 and interleukin-1β secretion through the GPER/cAMP/PKA and GPER/PI3K/AkT pathways. ### Effects of EMs on breast cancer cells In addition to ER-induced proliferative effects of the breast cancer cells, E2 is associated with the increased risk of breast cancer due to the DNA toxicity of its metabolites48. The metabolism of E2 can be divided into phase I and phase II reactions. In phase I reactions, E2 is metabolized to hydroxyl compounds by the cytochrome P450 enzyme (cytochrome P450, CYP) and in the phase II reaction, the hydroxyl compounds are converted to nontoxic, water-soluble compounds by catechol-O-methyltransferase (COMT). In detail, there are 3 main metabolic pathways for E2 in the liver, namely the 2-hydroxylation pathway, the 4-hydroxylation pathway, and the 16-hydroxylation pathway, of which the 2-hydroxylation pathway is the main metabolic pathway for E249. The metabolic pathways of E2 discussed below are shown in **Figure 2**. ![Figure 2](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/19/4/432/F2.medium.gif) [Figure 2](http://www.cancerbiomed.org/content/19/4/432/F2) Figure 2 Metabolic pathways of E2. There are 3 pathways of E2 metabolism: the 2-hydroxylation pathway, the 4-hydroxylation pathway, and the 16-hydroxylation pathway, of which the 2-hydroxylation pathway is the main metabolic pathway for E2. 2-OH E2, 2-hydroxyestradiol; COMT, catechol-O-methyltransferase; 4-OH E2, 4-hydroxyestradiol; 16α-OH E1, 16α-hydroxyestrone; E2-2,3-Q, E2-2,3-quinone; E2-3,4-Q, E2-3,4-quinone; ROS, reactive oxygen species. In the 2-hydroxylation pathway, E2 is metabolized to 2-hydroxyestradiol (2-OH E2) by cytochrome P450 enzymes (CYP1A1/2) in the phase I reaction. A previous study reported that 2-hydroxyestrone (2-OH E1) and 2-OH E2 significantly inhibited the hormone-induced proliferation of the human breast cancer cell lines, MCF-7 and T47D50. Subsequent *in vivo* tumorigenesis studies showed that tumor growth increases of nude mice treated with 1.5 mg E2 was 335.4%, and that there was no significant increase in the tumorigenesis of the 2-OH E1-treated group, when compared with that of the control group51. Moreover, 2-OH E2 is metabolized in the nucleus to E2-2,3-quinone (E2-2,3-Q) by peroxidase, with concomitant production of reactive oxygen species (ROS). The E2-2,3-Q readily binds to DNA to form DNA adducts such as 2-OH E2-6-N3 adenine that can cause DNA damage. In the phase II reaction, 2-OH E2 is further metabolized by COMT to 2-methoxyestradiol, which prevents ROS production and DNA adducts, thereby protecting the cells from the genotoxicity and cytotoxicity of catechol estrogens52. In the 4-hydroxylation pathway, E2 is transformed into 4-hydroxyestradiol (4-OH E2) by the cytochrome P450 enzyme CYP1B1 in the phase I reaction. Lareef et al.53 reported that 4-OH E2 increased cell proliferation and induced transformation in MCF-10F cells. Moreover, 4-OH-E2 was reported to induce malignant transformation of breast cells as well as tumor formation in nude mice18. The 4-OH E2 might be oxidized to E2-3,4-quinone (E2-3,4-Q) in the nucleus, along with the production of ROS, which increases DNA instability54. The E2-3,4-Q easily forms adducts with nuclear DNA, such as 4-OH E2-1-N3 adenine and 4-OH E2-1-N7 guanine, both of which have carcinogenic potential and might lead to DNA damage55. In the phase II reaction, 4-OH E2 can be further converted to 4-methoxyestradiol (4-methoxy E2) by COMT, thereby preventing the generation of large amounts of DNA adducts. Zahid et al.56 reported that MCF-10F cells oxidized 4-OH E2 to E2-3,4-Q. Additionally, the levels of 4-OH E2-1-N3 adenine and 4-OH E2-1-N7 guanine exhibited a 3- to 4-fold increase when methoxylation of 4-OH E2 was blocked with the COMT inhibitor, Ro41-0960. In the 16-hydroxylation pathway, E1 and E2 are exchanged through the action of 17β-HSD1 and 17β-HSD2, followed by the transformation of E1 into 2-hydroxyestradiol 16α-hydroxyestrone (16α-OH E1) by the cytochrome P450 enzyme (CYP3A4)57,58. The 16α-OH E1 has a stronger estrogenic activity than estradiol, and several studies have reported that 16α-OH E1 significantly increased the expressions of cyclin D1 and cyclin-dependent kinase 2, and promoted cell proliferation in MCF-7 cells51,59,60. There is currently a growing awareness regarding the impact of EMs on menopausal women. There is clinical evidence15 suggesting that certain EMs might be risk factors for breast cancer. The Breast and Bone Follow-up to the Fracture Intervention Trial (B˜FIT) cohort15 (*n* = 13,784) assessed the relationship between EMs and the risk of breast cancer in postmenopausal women during 12 follow-up years. They found an increased occurrence of the 2-hydroxylation metabolic pathway (HRQ5vsQ1 = 0.69; 95% CI: 0.46–1.05; *P* = 0.01) and also found that a higher ratio of 2/16-hydroxylation pathways (HRQ5vsQ1 = 0.60; 95% CI: 0.40–0.90; *P* = 0.002) were associated with a lower risk of breast cancer. Moreover, previous studies16,61 have provided in-depth analyses of the effects of MHT on EMs in postmenopausal women. For example, in a prospective case control study in the Women’s Health Initiative Hormone Trials, the concentration of 2-OH E1 increased 4-fold in the blood, and the ratio of 2-OHE1/16α-OHE1 improved approximately from 0.3 to 1.0, after 1 year of treatment with ET and EPT (*n* = 1,259)16. Additionally, Falk et al.61 reported that there might be some differences between the estrogen metabolic pathways of women receiving ET and EPT in the WHI Observational Study (*n* = 1,864). In comparison with the effects of EPT, ET was more likely to induce the 2-hydroxylation metabolism pathway than the 16-hydroxylation metabolism pathway. In the future, larger studies are necessary to better determine the relationships between the risks of breast cancer and EM levels with respect to the use of ET and EPT. In summary, estrogen as well as various EMs regulate genes that are involved in breast cancer cell proliferation. Although the effects of EMs in postmenopausal women have been reported, the underlying mechanisms need further study. ## Effect of progestogens on breast cancer proliferation Progestogens exert their effects on their target tissues primarily through genomic as well as non-genomic pathways (**Figure 3**). In the genomic pathway, the progesterone receptor (PR) directly binds to progesterone response elements (PREs) or other DNA-binding transcription factors to modify target gene expressions. In contrast, in non-genomic pathways, progestogens activate secondary messenger cascades through specific receptors, such as PRs, progesterone membrane receptors (mPRs), and progesterone receptor membrane component 1 (PGRMC1), to indirectly regulate gene transcription62. ![Figure 3](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/19/4/432/F3.medium.gif) [Figure 3](http://www.cancerbiomed.org/content/19/4/432/F3) Figure 3 Signaling pathways of progestogens leading to breast cancer growth by genomic and non-genomic pathways. In the genomic pathway, progestogens and progesterone receptors (PRs) bind directly to progesterone response elements (PREs) or other DNA-binding transcription factors to promote gene expressions. Rapid progestogenic effects can be induced by non-genomic pathways, in which progestogens (progesterone, medroxyprogesterone acetate, and norethindrone) through some receptors, such as PRs (A), progesterone membrane receptors (B), and progesterone receptor membrane component 1 (C), activate secondary messenger cascades and regulate gene transcription. The PRs belong to the steroid hormone receptor family, and the functional region of the PR consists of the DNA binding domain, the ligand-binding domain, and the transcription-activating functions63. There are 2 types of PRs, namely PR-A (94 kDa) and PR-B (120 kDa). These 2 types of receptors have different physiological functions; for example, PR-B is a more potent transcription factor than PR-A, but PR-A has an inhibitory effect on the transcriptional activity of PR-B, as well as other receptors, such as estrogen, androgen, and glucocorticoid receptors64,65. Under normal physiological conditions, PR-A and PR-B are similarly expressed in breast cells, but breast cancer cells overexpress PR-A more frequently than PR-B66. Some studies67,68 have suggested that imbalances in the expressions of PR-A and PR-B might play an important role in the early development of breast cancer. Breast cancer patients overexpressing PR-A have worse prognoses and lower chances of disease-free survival than breast cancer patients with PR-B overexpression. Mote et al.69 analyzed PR expression in tissue samples of 39 breast cancer patients and found that PR-A > PR-B accounted for 39% (15 out of 39 patients) cases, PR-A = PR-B accounted for 51% (20 out of 39 patients) cases, and PR-A < PR-B accounted for only 10% (4 out of 39 patients) cases. Moreover, Rojas et al.68 reported that among 222 PR+ breast cancer tissue samples, 52.3% (116 cases) had PRA-H (PR-A/PR-B) ≥ 1.2. The mPRs and PGRMC1 lack classical PRs, and their biological functions are not fully understood. The mPRs are novel 7-transmembrane receptors localized on the cell surface, and belong to the progestin and adipoQ receptor (PAQR) family. There are 5 types of mPRS, namely mPRα (PAQR7), mPRβ (PAQR8), mPRγ (PAQR5), mPRδ (PAQR6), and mPRɛ(PAQR9)70. The mPRs have similar GPCR functions, and exhibit a rapid response to non-genomic signaling71. The membrane-associated progesterone receptor (MAPR) family includes PGRMC1, PGRMC2, neudesin, and neuferricin, all of which contain a cytochrome b5-like heme/steroid binding domain72. Zhang et al.73 reported a 67.89% positive expression of PGRMC1 in breast cancer patients (74 out of 109 cases), and the expression of PGRMC1 was related to breast tumor size, lymph node metastasis, and prognoses. Patients with high PGRMC1 expression have lower disease-free survival as well as a lower overall survival, when compared with that of patients with low PGRMC1 expressions74. Even though there is limited information about PGRMC2, due to its 80% similarity to the cyt-b5 domain of PGRMC1, both of them might have overlapping functions and roles72. Causey et al.75 reported that PGRMC1 mRNA levels were significantly lower in stage II breast cancer patients than in stage III breast cancer patients, so measurement of PGRMC2 mRNA might be useful for the staging of breast adenocarcinoma. As previously mentioned, clinical use of progestogens can be categorized into natural and synthetic progestins, both of which are derived from the pregnane skeleton (C21 backbone), with similarities as well as differences in their pharmacological properties76. The WHI EPT Trial9 reported that administration of CEE + MPA increased the risk of breast cancer in women, whereas a large observational study of E3N from France12 reported that administration of E2 + P4 or E2 + dydrogesterone did not increase the risk of breast cancer in women; these differences might be related to the pharmacological properties of different progestins. ### Natural progestogens #### The P4 signaling pathways in breast cancer cells P4 can stimulate the receptor activator of the NF-κB ligand (RANKL)77–79, receptor activator of NF-κB (RANK), PI3K/Akt79–81, and MAPK/ERK82,83 pathways by binding to the PR, which promotes the proliferation of breast cells. For example, with respect to the RANKL/RANK pathway, P4 promotes the proliferation of mammary stem cells84 and breast cells with the breast cancer 1 protein (BRCA1)77. The incidence of breast cancer in *BRCA1* mutation carriers is 2.4% and 1.7% in North America and Poland, respectively; these occurrences are significantly higher than the incidence of breast cancer in individuals without the *BRCA1* mutation85. Additionally, P4 induces cyclin D1 through the RANKL/RANK/IKK/NF-κB pathway78. Boopalan et al.79 reported that levels of cyclin D1 decreased in MCF-7 breast cancer cells when the PR was blocked by the selective progesterone receptor antagonist, mifepristone. With respect to the PI3K/Akt pathway, Wang et al.80,81 reported that P4 reduced the concentration of p27 in the nucleus and facilitated breast cancer cell proliferation through the PI3K/Akt pathway. In this process, P4 activated the PI3K/Akt pathway and subsequently increased the phosphorylation of p27 at T157 and pT198 sites, ultimately leading to the retention of p27 in the cytoplasm. Activation of kinase-interacting stathmin induced the phosphorylation of p27 at serine 10 (S10) within the nucleus, thereby promoting the transfer of p27 from the nucleus to the cytoplasm. The transfer of p27 attenuated the inhibition of cyclin-dependent kinase activity in the nucleus, which ultimately led to cell proliferation. With respect to the MAPK pathway, P4 interacts with the SH3 structural domain of the cytoplasmic signaling molecules through the amino-terminal polyproline motif of PR and activates the MAPK pathway, thereby affecting the transcription of cyclin D1 (CCND1)82. Additionally, Wang86,87 reported that P4 promoted the proliferation and migration of breast cancer cells associated with ERK activation resulting from the direct binding of PR and the SH3 domain of cellular Src (c-Src). #### Effects of progesterone metabolites on breast cancer cells Many tissues have progesterone-metabolizing enzymes, that act on different parts of the progesterone molecule.88 In breast cancer cells, P4 is converted into various products by different enzymes (**Figure 4A**). It can be reduced to 5α-dihydroprogesterone [also known as 5α-pregnane-3,20-dione (5αP)] by 5α-reductase, it can be converted to 3α-dihydroprogesterone [also known as 4-pregnen-3α-ol-20-one (3αHP)] by 3α-hydroxysteroid oxidoreductase (3α-HSO), or it can be metabolized to 20α-dihydroprogesterone [also known as 4-pregnen-20α-ol-3-one (20αHP)] by 20α-hydroxysteroid oxidoreductase (20α-HSO). ![Figure 4](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/19/4/432/F4.medium.gif) [Figure 4](http://www.cancerbiomed.org/content/19/4/432/F4) Figure 4 Metabolic pathways of progesterone (A) and medroxyprogesterone acetate (MPA) (B). A: Progesterone can be reduced to 5α-pregnane-3,20-dione (5αP) by 5α-reductase, or be converted to 3αHP by 3α-hydroxysteroid oxidoreductase (3α-HSO), or be metabolized to 20αHP through 20α-HSO in breast cells. The 5αP promotes breast cancer cell proliferation and inhibits breast cancer cell apoptosis, while 3αHP does the opposite. Abbreviations: 3α-HSO, 3α-hydroxysteroid oxidoreductase; 20α-HSO, 20α-hydroxysteroid oxidoreductase. B: the metabolic pathways of MPA in human liver microsomes. Previous studies89,90 reported that 5αP inhibited apoptosis and promoted the proliferation of breast cancer cells. In contrast, 3αHP promoted apoptosis and inhibited the proliferation of breast cancer cells. Due to certain technical limitations, it was difficult to obtain precise *in vivo* measurements of low progesterone levels in postmenopausal woman; hence, only a few studies have examined the probable effects of progesterone metabolites on breast cancer cells. Trabert et al.91 improved the detection limit of progesterone from 3 ng/dL to 0.1 ng/dL using liquid chromatography-tandem mass spectrometry, which has significantly contributed to further studies of the association of progesterone metabolites with the risks of breast cancer. A previous study89 reported that the ratios of 5αP/3αHP in breast tumors and nontumor tissues of breast cancer patients were 18.16 ± 1.3 and 0.61 ± 0.16, respectively, thereby revealing a nearly 30-fold difference between the 2 groups. Moreover, Trabert et al.17 reported that the 5αP concentration (mean: 8.0; SD: 3.6) in the blood of 405 breast cancer patients was approximately 3-fold higher than the 3αHP concentration (mean: 2.5; SD: 1.3) in the B˜FIT cohort (as previously described). However, the B˜FIT cohort17 results reported that the ratio of 5αP/3αHP was not associated with an increased risk of postmenopausal breast cancer (HR: 1.00; 95% CI: 0.97–1.04; *P* = 0.85). Nevertheless, among women in the lowest tertile of 3αHP (< 1.72 ng/dL), those who belonged in the highest tertile of 5αP (> 9.12 ng/dL) were associated with an almost double risk of breast cancer (T3 *vs.* T1; HR: 1.96; 95% CI: 1.01–3.81; *P* = 0.04; *P* = 0.08 for interaction) when compared to the women in the lowest tertile of the 5αP group. Initially, these data did not support the hypothesis that 5αP exposure was associated with an increased risk of breast cancer. However, genetic variations in 5α-reductase activity in breast tissues might lead to differences in the 5αP concentrations in different individuals; hence, serum measurements might not be indicative of the real levels92. In the future, further studies are required to determine the exact roles 5αP and 3αHP in the development of breast cancer, as well as to verify whether they can be used as molecular markers for detecting the increased risk of breast cancer. In summary, P4, which plays an essential role in normal breast development during puberty and pregnancy, might be a potent driver of breast cancer cell proliferation by multiple signaling pathways. Previous studies indicated that P4 metabolites, namely 5αP and 3αHP, had contrasting effects on breast cancer cell proliferation. These results contributed to a comprehensive understanding of the effects of P4 on breast cancer cell proliferation, which provide suggestions for further studies. ### Progestins #### MPA P4 is a C21-steroid hormone in which the pregnane skeleton contains 2 ketone groups (3, 20-dione), 1 each at the C3 and C20 positions, with a double bond between the C4 and C5 atoms6. MPA is a synthetic derivative of P4, and its progestogenic effect is similar to that of P4. However, even small structural changes might cause large variations in the functional effects of a molecule; for example, when compared with P4, MPA shows a relatively high progestogen activity when a methyl group is added to C693. Similar to P4, MPA can also promote mammary cell proliferation through the PR-mediated activation of RANKL/RANK, MAPK, PI3K/Akt, signal transducer, and activator of transcription 3 (Stat3), and other pathways. For example, with respect to the RANKL/RANK pathway, Schramek et al.94 reported that the MPA-induced proliferation of breast cancer cells was significantly reduced in RANK knockout Cre mice. With respect to the PI3K/Akt and MAPK/ERK pathways, Saitoh et al.95 reported that MPA induced CCND1 expression through the PI3K/Akt/NF-κB pathway. Similarly, Giulianelli et al.96 reported that MPA simultaneously recruited PR and ERα to the promoters of CCND1 and MYC, thereby promoting their expressions, and the MAPK/ERK, PI3K/Akt, and JAK/STAT pathways were involved in this process. With respect to the Stat pathway, Elizalde et al.97,98 reported MPA significantly upregulated P21CIPI and CCND1 expressions in T47D breast cancer cells, resulting from assembling the Stat3/ErbB-2/PR transcriptional complex, where Stat3 bound to the P21CIPI proximal promoter while both ErbB-2 and PR functioned as Stat3 co-activators. Furthermore, P21CIPI and CCND1 were essential for MPA-driven breast cancer growth, both *in vitro* and *in vivo*. Additionally, MPA activates the ERK and JNK pathways in MCF10A cells (PR-negative benign breast epithelial cells) through mPR, which could help us to better understand the importance of MPA and mPR in the development of breast cancer99. However, unlike P4, MPA has a high affinity for the glucocorticoid receptor (GR)100. Courtin et al.101,102 reported that MPA facilitated the expression of fatty acid synthetase (FAS) through GR in MCF-7 breast cancer cells, and that FAS was closely related to the development of breast cancer. Furthermore, MPA displays a high affinity for androgen receptor (AR), and it can affect the proliferation of breast cancer cells through the AR signaling pathway103. It has been reported that administration of a high concentration of MPA (100 nM) could inhibit cell growth by activating the AR103–105, and a high concentration of MPA (> 500 mg/day) has been previously used as hormonal therapy for treating advanced breast cancer patients106. Birrell et al.103 reported that low concentrations of MPA (< 10 nM) exhibited an anti-androgen role, thereby interfering with the AR signaling pathway107. This interpretation was based on experiments in which MPA inhibited DHT-induced AR-N/C interactions103. However, this assumption is controversial because the inhibition of N/C interactions did not necessarily reflect the activity of an antagonist108. #### Effects of MPA metabolites In the USA, MPA is the most commonly used progestin for MHT109. Siddique et al.110 suggested that MPA underwent metabolic activation to generate genotoxic ROS through cytochrome P450- and NADPH-dependent processes. Previously, a study on synthetic progestins reported that the double bond between carbon 6 (C6) and carbon 7 (C7) might be significant with respect to genotoxicity110. MPA lacks the double bond between C6 and C7, so it produces various forms of quinones through redox cycling, which can lead to genotoxic effects. In contrast, megestrol acetate might undergo nucleophilic reactions and generate free radicals due to the presence of the C6-C7 double bond111 (**Figure 5**). The CYP-mediated biotransformation usually serves as the first step in steroid elimination. CYP3A4 is the major cytochrome P450 isoform involved in the metabolism of MPA113. The formation rate and relative abundances of the MPA metabolites follows the order, M-2 > M-4 > M-3 > M-5 ≈ M-1 in human liver microsomes113 (**Figure 4B**). M-2, M-3, and M-4 are the dominant metabolites, accounting for more than 85% of the 5 metabolites. Although only MPA is considered as the active form113, the biological activities of its metabolites need to be further studied to determine their relationships with breast cancer. Moreover, the mechanism of MPA metabolism within the breast tissue is still not clear. Therefore, a comprehensive study of the metabolic pathways of MPA is needed. ![Figure 5](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/19/4/432/F5.medium.gif) [Figure 5](http://www.cancerbiomed.org/content/19/4/432/F5) Figure 5 Possible mechanism of genotoxicity of synthetic progestins. The presence of the double bond between carbon 6 and carbon 7 might be significant for genotoxicity. Figure adapted from Siddique et al.112. Although many epidemiological studies have shown that the use of MPA in EPT was associated with an increased risk of breast cancer, the mechanism by which MPA affects the progression of breast cancer has not been fully elucidated. It is known that MPA binds to multiple steroid hormone receptors, such as PR, mPR, GR, and AR, and promotes the proliferation of breast cancer cells. However, administering higher and lower than normal doses of MPA has opposing biological effects on breast cancer proliferation due to their effects on the AR signaling pathway. Moreover, the mechanism by which MPA is metabolized in breast cancer cells and whether MPA metabolites increase the risk of breast cancer are yet to be determined. #### NET NET, which is mainly used in northern Europe, is a progestin compound with structural similarities to testosterone. In addition to its progestogenic effects, it also has very weak androgenic and estrogenic effects109,114. Previous studies115,116 have reported that 10−8 M–10−5 M NET significantly promoted the growth of MCF-7 cells overexpressing PGRMC1, and that this proliferative effect could be inhibited by administering the PGRMC1 antagonist, AG-205. Willibald et al.117 discovered that protein kinase CK2 (formerly known as casein kinase II) was involved in the phosphorylation of the PGRMC1 Ser181 site in MCF7/PGRMC1 cells (MCF-7 cells overexpressing PGRMC1) treated with NET, and that this was a prerequisite for the NET-induced proliferation of MCF7/PGRMC1 cells. Protein kinase CK2 is a serine/threonine protein kinase that is widely expressed and highly conserved in eukaryotic cells118. It is involved in various cellular processes, including metabolism, proliferation, differentiation, and apoptosis119. Moreover, previous studies120,121 reported that the E2/NET combination enhanced the proliferation of PGRMC1-overexpressing breast cancer cells, both *in vivo* and *in vitro*. The MWS observational study10 reported that the relative risk of using estrogen + NET with respect to the incidence of breast cancer was 1.53 (95% CI: 1.35–1.75), which was consistent with numerous previous studies. In contrast, in another study, the Danish Osteoporosis Prevention Study (DOPS; *n* = 1,006)23, reported that administration of E2 + NET did not increase the risk of breast cancer. Because PGRMC1 is expressed in breast tissue and overexpressed in breast cancer tissue73, the molecular mechanism of PGRMC1 associated with the proliferation of breast cancer cells requires further study. It is important to determine whether breast cells overexpressing PGRMC1 are more likely to develop into tumor cells in women receiving E2/NET hormone therapy, which would help to determine the risks of MHT in relation to breast cancer. ## Conclusions and perspectives By combining the results of recent MHT clinical trial studies, it has been suggested that there is a lower risk of developing breast cancer in women who use ET and E2 + P4, when compared to the breast cancer risk in women who use E2 + MPA. However, there are differences among the clinical trials with respect to the basic profile of the study population (age composition, time of menopause, and time of initiation of MHT), drug use (estrogen and progestin type, dosage, dosage form, and time of administration), dosing regimen (sequential or combined), and route of administration. Due to the great heterogeneity of these studies, it is difficult and inappropriate to reach a unified conclusion. Hence, long-term studies on the associations between increased risks of breast cancer and the underlying mechanisms of hormone therapy using different estrogen and progesterone regimens in early menopausal women are needed108. To understand the molecular mechanisms of estrogen and progestogens and their relationships with breast cancer cell proliferation, we analyzed previous studies on estrogen and progesterone receptors in breast cancer cells as well as their signaling pathways, respectively. The genomic and non-genomic effects mediated by the hormone receptors played an important role in the proliferation of breast cancer cells. Furthermore, the biological functions of estrogen and progestogen metabolites were also linked with the proliferation of breast cancer cells, although this area of research required further study. Current studies on the metabolites of E2, MPA, and NET have focused on liver tissues. Notably, the metabolic pathways of all 3 were similar, and were associated with cytochrome P450 enzymes. Because E2 and MPA lack the C6-C7 double bond, they can generate various quinones through redox cycling, to produce genotoxic effects. However, the relevant metabolic enzymes and metabolic pathways of E2, MPA, and NET in breast tissue are still not clear, and the concentrations of their metabolites are difficult to measure by conventional methods due to their low concentrations. These limitations provide a challenge for further research dealing with the associations of the estrogen and progestogen metabolites with the risks of breast cancer. ## Grant support This study was supported by grants from the National Natural Science Foundation of China (Grant Nos. 61673024 and 81971348), the Clinical Medicine Plus X-Young Scholar Project, Peking University, and the Fundamental Research Funds for the Central University (Grant No. PKU2018LCXQ001). ## Conflict of interest statement No potential conflicts of interest are disclosed. ## Author contributions Conceived and designed the analysis: Yu Deng and Hongyan Jin. Collected the data: Yu Deng and Hongyan Jin. Wrote the manuscript: Yu Deng and Hongyan Jin. ## Acknowledgements We thank Dr. He Huang at the Peking University First Hospital for linguistic assistance during the revision of this manuscript. * Received June 5, 2021. * Accepted September 30, 2021. * Copyright: © 2022, Cancer Biology & Medicine [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/) This is an open access article distributed under the terms of the [Creative Commons Attribution License (CC BY) 4.0](https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. ## References 1. 1. Santen RJ, Allred DC, Ardoin SP, Archer DF, Boyd N, Braunstein GD, et al. Postmenopausal hormone therapy: an endocrine society scientific statement. J Clin Endocrinol Metab. 2010; 95: s1–66. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/jc.2009-2509&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20566620&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 2. 2. Stramba-Badiale M. Postmenopausal hormone therapy and the risk of cardiovascular disease. J Cardiovasc Med (Hagerstown). 2009; 10: 303–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19430340&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 3. 3. Santoro N, Epperson CN, Mathews SB. Menopausal symptoms and their management. Endocrinol Metab Clin North Am. 2015; 44: 497–515. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ecl.2015.05.001&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26316239&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 4. 4. Baber RJ, Panay N, Fenton A. 2016 IMS recommendations on women’s midlife health and menopause hormone therapy. Climacteric. 2016; 19: 109–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3109/13697137.2015.1129166&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26872610&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 5. 5. Stanczyk FZ. Pharmacokinetics and potency of progestins used for hormone replacement therapy and contraception. Rev Endocr Metab Disord. 2002; 3: 211–24. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1023/A:1020072325818&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12215716&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 6. 6. Sitruk-Ware R. Pharmacological profile of progestins. Maturitas. 2004; 47: 277–83. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.maturitas.2004.01.001&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15063480&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000220897700007&link_type=ISI) 7. 7. Schindler AE, Campagnoli C, Druckmann R, Huber J, Pasqualini JR, Schweppe KW, et al. Classification and pharmacology of progestins. Maturitas. 2008; 61: 171–80. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.maturitas.2008.11.013&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19434889&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 8. 8. Brisken C, Hess K, Jeitziner R. Progesterone and overlooked endocrine pathways in breast cancer pathogenesis. Endocrinology. 2015; 156: 3442–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/en.2015-1392&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26241069&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 9. 9. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, 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. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.288.3.321&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12117397&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000176807000024&link_type=ISI) 10. 10. Beral V. Breast cancer and hormone-replacement therapy in the million women study. Lancet. 2003; 362: 419–27. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0140-6736(03)14065-2&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12927427&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000184651100007&link_type=ISI) 11. 11. 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. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.20710&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15551359&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000227223100018&link_type=ISI) 12. 12. 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. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s10549-007-9523-x&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17333341&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000251235200012&link_type=ISI) 13. 13. Santen RJ, Yue W. Cause or prevention of breast cancer with estrogens: Analysis from tumor biologic data, growth kinetic model and women’s health initiative study. Climacteric. 2019; 22: 3–12. 14. 14. Santen RJ, Yue W, Heitjan DF. Modeling of the growth kinetics of occult breast tumors: role in interpretation of studies of prevention and menopausal hormone therapy. Cancer Epidemiol Biomarkers Prev. 2012; 21: 1038–48. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiY2VicCI7czo1OiJyZXNpZCI7czo5OiIyMS83LzEwMzgiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xOS80LzQzMi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 15. 15. Dallal CM, Tice JA, Buist DS, Bauer DC, Lacey Jr JV, Cauley JA, et al. Estrogen metabolism and breast cancer risk among postmenopausal women: a case-cohort study within B˜FIT. Carcinogenesis. 2014; 35: 346–55. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/carcin/bgt367&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24213602&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 16. 16. Mackey RH, Fanelli TJ, Modugno F, Cauley JA, McTigue KM, Brooks MM, et al. Hormone therapy, estrogen metabolism, and risk of breast cancer in the women’s health initiative hormone therapy trial. Cancer Epidemiol Biomarkers Prev. 2012; 21: 2022–32. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiY2VicCI7czo1OiJyZXNpZCI7czoxMDoiMjEvMTEvMjAyMiI7czo0OiJhdG9tIjtzOjE4OiIvY2JtLzE5LzQvNDMyLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 17. 17. Trabert B, Bauer DC, Buist DSM, Cauley JA, Falk RT, Geczik AM, et al. Association of circulating progesterone with breast cancer risk among postmenopausal women. JAMA Netw Open. 2020; 3: e203645. 18. 18. Miao S, Yang F, Wang Y, Shao C, Zava DT, Ding Q, et al. 4-Hydroxy estrogen metabolite, causing genomic instability by attenuating the function of spindle-assembly checkpoint, can serve as a biomarker for breast cancer. Am J Transl Res. 2019; 11: 4992–5007. 19. 19. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: The women’s health initiative randomized controlled trial. JAMA. 2004; 291: 1701–12. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.291.14.1701&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15082697&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000220788700027&link_type=ISI) 20. 20. LaCroix AZ, Chlebowski RT, Manson JE, Aragaki AK, Johnson KC, Martin L, et al. Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy: a randomized controlled trial. JAMA. 2011; 305: 1305–14. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.2011.382&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21467283&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000289162400019&link_type=ISI) 21. 21. Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE, Prentice RL, et al. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the women’s health initiative randomized trials. JAMA. 2013; 310: 1353–68. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.2013.278040&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24084921&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000325098100021&link_type=ISI) 22. 22. Manson JE, Aragaki AK, Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, et al. Menopausal hormone therapy and long-term all-cause and cause-specific mortality: the women’s health initiative randomized trials. JAMA. 2017; 318: 927–38. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.2017.11217&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28898378&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 23. 23. Schierbeck LL, Rejnmark L, Tofteng CL, Stilgren L, Eiken P, Mosekilde L, et al. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ. 2012; 345: e6409. 24. 24. Bakken K, Fournier A, Lund E, Waaseth M, Dumeaux V, Clavel-Chapelon F, et al. Menopausal hormone therapy and breast cancer risk: impact of different treatments. The european prospective investigation into cancer and nutrition. Int J Cancer. 2011; 128: 144–56. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.25314&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20232395&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000284209700015&link_type=ISI) 25. 25. 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. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s10549-014-2934-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24781971&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000336370200026&link_type=ISI) 26. 26. Holm M, Olsen A, Au Yeung SL, Overvad K, Lidegaard Ø, Kroman N, et al. Pattern of mortality after menopausal hormone therapy: long-term follow up in a population-based cohort. BJOG. 2019; 126: 55–63. 27. 27. Vinogradova Y, Coupland C, Hippisley-Cox J. Use of hormone replacement therapy and risk of breast cancer: nested case-control studies using the QResearch and CPRD databases. BMJ. 2020; 371: m3873. 28. 28. Gompel A, Plu-Bureau G. Progesterone, progestins and the breast in menopause treatment. Climacteric. 2018; 21: 326–32. 29. 29. Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, et al. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997; 138: 863–70. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/en.138.3.863&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=9048584&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=A1997WJ73900002&link_type=ISI) 30. 30. Denver N, Khan S, Stasinopoulos I, Church C, Homer NZ, MacLean MR, et al. Derivatization enhances analysis of estrogens and their bioactive metabolites in human plasma by liquid chromatography tandem mass spectrometry. Anal Chim Acta. 2019; 1054: 84–94. 31. 31. Gustafsson JA. What pharmacologists can learn from recent advances in estrogen signalling. Trends Pharmacol Sci. 2003; 24: 479–85. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0165-6147(03)00229-3&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12967773&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000185448600009&link_type=ISI) 32. 32. Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, et al. Mechanisms of estrogen action. Physiol Rev. 2001; 81: 1535–65. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1152/physrev.2001.81.4.1535&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11581496&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000171442500005&link_type=ISI) 33. 33. Wilkenfeld SR, Lin C, Frigo DE. Communication between genomic and non-genomic signaling events coordinate steroid hormone actions. Steroids. 2018; 133: 2–7. 34. 34. Hayashi S, Yamaguchi Y. Estrogen signaling pathway and hormonal therapy. Breast Cancer. 2008; 15: 256–61. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s12282-008-0070-z&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18818989&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 35. 35. Lösel R, Wehling M. Nongenomic actions of steroid hormones. Nat Rev Mol Cell Biol. 2003; 4: 46–56. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nrm1009&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12511868&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000180170900015&link_type=ISI) 36. 36. Losel RM, Falkenstein E, Feuring M, Schultz A, Tillmann HC, Rossol-Haseroth K, et al. Nongenomic steroid action: controversies, questions, and answers. Physiol Rev. 2003; 83: 965–1016. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1152/physrev.00003.2003&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12843413&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000183941300008&link_type=ISI) 37. 37. Saczko J, Michel O, Chwilkowska A, Sawicka E, Maczynska J, Kulbacka J. Estrogen receptors in cell membranes: regulation and signaling. Adv Anat Embryol Cell Biol. 2017; 227: 93–105. 38. 38. Levin ER. Bidirectional signaling between the estrogen receptor and the epidermal growth factor receptor. Mol Endocrinol. 2003; 17: 309–17. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/me.2002-0368&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12554774&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000181424400001&link_type=ISI) 39. 39. Giuliano M, Trivedi MV, Schiff R. Bidirectional crosstalk between the estrogen receptor and human epidermal growth factor receptor 2 signaling pathways in breast cancer: molecular basis and clinical implications. Breast Care (Basel). 2013; 8: 256–62. 40. 40. Raffo D, Pontiggia O, Bal de Kier Joffe E, Simian M. Non-genomic actions of estradiol and 4-OH-tamoxifen on murine breast cancer cells. Oncol Rep. 2015; 33: 439–47. 41. 41. Aronica SM, Kraus WL, Katzenellenbogen BS. Estrogen action via the cAMP signaling pathway: stimulation of adenylate cyclase and cAMP-regulated gene transcription. Proc Natl Acad Sci U S A. 1994; 91: 8517–21. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiOTEvMTgvODUxNyI7czo0OiJhdG9tIjtzOjE4OiIvY2JtLzE5LzQvNDMyLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 42. 42. Garrido P, Morán J, Alonso A, González S, González C. 17β-estradiol activates glucose uptake via GLUT4 translocation and PI3K/Akt signaling pathway in MCF-7 cells. Endocrinology. 2013; 154: 1979–89. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/en.2012-1558&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23546602&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000319208200007&link_type=ISI) 43. 43. Carmeci C, Thompson DA, Ring HZ, Francke U, Weigel RJ. Identification of a gene (GPR30) with homology to the G-protein-coupled receptor superfamily associated with estrogen receptor expression in breast cancer. Genomics. 1997; 45: 607–17. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1006/geno.1997.4972&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=9367686&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=A1997YF97300015&link_type=ISI) 44. 44. Yang H, Wang C, Liao H, Wang Q. Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR-124/CD151 pathway. Oncol Lett. 2021; 21: 432. 45. 45. Filardo EJ, Quinn JA, Bland KI, Frackelton Jr AR. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol. 2000; 14: 1649–60. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/me.14.10.1649&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11043579&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000089633200011&link_type=ISI) 46. 46. Vivacqua A, Romeo E, De Marco P, De Francesco EM, Abonante S, Maggiolini M. GPER mediates the Egr-1 expression induced by 17β-estradiol and 4-hydroxitamoxifen in breast and endometrial cancer cells. Breast Cancer Res Treat. 2012; 133: 1025–35. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s10549-011-1901-8&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22147081&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000305914900020&link_type=ISI) 47. 47. Deng Y, Miki Y, Nakanishi A. Estradiol/GPER affects the integrity of mammary duct-like structures in vitro. Scientific Reports. 2020; 10: 1386. 48. 48. Santen RJ, Yue W, Wang J-P. Estrogen metabolites and breast cancer. Steroids. 2015; 99: 61–6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.steroids.2014.08.003&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25168343&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 49. 49. Samavat H, Kurzer MS. Estrogen metabolism and breast cancer. Cancer Lett. 2015; 356: 231–43. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.canlet.2014.04.018&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24784887&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 50. 50. Gupta M, McDougal A, Safe S. Estrogenic and antiestrogenic activities of 16alpha- and 2-hydroxy metabolites of 17beta-estradiol in MCF-7 and T47D human breast cancer cells. J Steroid Biochem Mol Biol. 1998; 67: 413–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0960-0760(98)00135-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=10030690&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000078389400005&link_type=ISI) 51. 51. Suto A, Telang NT, Tanino H, Takeshita T, Ohmiya H, Osborne MP, et al. In vitro and in vivo modulation of growth regulation in the human breast cancer cell line MCF-7 by estradiol metabolites. Breast Cancer. 1999; 6: 87–92. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11091698&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 52. 52. Yager JD. Mechanisms of estrogen carcinogenesis: the role of E2/E1-quinone metabolites suggests new approaches to preventive intervention--a review. Steroids. 2015; 99: 56–60. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.steroids.2014.08.006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25159108&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 53. 53. Lareef MH, Garber J, Russo PA, Russo IH, Heulings R, Russo J. The estrogen antagonist ICI-182-780 does not inhibit the transformation phenotypes induced by 17-beta-estradiol and 4-OH estradiol in human breast epithelial cells. Int J Oncol. 2005; 26: 423–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15645127&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000226481500015&link_type=ISI) 54. 54. Tian H, Gao Z, Wang G, Li H, Zheng J. Estrogen potentiates reactive oxygen species (ROS) tolerance to initiate carcinogenesis and promote cancer malignant transformation. Tumour Biol. 2016; 37: 141–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s13277-015-4370-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26566628&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 55. 55. Zahid M, Kohli E, Saeed M, Rogan E, Cavalieri E. The greater reactivity of estradiol-3,4-quinone vs estradiol-2,3-quinone with DNA in the formation of depurinating adducts: Implications for tumor-initiating activity. Chem Res Toxicol. 2006; 19: 164–72. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1021/tx050229y&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16411670&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000234822600021&link_type=ISI) 56. 56. Zahid M, Saeed M, Lu F, Gaikwad N, Rogan E, Cavalieri E. Inhibition of catechol-O-methyltransferase increases estrogen-DNA adduct formation. Free Radic Biol Med. 2007; 43: 1534–40. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.freeradbiomed.2007.08.005&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17964424&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 57. 57. Sood D, Johnson N, Jain P, Siskos AP, Bennett M, Gilham C, et al. CYP3A7*1C allele is associated with reduced levels of 2-hydroxylation pathway oestrogen metabolites. British J Cancer. 2017; 116: 382–8. 58. 58. Bradlow HL, Davis DL, Lin G, Sepkovic D, Tiwari R. Effects of pesticides on the ratio of 16 alpha/2-hydroxyestrone: A biologic marker of breast cancer risk. Environ Health Perspect. 1995; 103 (Suppl 7): 147–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1289/ehp.95103s6147&link_type=DOI) 59. 59. Lewis JS, Thomas TJ, Klinge CM, Gallo MA, Thomas T. Regulation of cell cycle and cyclins by 16alpha-hydroxyestrone in MCF-7 breast cancer cells. J Mol Endocrinol. 2001; 27: 293–307. [Abstract](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoiam1lIjtzOjU6InJlc2lkIjtzOjg6IjI3LzMvMjkzIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 60. 60. Lewis JS, Thomas TJ, Pestell RG, Albanese C, Gallo MA, Thomas T. Differential effects of 16alpha-hydroxyestrone and 2-methoxyestradiol on cyclin D1 involving the transcription factor ATF-2 in MCF-7 breast cancer cells. J Mol Endocrinol. 2005; 34: 91–105. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoiam1lIjtzOjU6InJlc2lkIjtzOjc6IjM0LzEvOTEiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xOS80LzQzMi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 61. 61. Falk RT, Manson JE, Barnabei VM, Anderson GL, Brinton LA, Rohan TE, et al. Estrogen metabolism in menopausal hormone users in the women’s health initiative observational study: Does it differ between estrogen plus progestin and estrogen alone? Int J Cancer. 2019; 144: 730–40. 62. 62. Mueck AO, Ruan X, Seeger H, Fehm T, Neubauer H. Genomic and non-genomic actions of progestogens in the breast. J Steroid Biochem Mol Biol. 2014; 142: 62–7. 63. 63. Anderson E. Progesterone receptors – animal models and cell signaling in breast cancer: the role of oestrogen and progesterone receptors in human mammary development and tumorigenesis. Breast Cancer Res. 2002; 4: 197. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/bcr452&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12223124&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000177785100011&link_type=ISI) 64. 64. Boonyaratanakornkit V, Bi Y, Rudd M, Edwards DP. The role and mechanism of progesterone receptor activation of extra-nuclear signaling pathways in regulating gene transcription and cell cycle progression. Steroids. 2008; 73: 922–8. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.steroids.2008.01.010&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18321550&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000257519900014&link_type=ISI) 65. 65. Jacobsen BM, Horwitz KB. Progesterone receptors, their isoforms and progesterone regulated transcription. Mol Cell Endocrinol. 2012; 357: 18–29. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.mce.2011.09.016&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21952082&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 66. 66. Graham JD, Yeates C, Balleine RL, Harvey SS, Milliken JS, Bilous AM, et al. Characterization of progesterone receptor A and B expression in human breast cancer. Cancer Res. 1995; 55: 5063–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI1NS8yMS81MDYzIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 67. 67. Hopp TA, Weiss HL, Hilsenbeck SG, Cui Y, Allred DC, Horwitz KB, et al. Breast cancer patients with progesterone receptor PR-A-rich tumors have poorer disease-free survival rates. Clin Cancer Res. 2004; 10: 2751–60. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMTAvOC8yNzUxIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 68. 68. Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martinez P, et al. Progesterone receptor isoform ratio: a breast cancer prognostic and predictive factor for antiprogestin responsiveness. J Natl Cancer Inst. 2017; 109: djw317. 69. 69. Mote PA, Bartow S, Tran N, Clarke CL. Loss of co-ordinate expression of progesterone receptors A and B is an early event in breast carcinogenesis. Breast Cancer Res Treat. 2002; 72: 163–72. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1023/A:1014820500738&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12038707&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000174634400008&link_type=ISI) 70. 70. Valadez-Cosmes P, Vázquez-Martínez ER, Cerbón M, Camacho-Arroyo I. Membrane progesterone receptors in reproduction and cancer. Mol Cell Endocrinol. 2016; 434: 166–75. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.mce.2016.06.027&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27368976&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 71. 71. Zhu Y, Hanna RN, Schaaf MJ, Spaink HP, Thomas P. Candidates for membrane progestin receptors--past approaches and future challenges. Comp Biochem Physiol C Toxicol Pharmacol. 2008; 148: 381–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cbpc.2008.05.019&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18602498&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000261600100011&link_type=ISI) 72. 72. Kimura I, Nakayama Y, Konishi M, Terasawa K, Ohta M, Itoh N, et al. Functions of MAPR (membrane-associated progesterone receptor) family members as heme/steroid-binding proteins. Curr Protein Pept Sci. 2012; 13: 687–96. 73. 73. Zhang Y, Ruan X, Willibald M, Seeger H, Fehm T, Neubauer H, et al. May progesterone receptor membrane component 1 (PGRMC1) predict the risk of breast cancer? Gynecol Endocrinol. 2016; 32: 58–60. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3109/09513590.2015.1078303&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26303031&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 74. 74. Ruan X, Zhang Y, Mueck AO, Willibald M, Seeger H, Fehm T, et al. Increased expression of progesterone receptor membrane component 1 is associated with aggressive phenotype and poor prognosis in ER-positive and negative breast cancer. Menopause. 2017; 24: 203–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1097/GME.0000000000000739&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27749742&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 75. 75. Causey MW, Huston LJ, Harold DM, Charaba CJ, Ippolito DL, Hoffer ZS, et al. Transcriptional analysis of novel hormone receptors PGRMC1 and PGRMC2 as potential biomarkers of breast adenocarcinoma staging. J Surg Res. 2011; 171: 615–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.jss.2010.04.034&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20655063&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 76. 76. Sitruk-Ware R. Pharmacological profile of progestins. Maturitas. 2008; 61: 151–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.maturitas.2008.11.011&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19434887&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 77. 77. Nolan E, Vaillant F, Branstetter D, Pal B, Giner G, Whitehead L, et al. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers. Nat Med. 2016; 22: 933–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nm.4118&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27322743&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 78. 78. Cao Y, Bonizzi G, Seagroves TN, Greten FR, Johnson R, Schmidt EV, et al. IKKalpha provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell. 2001; 107: 763–75. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0092-8674(01)00599-2&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11747812&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000172771800009&link_type=ISI) 79. 79. Boopalan T, Arumugam A, Parada J, Saltzstein E, Lakshmanaswamy R. Receptor activator for nuclear factor-κB ligand signaling promotes progesterone-mediated estrogen-induced mammary carcinogenesis. Cancer Sci. 2015; 106: 25–33. 80. 80. Wang HC, Lee WS. Molecular mechanisms underlying progesterone-induced cytoplasmic retention of p27 in breast cancer cells. J Steroid Biochem Mol Biol. 2018; 183: 202–9. 81. 81. Boehm M, Yoshimoto T, Crook MF, Nallamshetty S, True A, Nabel GJ, et al. A growth factor-dependent nuclear kinase phosphorylates p27(Kip1) and regulates cell cycle progression. EMBO J. 2002; 21: 3390–401. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZW1ib2pubCI7czo1OiJyZXNpZCI7czoxMDoiMjEvMTMvMzM5MCI7czo0OiJhdG9tIjtzOjE4OiIvY2JtLzE5LzQvNDMyLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 82. 82. Boonyaratanakornkit V, Scott MP, Ribon V, Sherman L, Anderson SM, Maller JL, et al. Progesterone receptor contains a proline-rich motif that directly interacts with SH3 domains and activates c-Src family tyrosine kinases. Mol Cell. 2001; 8: 269–80. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1097-2765(01)00304-5&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11545730&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000170827000005&link_type=ISI) 83. 83. Faivre E, Skildum A, Pierson-Mullany L, Lange CA. Integration of progesterone receptor mediated rapid signaling and nuclear actions in breast cancer cell models: role of mitogen-activated protein kinases and cell cycle regulators. Steroids. 2005; 70: 418–26. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.steroids.2005.02.012&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15862825&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000229342800011&link_type=ISI) 84. 84. Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL, et al. Progesterone induces adult mammary stem cell expansion. Nature. 2010; 465: 803–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature09091&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20445538&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000278551800050&link_type=ISI) 85. 85. Lubinski J, Huzarski T, Byrski T, Lynch HT, Cybulski C, Ghadirian P, et al. The risk of breast cancer in women with a BRCA1 mutation from North America and Poland. Int J Cancer. 2012; 131: 229–34. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.26369&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21834074&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 86. 86. Wang H-C, Lee W-S. Molecular mechanisms underlying progesterone-enhanced breast cancer cell migration. Sci. Rep. 2016; 6: 31509. 87. 87. Wang HC, Huo YN, Lee WS. Folic acid prevents the progesterone-promoted proliferation and migration in breast cancer cell lines. Eur J Nutr. 2020; 59: 2333–44. 88. 88. Wiebe JP. Progesterone metabolites in breast cancer. Endocr Relat Cancer. 2006; 13: 717–38. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZXJjIjtzOjU6InJlc2lkIjtzOjg6IjEzLzMvNzE3IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 89. 89. Wiebe JP, Muzia D, Hu J, Szwajcer D, Hill SA, Seachrist JL. The 4-pregnene and 5alpha-pregnane progesterone metabolites formed in nontumorous and tumorous breast tissue have opposite effects on breast cell proliferation and adhesion. Cancer Res. 2000; 60: 936–43. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjg6IjYwLzQvOTM2IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 90. 90. Wiebe JP, Beausoleil M, Zhang G, Cialacu V. Opposing actions of the progesterone metabolites, 5alpha-dihydroprogesterone (5alphaP) and 3alpha-dihydroprogesterone (3alphaHP) on mitosis, apoptosis, and expression of Bcl-2, Bax and p21 in human breast cell lines. J Steroid Biochem Mol Biol. 2010; 118: 125–32. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19931389&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 91. 91. Trabert B, Falk RT, Stanczyk FZ, McGlynn KA, Brinton LA, Xu X. Reproducibility of an assay to measure serum progesterone metabolites that may be related to breast cancer risk using liquid chromatography-tandem mass spectrometry. Horm Mol Biol Clin Investig. 2015; 23: 79–84. 92. 92. Khan SA. Progesterone exposure and breast cancer risk-addressing barriers. JAMA Netw Open. 2020; 3: e203608. 93. 93. Cabeza M, Heuze Y, Sánchez A, Garrido M, Bratoeff E. Recent advances in structure of progestins and their binding to progesterone receptors. J Enzyme Inhib Med Chem. 2015; 30: 152–9. 94. 94. Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010; 468: 98–102. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature09387&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20881962&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000283786900043&link_type=ISI) 95. 95. Saitoh M, Ohmichi M, Takahashi K, Kawagoe J, Ohta T, Doshida M, et al. Medroxyprogesterone acetate induces cell proliferation through up-regulation of cyclin D1 expression via phosphatidylinositol 3-kinase/Akt/nuclear factor-kappaB cascade in human breast cancer cells. Endocrinology. 2005; 146: 4917–25. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/en.2004-1535&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16123159&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000232585200039&link_type=ISI) 96. 96. Giulianelli S, Vaqué JP, Soldati R, Wargon V, Vanzulli SI, Martins R, et al. Estrogen receptor alpha mediates progestin-induced mammary tumor growth by interacting with progesterone receptors at the cyclin D1/MYC promoters. Cancer Res. 2012; 72: 2416–27. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjk6IjcyLzkvMjQxNiI7czo0OiJhdG9tIjtzOjE4OiIvY2JtLzE5LzQvNDMyLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 97. 97. Diaz Flaqué MC, Vicario R, Proietti CJ, Izzo F, Schillaci R, Elizalde PV. Progestin drives breast cancer growth by inducing p21(CIP1) expression through the assembly of a transcriptional complex among Stat3, progesterone receptor and ErbB-2. Steroids. 2013; 78: 559–67. 98. 98. Béguelin W, Díaz Flaqué MC, Proietti CJ, Cayrol F, Rivas MA, Tkach M, et al. Progesterone receptor induces ErbB-2 nuclear translocation to promote breast cancer growth via a novel transcriptional effect: ErbB-2 function as a coactivator of Stat3. Mol Cell Biol. 2010; 30: 5456–72. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoibWNiIjtzOjU6InJlc2lkIjtzOjEwOiIzMC8yMy81NDU2IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 99. 99. Salazar M, Lerma-Ortiz A, Hooks GM, Ashley AK, Ashley RL. Progestin-mediated activation of MAPK and AKT in nuclear progesterone receptor negative breast epithelial cells: the role of membrane progesterone receptors. Gene. 2016; 591: 6–13. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.gene.2016.06.044&link_type=DOI) 100.100. Africander D, Verhoog N, Hapgood JP. Molecular mechanisms of steroid receptor-mediated actions by synthetic progestins used in HRT and contraception. Steroids. 2011; 76: 636–52. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.steroids.2011.03.001&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21414337&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000291901800002&link_type=ISI) 101.101. Courtin A, Communal L, Vilasco M, Cimino D, Mourra N, de Bortoli M, et al. Glucocorticoid receptor activity discriminates between progesterone and medroxyprogesterone acetate effects in breast cells. Breast Cancer Res Treat. 2012; 131: 49–63. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s10549-011-1394-5&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21336598&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000298006300006&link_type=ISI) 102.102. Esslimani-Sahla M, Thezenas S, Simony-Lafontaine J, Kramar A, Lavaill R, Chalbos D, et al. Increased expression of fatty acid synthase and progesterone receptor in early steps of human mammary carcinogenesis. Int J Cancer. 2007; 120: 224–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.22202&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17044016&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 103.103. Birrell SN, Butler LM, Harris JM, Buchanan G, Tilley WD. Disruption of androgen receptor signaling by synthetic progestins may increase risk of developing breast cancer. FASEB J. 2007; 21: 2285–93. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1096/fj.06-7518com&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17413000&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000248454400003&link_type=ISI) 104.104. Izuo M, Iino Y, Endo K. Oral high-dose medroxyprogesterone acetate (MAP) in treatment of advanced breast cancer. A preliminary report of clinical and experimental studies. Breast Cancer Res Treat. 1981; 1: 125–30. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=6216931&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 105.105. Bentel JM, Birrell SN, Pickering MA, Holds DJ, Horsfall DJ, Tilley WD. Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells. Mol Cell Endocrinol. 1999; 154: 11–20. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0303-7207(99)00109-4&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=10509795&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000082459800002&link_type=ISI) 106.106. Ganzina F. High-dose medroxyprogesterone acetate (MPA) treatment in advanced breast cancer. A review. Tumori. 1979; 65: 563–85. 107.107. Kemppainen JA, Langley E, Wong CI, Bobseine K, Kelce WR, Wilson EM. Distinguishing androgen receptor agonists and antagonists: distinct mechanisms of activation by medroxyprogesterone acetate and dihydrotestosterone. Mol Endocrinol. 1999; 13: 440–54. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1210/me.13.3.440&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=10077001&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000078929400009&link_type=ISI) 108.108. Stanczyk FZ, Bhavnani BR. Use of medroxyprogesterone acetate for hormone therapy in postmenopausal women: is it safe? J Steroid Biochem Mol Biol. 2014; 142: 30–8. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.jsbmb.2013.11.011&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24291402&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 109.109. Campagnoli C, Clavel-Chapelon F, Kaaks R, Peris C, Berrino F. Progestins and progesterone in hormone replacement therapy and the risk of breast cancer. J Steroid Biochem Mol Biol. 2005; 96: 95–108. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.jsbmb.2005.02.014&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15908197&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000231515100001&link_type=ISI) 110.110. Siddique YH, Ara G, Beg T, Afzal M. Genotoxic potential of medroxyprogesterone acetate in cultured human peripheral blood lymphocytes. Life Sci. 2006; 80: 212–8. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.lfs.2006.09.005&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17023004&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 111.111. Siddique YH, Beg T, Afzal M. Antigenotoxic effects of ascorbic acid against megestrol acetate-induced genotoxicity in mice. Hum Exp Toxicol. 2005; 24: 121–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1191/0960327104ht508oa&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15901051&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 112.112. Siddique YH, Afzal M. A review on the genotoxic effects of some synthetic progestins. Int J Pharmacol. 2008; 4: 410–30. 113.113. Zhang J-W, Liu Y, Zhao J-Y, Wang L-M, Ge G-B, Gao Y, et al. Metabolic profiling and cytochrome P450 reaction phenotyping of medroxyprogesterone acetate. Drug Metab Dispos. 2008; 36: 2292. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZG1kIjtzOjU6InJlc2lkIjtzOjEwOiIzNi8xMS8yMjkyIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTkvNC80MzIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 114.114. Schoonen WG, Deckers GH, de Gooijer ME, de Ries R, Kloosterboer HJ. Hormonal properties of norethisterone, 7alpha-methyl-norethisterone and their derivatives. J Steroid Biochem Mol Biol. 2000; 74: 213–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0960-0760(00)00125-4&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11162927&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 115.115. Schneck H, Ruan X, Seeger H, Cahill MA, Fehm T, Mueck AO, et al. Membrane-receptor initiated proliferative effects of dienogest in human breast cancer cells. Gynecol Endocrinol. 2013; 29: 160–2. 116.116. Ruan X, Neubauer H, Yang Y, Schneck H, Schultz S, Fehm T, et al. Progestogens and membrane-initiated effects on the proliferation of human breast cancer cells. Climacteric. 2012; 15: 467–72. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3109/13697137.2011.648232&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22335423&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000308942500010&link_type=ISI) 117.117. Willibald M, Bayer G, Stahlhut V, Poschmann G, Stühler K, Gierke B, et al. Progesterone receptor membrane component 1 is phosphorylated upon progestin treatment in breast cancer cells. Oncotarget. 2017; 8: 72480–93. 118.118. Guerra B, Issinger OG. Protein kinase CK2 in human diseases. Curr Med Chem. 2008; 15: 1870–86. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.2174/092986708785132933&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18691045&link_type=MED&atom=%2Fcbm%2F19%2F4%2F432.atom) 119.119. Ortega CE, Seidner Y, Dominguez I. Mining CK2 in cancer. PLoS One. 2014; 9: e115609. 120.120. Neubauer H, Ruan X, Schneck H, Seeger H, Cahill MA, Liang Y, et al. Overexpression of progesterone receptor membrane component 1: possible mechanism for increased breast cancer risk with norethisterone in hormone therapy. Menopause. 2013; 20: 504–10. 121.121. Zhao Y, Ruan X, Wang H, Li X, Gu M, Wang L, et al. The presence of a membrane-bound progesterone receptor induces growth of breast cancer with norethisterone but not with progesterone: a xenograft model. Maturitas. 2017; 102: 26–33.