Fibroid growth and medical options for treatment Nathalie Chabbert-Buffet, M.D., Ph.D.,a Nathalie Esber, M.Sc.,b,c and Philippe Bouchard, M.D.d,e a Obstetrics, Gynecology and Reproductive Medicine Department, AP-HP, Hospital Tenon, Paris; b INSERM, U693, Le  de Me  decine Paris-Sud, Le Kremlin-Bice ^tre; d Univ Paris-Sud, UMR-S693, Le Kremlin-Bice ^ tre; ^tre; c Faculte Kremlin-Bice and e UPMC Paris 06, Paris, France

Although fibroids are common benign tumors, their impact on women's quality of life can be considerable. The most frequent symptoms are uterine bleeding, resulting in anemia, and pelvic pain. Fibroids can be of genetic or hormonal origin or arise from intrauterine events. Current options for medical treatment include control of estradiol and progesterone production or action and are discussed in this review. Although curative treatment of fibroids relies on surgical strategies, the current trend is for uterine-sparing treatment to preserve fertility and avoid unnecessary surgery. Currently approved medical treatments include intrauterine progestin delivery to reduce uterine bleeding, GnRH analogues, and, more recently, selective progesterone receptor modulators to control uterine bleeding and reduce fibroid volume. (Fertil SterilÒ 2014;102: Use your smartphone 630–9. Ó2014 by American Society for Reproductive Medicine.) to scan this QR code Key Words: Fibroids, sex steroids, fibroids treatment, progestins, selective progesterone and connect to the receptor modulators, selective estrogen receptor modulators Discuss: You can discuss this article with its authors and other ASRM members at http:// fertstertforum.com/chabbertbuffetn-fibroids-growth-medical-treatment/

F

ibroids are the most common gynecologic tumors, occurring in 70% of women over 30 years of age (1). They are benign tumors developing in the myometrium. Despite the high incidence of the disease and its impact on women's quality of life, there has been relatively little research on fibroids until recently (2). Fibroids develop from a single myometrial smooth muscle cell and are therefore classified as a clonal disease (3, 4). Risk factors for developing fibroids, apart from ethnic origin and heredity, include situations causing prolonged high exposure to estrogens and/or progesterone, such as early age of menarche (5, 6), polycystic ovary syndrome (7), obesity (8), and late pregnancy (9). Consistent with these epidemiologic data, the best known stimulator of tumor

growth is the combined action of estrogens and progesterone. Growth factors, cytokines, and chemokines (1) have also been described as playing a role. Several animal and in vitro models (5) developed over the past decade have contributed to a better understanding of the disease. Fibroids are mostly asymptomatic (80%) but can induce symptoms with a high impact on women's health (10) depending on their size and location. Until recently, they were largely classified as being either subserosal or interstitial. However, the International Federation of Gynecology and Obstetrics now recognizse eight subtypes: types 0–2, submucosal; 3–5, interstitial; 5–7, subserosal; and 8, extrauterine (e.g., parametrium; Fig. 1) (11). Heavy menstrual bleeding is the most frequent fibroid-related symptom, re-

Received June 9, 2014; revised and accepted July 15, 2014. N.C.-B. is a member of the French Gedeon Richter's scientific advisory board. N.E. is funded by HRA Pharma for her Ph.D. P.B. is a member of Preglem's scientific advisory board. Reprint requests: Nathalie Chabbert-Buffet, M.D., Ph.D., Obstetrics Gynecology and Reproductive Medicine Department, Tenon Hospital, 4 rue de la Chine, 75 020 Paris, France (E-mail: [email protected]). Fertility and Sterility® Vol. 102, No. 3, September 2014 0015-0282/$36.00 Copyright ©2014 American Society for Reproductive Medicine, Published by Elsevier Inc. http://dx.doi.org/10.1016/j.fertnstert.2014.07.1238 630

discussion forum for this article now.*

* Download a free QR code scanner by searching for “QR scanner” in your smartphone’s app store or app marketplace.

sulting in reduced quality of life and anemia (5), and it can be related either to the location of the fibroid (submucosal) or to fibroid-related endometrial dysfunction (11). In addition, associated endometrial lesions may be present in up to 20% women with abnormal uterine bleeding, particularly adenomyosis (12–15). Pretherapeutic workup should, of course, aim to identify these associated conditions, because they may require specific care. Symptoms related to tumor volume are less frequent and include pelvic pain, dyspareunia, and urinary symptoms, such as pollakiuria and dysuria. Fibroid volume can also occasionally result in urinary tract compression and ureteral dilation, which can lead to renal dysfunction. The role of fibroids in infertility is limited and mostly related to submucosal lesions resulting in implantation defects (16). Whether large interstitial fibroids can cause infertility through deformation of the uterine cavity is still under discussion (16). However, although African women have a high incidence of fibroids and more severe lesions, their rate of fertility remains high even VOL. 102 NO. 3 / SEPTEMBER 2014

Fertility and Sterility®

FIGURE 1

The 2011 FIGO classification of fibroids (reproduced from Munro et al. [11] with permission). Chabbert-Buffet. Fibroids growth and treatment. Fertil Steril 2014.

though pregnancy usually occurs at an age where the rate of fibroids is already high: 25–35 years (17). Although bleeding may have a marked impact on their quality of life, women often delay seeking medical advice. This may be because they underestimate the importance of the bleeding (18) or because of a fear of surgical treatment. It is therefore important that women are better informed about medical treatment options. The only curative treatment to date is surgical removal either by myomectomy or hysterectomy. Although alternative surgical strategies have resulted in a reduction of the number of hysterectomies (19), 55,000 hysterectomies a year are still performed for fibroids in the United Kingdom and 600,000 in the United States (20), resulting in a heavy economic burden (21). Destruction techniques, by ultrasound or radiofrequency (22), are under evaluation as complementary or associated techniques. Endometrial ablation, either by surgical or physical means, such as thermablation (23), is an alternative option for women close to menopause who would like to avoid surgery (20). However, the current trend is for uterine-sparing therapeutic strategies, mainly because the age of a first pregnancy is on the increase. Because myomectomy can be detrimental on pregnancy outcome (24), medical options are frequently offered as a first-line option. Medical therapy is also required before surgery in women with fibroid-related anemia to reduce postoperative morbidity (25) or to facilitate endometrial ablation techniques (26). Most medical treatments reduce bleeding. Modulators of estrogen signaling, such as aromatase inhibitors, and selective estrogen receptor modulators (SERMs) have been evaluated. Progestins are also used for this purpose but have limited efficacy and can induce tumor growth as discussed below. Finally GnRH analogues and selective progesterone receptor modulators (SPRMs) can be used to reduce both tumor volume and bleeding (Table 1).

FIBROID GROWTH Although quite a lot is known about the factors contributing to fibroid growth, the pathophysiology of this disease remains largely unelucidated (1, 5). VOL. 102 NO. 3 / SEPTEMBER 2014

Histology Fibroids are benign tumors developing in the myometrium (Fig. 2) and appear as a disordered organization of the myometrial cells forming spheres and nesting in abundant extracellular matrix (ECM) (9, 27). This results in the classic surgical description of fibroids as firm tumors with a clear dissection plane and easy to enucleate. Other histologic presentations have been described, such as cellular fibroids with scarce ECM component or bizarre fibroids with unusual, but nonsuspect, tissue organization. Numerous other forms have also been described (9), including vascular fibroids, intravascular fibroids, and benign disseminating fibroids (leiomyomatosis) (28). These latter forms, though benign, can be life-threatening. Preoperative differential diagnosis with sarcoma is mostly based on the analysis of the tumor vascularization on Doppler-coupled ultrasound scan and magnetic resonance imaging (29). This differential diagnosis is important, especially in the case of a nonsurgical therapeutic option, even though sarcomas are extremely rare, occurring in 10 out of 1 million women every year (30). Stem cells may be involved in fibroid growth. Specific myometrial cellular subpopulations exhibiting myometrial stem cell characteristics have been identified (31) and found to be necessary for fibroid growth in an animal model (32). These cells appear to be pluripotent and may be tumor initiating (33). One single stem cell is thought to give birth to a specific fibroid (which is why it is called a clonal disease) (3, 4). Stem cells present in fibroids, compared with stem cells in normal myometrium, carry mutations of the MED12 (mediator complex subunit 12) gene located on the X chromosome (32). This suggests a primary role of genetic events in the stem cells to allow tumor growth. MED12 is one of the components involved in the control of transcription initiation in association with the preinitiation complex (34). Finally, the role of the ECM in fibroid growth, as part of the tumor microenvironment, is also thought to be very

FIGURE 2

Fibroid growth. CCR ¼ complex chromosomal rearrangement; ECM ¼ extracellular matrix; ER ¼ estrogen receptor; PR ¼ progesterone receptor. Chabbert-Buffet. Fibroids growth and treatment. Fertil Steril 2014.

631

VIEWS AND REVIEWS

TABLE 1 Fibroid treatment: hormone production and signalling modulators. Therapeutic class Progestins GnRH analogues SPRMs

Molecules Danazol DMPA LNG-IUS Leuprorelin acetate Mifepristone Ulipristal acetate Asoprisnil Telapristone acetate

Aromatase inhibitors SERMs

Raloxifene

Therapeutic effects in women with fibroids Bleeding control Bleeding control, tumor volume reduction Rapid bleeding control Tumor volume control Bleeding control Tumor volume control Tumor volume control; limited data Questionable efficacy

Main adverse events Fibroid growth Weight gain Expulsion Hot flushes, bone demineralization PAECs

Status

FDA approval FDA approval FDA approval

Reference(s) (87) (26, 149) (88) (90) (150) (10, 75, 121)

Phase II Hot flushes, joint pain

Phase II

(102)

Hot flushes, DVT

Phase II

(100)

Note: DMPA ¼ depot medroxyprogesterone acetate; DVT ¼ deep vein thrombosis; LNG-IUS ¼ levonorgestrel-delivering intrauterine system; PAECs ¼ progesterone receptor modulator–associated endometrial changes; SERMs ¼ selective estrogen receptor modulators; SPRMs ¼ selective progesterone receptor modulators. Chabbert-Buffet. Fibroids growth and treatment. Fertil Steril 2014.

important. As previously described, an increase in tumor volume can be related to cell proliferation (cellular fibroids, bizarre fibroids) or ECM accumulation (usual fibroids). ECM in fibroids is abnormal in structure and physical properties (35) compared with the normal myometrial ECM. Previous nonhuman animal models have shown discrepancies between human and nonhuman pathophysiology, because tumors in other animals do not include excess ECM (5). Important discrepancies have also been observed between cellular models of fibroids, without ECM, and in vivo experiments. The role of ECM has been confirmed in a recently developed model of human fibroid xenograft in immunodeficient mice (36). Tumor growth control by sex steroids in a paracrine system of regulation has also been described (37). Estradiol appears to be necessary to allow the progesterone stimulatory action on fibroid growth as discussed below. In addition, E2 and P action seems to be necessary for the production of ECM components, such as collagen types I and II. In fact collagen fibers are overexpressed in the follicular phase of the cycle (38) and the P receptor antagonist asoprisnil down-regulates collagen fiber synthesis in an in vitro model (39).

Genetics Apart from the very rare syndromic fibroids associated with aggressive renal cancer in individuals with fumarate hydratase (FH) gene mutations on chromosome 1 (40), the genetic component of fibroids remains speculative. Studies into somatic genetic alterations revealed that 50% of fibroids have chromosomal abnormalities. Genetic alterations have been described in 12 out of 26 chromosomes, and nine main candidate genes have been identified: MED12, HMGA2, HMGA1, FH, BHD, TSC2, PCOLCE, ORC5L, and LHFPL3 (1, 9). In addition to somatic gene alterations, epigenetic mechanisms also may be involved independently from specific modifications of the DNA sequence. Differences in DNA methylation, involved in gene transcription repression, have been described in fibroids compared with the normal surrounding myometrium (41). Most of these DNA methylation 632

modifications occur in the promoter region of the involved genes, including a number of tumor repressors, such as KLF11, a P receptor target gene. These modifications result in silencing the corresponding genes in fibroids, therefore promoting tumor growth. Other epigenetic modifications may include histone modifications, which modulate gene transcription, and microRNAs which modulate protein production downstream of gene transcription. Data on histone modifications are extremely limited to date (42). Data regarding microRNA (43, 44) show that several microRNAs are deregulated in fibroids, as well as their target genes. More recently, a whole-genome sequencing study of fibroids from 30 patients (45) has provided complementary information. It was shown that identical variants may be identified in different nodules—up to five different nodules from the same patient—suggesting a common cellular origin. Single mutations have been observed mostly in MED12 and FH genes. Modifications at chromosome 7q locus were frequent, particularly the COL4A5–COL4A6 locus, as well as modifications of the HMGA2 locus on chromosome 12 and RAD51B on chromosome 14. Complex chromosomal rearrangements (CCRs) in fibroids were also observed, sharing common features with chromothripsis, a single-genomic event that results in focal losses and rearrangements in multiple genomic regions. This leads to cell death and apoptosis, but is also involved in carcinogenesis. In fibroids, chromothripsis-like events appear to lead to benign tumor development. None of the samples studied carried TP53 mutations, confirming benignity. CCRs are most probably due to multiple chromosomal breaks and random reassembly, and they promote tumor growth.

Risk Factors for Fibroids The epidemiology of fibroids suggests two distinct patterns depending on ethnic background (46). In women of African origin, fibroids occur early in their reproductive life, they are more often numerous and larger in size, recurrence occurs more often, and they do not regress after menopause (47). Other VOL. 102 NO. 3 / SEPTEMBER 2014

Fertility and Sterility® risk factors identified in this population (7, 48–52) include age, early menarche, overweight, and polycystic ovary syndrome. Tumoral molecular biology studies suggest that E2 metabolism may be altered in women of African origin, because polymorphism in the catecholamine-O-methyl transferase (COMT) gene leading to reduced enzyme activity has been correlated with increased tumor size (53). However, this polymorphism does not appear to alter fibroid prevalence (54). The product of E2 transformation by COMT, 2-methoxyE2, has been shown to be involved in the regulation of aromatase activity and estrogen signaling in fibroids. In women of European origin (46), fibroids are most frequent in women in their 40s, suggesting an important role of the duration of exposure to sex steroids (55). Fibroids regress after menopause, suggesting that tissue aging alone is not a sufficient mechanism. Obesity is a major risk factor, and adiponectin (56, 57), a cytokine secreted by the adipose tissue, may be involved as well. Endocrine disruptors may also play a role. Environmental exposure to phthalates (58) is associated with fibroid development, as well as with endometriosis. Intrauterine exposure to diethylstilbestrol (59) also has been related to an increased risk of developing fibroids. The number and size of fibroids vary considerably from one woman to another.

Role of Sex Steroids Data from in vitro and nonhuman animal models over the years suggest that E2 plays a central role in fibroid growth via its receptor, ERa. Fibroid cells cultured in vitro and treated with estrogens proliferate. In mouse (60) and guinea pig (61) models, exogenous E2 has been shown to promote tumor growth (37). Estradiol from the general circulation as well as locally produced (62) is involved. This is supported mostly by clinical data showing that aromatase inhibitors are as effective as GnRH analogues in reducing fibroid volume in premenopausal women, with circulating E2 levels remaining normal to high in aromatase inhibitor users and low in GnRH analogue users (63). Estradiol therefore acts via its a receptor (64). However, while high levels of ERa/ERb mRNA have been described in fibroids compared with normal adjacent myometrium (65) subsequent analysis of the corresponding protein expression failed to confirm that overexpression or unbalanced expression of ER isoforms plays a potential specific role (66). Initial data on the action of P were more controversial (37). Depending on the experimental conditions, progestins were found to stimulate or inhibit cell proliferation in in vitro models. In a mouse model, P was able to inhibit estrogeninducing tumor growth (36). In the Eker rat model (67) P was unable to stimulate tumor growth whereas estrogens could. In parallel, clinical data suggested a major role of P in fibroid growth. Proliferation markers are overexpressed in fibroids during the luteal phase (68), as well as in postmenopausal women treated with estrogens and progestins, but not with estrogens only (69). In postmenopausal women, the stimulatory effect of hormone replacement therapy on fibroid growth has been correlated with the progestin dose (70). Whereas treatment with GnRH analogues results in fibroid shrinkage, VOL. 102 NO. 3 / SEPTEMBER 2014

cotreatment with progestins limited their therapeutic efficacy (71). This was not observed in women cotreated with estrogens and progestins (72). Finally, recent clinical studies have shown that four different SPRMs cause fibroids to shrink (73, 74) in the efficacy range of GnRH analogues (75). This effect was observed despite maintained E2 levels (75) and brings further evidence of the role of the P receptor (PR) and its ligand in fibroid growth. In fibroids, P acts through its nuclear receptor as well as other nonnuclear signaling systems. The nuclear PR exists as two main isoforms (PRA and PRB) arising from the same gene (76). Interestingly, the ratio of the two isoforms appears to be balanced in fibroids as well as in the adjacent myometrium (77). This is in contrast with data from other proliferating benign or malignant diseases, such as endometriosis or breast cancer, where an unbalanced PRA/ PRB ratio is involved in tumor growth (78, 79). Progesterone may also signal through membrane proteins (80), which have not been evaluated in fibroids. Recently a truncated form of the nuclear PR, deleted of its DNA-binding region and localized to the mitochondria (PR-M), has been cloned. An increase in PR-M expression has been observed in fibroids and may be involved in fibroid growth through altered mitochondrial activity and cellular respiration (81). Biologic actions of the ligand-PR complex have been evaluated in genome-wide PR binding experiments (1). Ligand-receptor interaction induces PR binding to a very high number of distinct sites over the genome and activation of multiple gene transcription. Further research is needed to correlate these complex events with the role of genetic events discussed above or with therapeutic effects. Finally, in the fibroid xenograft mouse model (36) in which both fibroids and the microenvironment can be evaluated, it has been shown that P and E2 together, but not administered alone, cause cell proliferation and ECM accumulation. This synergistic effect was reverted by the SPRM mifepristone. Taken together, these data suggest that both E2 and P, and their specific receptors ERa, PRA, and PRB, are involved in fibroid growth. Both steroids appear to act in a collaborative system where E2 allows the expression of PRs which in turn control cell proliferation and ECM production. However, although fibroid growth appears to depend on sex steroids, ERs and PRs are not expressed in all tumor cells, suggesting paracrine mechanisms. In particular, stem cells express extremely low levels of sex steroid receptors (32). In vitro data suggest that coculture with fibroid cells allow stem cells to grow into large tumors when grafted to immunodeficient mice (32). Other components of the tumor microenvironment, including adipocytes, may exert paracrine regulation through growth factors, cytokines, and chemokines (56, 82–86).

FIBROID TREATMENT For years the only curative treatment for fibroids was considered to be surgery, but alternate options are available today. Although hysterectomy remains the first treatment option for fibroids, women are increasingly choosing to avoid hysterectomy either because they are approaching menopause or because they want to preserve pregnancy opportunities. 633

VIEWS AND REVIEWS Other options include: limited surgery, i.e., myomectomy either by hysteroscopy, laparoscopy, or laparotomy for some cases (depending on the number, size and type of tumor); embolization of fibroids; and physical destruction by ultrasound and radiofrequency, which is currently under evaluation. Medical treatment is initially used to relieve heavy menstrual bleeding. It improves a woman's quality of life, relieves anemia-related symptoms, and reduces subsequent surgical morbidity. In addition, medical treatment reducing the uterine volume can facilitate surgical management. It is important to note that all medical treatment options to date induce anovulation or are contraindicated in pregnant women and, therefore, in women wishing to conceive.

Progestins Oral or intramuscular progestins may be used to treat dysfunctional uterine bleeding associated with fibroids by reducing endometrial hyperplasia (Table 1). However, data from the literature are limited (87, 88) and tumor volume reduction has not been confirmed (89). This is probably the consequence of the multiple effects of progestins. Some of these are beneficial, such as endometrial atrophy or their effect in suppressing gonadotropin secretion. However, as already discussed, progestins such as P are growth factors for fibroids. Endometrial thinning before hysteroscopic surgery can be obtained with the use of danazol (26, 90, 91), although GnRH analogues have been shown to be more effective. Intrauterine delivery of progestins by means of an intrauterine system (IUS) is also an option (92). The levonorgestrelreleasing IUS (LNG-IUS) has been approved by the United States Food and Drug Administration to treat heavy menstrual bleeding in IUS users. In women with fibroid-related uterine bleeding, the LNG-IUS has been shown to reduce bleeding and improve anemia (88, 93). However, tumor volume was not reduced. In addition, insertion of an IUS can be difficult in women with submucosal fibroids, and the device can be expelled more frequently (94). However, the use of LNG-IUS may reduce the rate of hysterectomy and improve patient satisfaction (93).

Selective Estrogen Receptor Modulators SERMs are nonsteroidal estrogen receptor ligands with agonist or antagonist effects depending on the tissue. In contrast to tamoxifen, the widely used SERM in breast cancer, raloxifene exerts antiestrogenic effects in fibroids. Raloxifene reduces cell proliferation (95–98) and has no endometrial agonist activity (99). Three randomized controlled trials have evaluated raloxifene in premenopausal women with confirmed fibroids (Table 1) (100). Two of these trials, including 215 women, showed therapeutic efficacy of raloxifene, but the third did not. This may be due to the rise in E2 secretion observed in premenopausal women following SERM treatment. Clinical efficacy of raloxifene is therefore considered to be limited. Furthermore, tamoxifen does not appear to be suitable, owing to its agonistic effect in the endometrium (101). 634

Aromatase Inhibitors Aromatase is a cytochrome P450 enzyme (CYP-19) that allows the transformation of androgens into estrogens. Aromatase inhibitors are steroidal (exemestane) or nonsteroidal (anastrozole, letrozole) compounds that interact with the hormone-binding site of the molecule (exemestane) or with its catalytic subunit (anastrozole, letrozole). In premenopausal women, their use results in a rise in plasma E2 levels, so they are unlikely to be suitable to treat fibroids. However, they may contribute to a local decrease in E2 production within fibroids, as has been previously discussed (63). A single clinical trial has been included in a recent Cochrane data base review (Table 1) (102). This trial compared the aromatase inhibitor letrozole to the GnRH analogue triptorelin. It showed that letrozole reduces fibroid volumes by 46% (vs. 32% in the GnRH analogue group). However, no significant data are available regarding their impact on uterine bleeding, even though pilot studies have suggested that aromatase inhibitors may help to control uterine bleeding (103). To date, clinical data are too limited to allow aromatase inhibitors to be used for fibroid treatment in clinical practice.

GnRH Analogues GnRH analogues are synthetic peptidic compounds structurally close to the natural GnRH molecule. After binding to the GnRH receptors, agonistic analogues induce subsequent stimulation of gonadotropin secretion followed by desensitization, and thus they delay the gonadotropic axis blockade. In contrast, GnRH antagonists induce immediate blockade of GnRH receptors and immediate decrease in LH and FSH secretion. As a consequence, both E2 and P levels reach postmenopausal levels. This property has been used to treat fibroids, in part because of the ability of GnRH analogues to induce amenorrhea. In line with the already discussed pathophysiology, additional biologic effects related to low E2 and P levels have been described. In addition to their effects on circulating steroid levels, GnRH analogues have been shown to decrease different paracrine mitogenic and angiogenic factor expression, such as vascular endothelial growth factor, fibroblastic growth factor, and plateletderived growth factor (104, 105). In vitro GnRH analogues inhibit cell proliferation and induce apoptosis (106). Studies of surgical samples of fibroids have contributed to understanding some of the pathways involved in apoptosis activation, such as down-regulation of the PI3 kinase protein kinase B pathway (107). Clinical randomized studies have shown that GnRH analogues can control bleeding, thus correcting anemia, and reduce fibroid volume by 50%. Surgical procedures are subsequently facilitated (Table 1) (90). Treatments with GnRH analogues have been evaluated, either alone or with an add-back therapy to limit their side effects, particularly the consequences of low estrogen secretion, such as hot flushes and a decrease in bone density. Add-back treatments include progestins, estrogen and progestin combinations, and raloxifene (91). However, the use of progestins as an add-back therapy appears to reduce the effect of GnRH analogues on fibroid volume. Alternatively, raloxifene treatment may VOL. 102 NO. 3 / SEPTEMBER 2014

Fertility and Sterility® help further reduce fibroid volume but is unable to decrease the intensity and frequency of hot flushes. Finally, the use of GnRH analogues results in fibroid shrinkage, but for a limited period corresponding to the duration of treatment, and E2 suppression (75). Fibroid volume increases after cessation of treatment, and mean changes from screening at week 38 (25 weeks, or 6 months, after treatment discontinuation) were 16.5% in the leuprorelin-treated group compared with 44.8% and 54.8% in the 5 mg/d and 10 mg/d ulipristal acetate (UPA)–treated groups, respectively, in the PEARL II study (75).

Selective Progesterone Receptor Modulators SPRMs are PR ligands with agonist, antagonist, or mixed activity depending on the cellular and molecular context (108). In vitro, cultured fibroid cells treated with the SPRM UPA, telapristone acetate, or asoprisnil showed a decrease in cell proliferation as well as an induction of apoptosis pathways (109–114). In contrast, these agents have no effect on the proliferation or apoptosis of normal myometrial cells. Asoprisnil and UPA also down-regulate a number of growth factors and their receptors in vitro (115, 116). Finally, asoprisnil and UPA can reduce ECM production in cultured fibroid cells by up-regulating ECM metalloproteinase activity in vitro (39, 110). Recently, looking for a molecular explanation, and potentially for a predictive marker of SPRM efficacy, Engman et al. suggested that the enzyme glutathione-S transferase mu 1 overexpression is associated with a significant response to treatment (117). A number of clinical trials have investigated the efficacy and safety of SPRMs as a treatment for fibroids, showing that mifepristone (118–120), UPA (74, 75, 118–121), asoprisnil (122, 123), and telapristone acetate (73) are all effective in reducing fibroid and uterine volume (73). Fibroid volumes are reduced by 17%–57% and uterine volume by 9%–53% following treatment with SPRMs compared with placebo (74). In contrast to GnRH agonist therapy, the SPRMs control tumor volume (75) for a prolonged period of up to 6 months after treatment discontinuation (75, 124). SPRMs are also able to suppress bleeding more rapidly than GnRH analogues in women with fibroids. In a randomized trial, bleeding control occurred within 7 days in a UPA group compared with 21 days in a leuprorelin group (74). Most women treated with UPA, asoprisnil, or mifepristone experience amenorrhea during the entire treatment (75, 123, 125). Importantly, bleeding control after treatment with an SPRM is not associated with signs of E2 deprivation, and E2 plasma levels are maintained at 60 pg/mL. In contrast, GnRH analogues induce postmenopausal levels of E2 and cause subsequent hot flushes and bone loss (75). All studies with SPRMs have reported an improvement in quality of life scales in subjects treated with UPA, asoprisnil, or mifepristone (126–130). UPA administered for 3 months at a daily dose of 5 mg is currently used in clinical practice to treat fibroids in the preoperative period in women eligible for surgical treatment. The recently published PEARL III study and its extension (121) resulted in the approval of the extension of treatment to VOL. 102 NO. 3 / SEPTEMBER 2014

a second course of 3 months in Europe. In that study, women received an initial 3-month course of 10 mg/d UPA. They could then be enrolled in the extension phase of the study where they were randomized to receive norethisterone acetate (NETA) or placebo for 10 days. A subsequent 3-month treatment with UPA was then started on the 4th day of bleeding, followed by a 10-days NETA/placebo period, up to a final of four courses of UPA. The study showed that amenorrhea was obtained after a mean of 3.5 days in the first UPA treatment period, and 2–3 days in subsequent courses. Ninety percent of women were amenorheic after the first course and 93%–94% of them had spotting or no bleeding at all in the subsequent courses. Fibroid volume reduction reached 45% after the first course and continued to decrease in subsequent treatment rounds to 72% after the fourth treatment course. We are currently awaiting further data from the Pearl IV study exploring long-term administration of UPA at 5 mg/d versus 10 mg/d (in 500 women) in Europe and from the Premya phase IV study also conducted in Europe.

Endometrium Changes There has been some concern regarding endometrial changes induced by medium- to long-term (3–6 months) continuous daily dosing of mifepristone (2–200 mg/d) (131) and asoprisnil (132, 133). Nonphysiologic endometrial changes characterized by dilated weakly secretory endometrial glands with few mitotic figures, and stromal effects ranging from compaction to nonuniform edema have been described. A panel of expert pathologists concluded that no safety concerns arise from these changes, which have been termed ‘‘progesterone receptor modulator–associated endometrial changes’’ (PAECs) (134, 135). Additional data from the PEARL I, II, and III studies (74, 75, 121) confirmed that PAECs were not a matter of concern. Endometrial thickening >16 mm occurred in 10%–12% of women, and endometrial histologic samples revealed no atypia, either simple or complex, in the treated groups (136). PAECs always disappear after treatment cessation (74, 75, 121). Administration of a progestin (121) after UPA discontinuation does not speed up regression of PAECs. However, data on PAECs require further evaluation, because this aspect has also been observed in women not receiving SPRMs. Screening of endometrial samples showed that 10% of women with fibroids had PAECs before administration of SPRMs (121).

Other Potential Therapeutic Options A number of potential treatments for fibroids are currently in preclinical or phase II studies (9). Additional studies are needed in women suffering from fibroids, particularly in women planning to conceive. Herbal therapy is being investigated in phase II clinical trials. For example, epigallocatechin gallate, found in green tea, has been shown to inhibit fibroid cell proliferation and induce apoptosis in vitro and in nonhuman animal models (137, 138). Inhibitors of fibrosis formation, such as pirfenidone (139), also have been evaluated. 635

VIEWS AND REVIEWS Correction of vitamin D deficiency, a common condition in northern countries, may be an important issue as well. Decreased vitamin D level has been described as a risk factor for developing fibroids (relative risk 2.4) (140), and sufficient vitamin D levels (>20 mg/mL) are associated with a decreased risk of developing fibroids independently from ethnic origin (odds ratio 0.68, 95% confidence interval 0.48–0.96) (141). In vitro, vitamin D reduces ECM production (142–144) and cellular proliferation in fibroid cells as well as in normal myometrial cells (145). In the Eker rat model, vitamin D causes fibroid shrinkage (146). In addition, the impact on bone mineral density of GnRH analogues used for fibroid treatment may be limited, or even reverted, by vitamin D supplementation (147, 148). Finally, molecules that control fibroid cell proliferation in vitro or in vivo may deserve clinical evaluation in the future. Growth factor receptor antagonists, such as AG1478 and TKS050, which block EGF signalling, or SB525334, which is a transforming growth factor beta signaling blocker, have been shown to control fibroid cell proliferation in vitro (9). Peroxisome proliferator–activated receptor gamma receptor ligands, such as roziglitazone and ciglitizone, also have been evaluated in vitro and shown to reduce fibroid cell proliferation (9).

CONCLUSION SPRMs appear to be the best therapy for women with symptomatic fibroids who prefer not to undergo surgery, especially when administered after a long-term discontinuous pattern (121). They may facilitate surgical procedures pending the review analysis of worldwide surgical experience. Apart from providing an option for women who wish to preserve childbearing potential, they may also be advantageous for women in their late 40s to delay invasive procedures until menopause, when the fibroids could regress naturally. This new therapeutic option appears to be safe and contributes to a rapid improvement of quality of life in women suffering from fibroids. We think that in the future, SPRMs may be combined with other more recent therapeutic options, such as growth factor modulators.

8. 9.

10.

11.

12.

13. 14. 15.

16. 17. 18. 19.

20. 21.

22.

23.

24.

25.

REFERENCES 1. 2. 3.

4. 5.

6.

7.

636

Bulun SE. Uterine fibroids. N Engl J Med 2013;369:1344–55. Walker CL, Stewart EA. Uterine fibroids: the elephant in the room. Science 2005;308:1589–92. Holdsworth-Carson SJ, Zaitseva M, Vollenhoven BJ, Rogers PA. Clonality of smooth muscle and fibroblast cell populations isolated from human fibroid and myometrial tissues. Mol Hum Reprod 2014;20:250–9. Linder D, Gartler SM. Glucose-6-phosphate dehydrogenase mosaicism: utilization as a cell marker in the study of leiomyomas. Science 1965;150:67–9. Segars JH, Parrott EC, Nagel JD, Guo XC, Gao X, Birnbaum LS, et al. Proceedings from the Third National Institutes of Health International Congress on Advances in Uterine Leiomyoma Research: comprehensive review, conference summary and future recommendations. Hum Reprod Update 2014;20:309–33. Velez Edwards DR, Baird DD, Hartmann KE. Association of age at menarche with increasing number of fibroids in a cohort of women who underwent standardized ultrasound assessment. Am J Epidemiol 2013;178:426–33. Wise LA, Palmer JR, Stewart EA, Rosenberg L. Polycystic ovary syndrome and risk of uterine leiomyomata. Fertil Steril 2007;87:1108–15.

26.

27.

28.

29.

30. 31.

Okolo S. Incidence, aetiology and epidemiology of uterine fibroids. Best Pract Res Clin Obstet Gynaecol 2008;22:571–88. Islam MS, Protic O, Giannubilo SR, Toti P, Tranquilli AL, Petraglia F, et al. Uterine leiomyoma: available medical treatments and new possible therapeutic options. J Clin Endocrinol Metab 2013;98:921–34. Downes E, Sikirica V, Gilabert-Estelles J, Bolge SC, Dodd SL, Maroulis C, et al. The burden of uterine fibroids in five European countries. Eur J Obstet Gynecol Reprod Biol 2010;152:96–102. Munro MG, Critchley HO, Fraser IS. The FIGO classification of causes of abnormal uterine bleeding in the reproductive years. Fertil Steril 2011; 95:2204–8. 8.e1–3. Brucker SY, Huebner M, Wallwiener M, Stewart EA, Ebersoll S, Schoenfisch B, et al. Clinical characteristics indicating adenomyosis coexisting with leiomyomas: a retrospective, questionnaire-based study. Fertil Steril 2013;101:237–41.e1. Hanafi M. Ultrasound diagnosis of adenomyosis, leiomyoma, or combined with histopathological correlation. J Hum Reprod Sci 2013;6:189–93. Naftalin J, Hoo W, Pateman K, Mavrelos D, Foo X, Jurkovic D. Is adenomyosis associated with menorrhagia? Hum Reprod 2014;29:473–9. Nomelini RS, Ferreira FA, Borges RC, Adad SJ, Murta EF. Frequency of endometriosis and adenomyosis in patients with leiomyomas, gynecologic premalignant, and malignant neoplasias. Clin Exp Obstet Gynecol 2013;40:40–4. Pritts EA, Parker WH, Olive DL. Fibroids and infertility: an updated systematic review of the evidence. Fertil Steril 2009;91:1215–23. National Institute of Demography of Burkina Faso. Evolution of fertility rates by age category (Table 3). In: Population, 2014. Reid PC, Coker A, Coltart R. Assessment of menstrual blood loss using a pictorial chart: a validation study. BJOG 2000;107:320–2. Reid PC, Mukri F. Trends in number of hysterectomies performed in England for menorrhagia: examination of health episode statistics, 1989 to 2002–3. BMJ 2005;330:938–9. Khan AT, Shehmar M, Gupta JK. Uterine fibroids: current perspectives. Int J Womens Health 2014;6:95–114. Cardozo ER, Clark AD, Banks NK, Henne MB, Stegmann BJ, Segars JH. The estimated annual cost of uterine leiomyomata in the United States. Am J Obstet Gynecol 2012;206:211.e1–9. Trumm CG, Stahl R, Clevert DA, Herzog P, Mindjuk I, Kornprobst S, et al. Magnetic resonance imaging-guided focused ultrasound treatment of symptomatic uterine fibroids: impact of technology advancement on ablation volumes in 115 patients. Invest Radiol 2013;48:359–65. Hachmann-Nielsen E, Rudnicki M. Clinical outcome after hydrothermal ablation treatment of menorrhagia in patients with and without submucous myomas. J Minim Invasive Gynecol 2012;19:212–6. Kim MS, Uhm YK, Kim JY, Jee BC, Kim YB. Obstetric outcomes after uterine myomectomy: laparoscopic versus laparotomic approach. Obstet Gynecol Sci 2014;56:375–81. Musallam KM, Jamali FR, Rosendaal FR, Richards T, Spahn DR, Khavandi K, et al. Preoperative hematocrit concentration and the risk of stroke in patients undergoing isolated coronary-artery bypass grafting. Anemia 2013;2013:206829. Tan YH, Lethaby A. Pre-operative endometrial thinning agents before endometrial destruction for heavy menstrual bleeding. Cochrane Database Syst Rev 2013:CD010241. Malik M, Norian J, McCarthy-Keith D, Britten J, Catherino WH. Why leiomyomas are called fibroids: the central role of extracellular matrix in symptomatic women. Semin Reprod Med 2010;28:169–79. Awonuga AO, Shavell VI, Imudia AN, Rotas M, Diamond MP, Puscheck EE. Pathogenesis of benign metastasizing leiomyoma: a review. Obstet Gynecol Surv 2010;65:189–95. Yoshida Y, Kiyono Y, Tsujikawa T, Kurokawa T, Okazawa H, Kotsuji F. Additional value of 16alpha-[18F]fluoro-17beta-oestradiol PET for differential diagnosis between uterine sarcoma and leiomyoma in patients with positive or equivocal findings on [18F]fluorodeoxyglucose PET. Eur J Nucl Med Mol Imaging 2011;38:1824–31. Cancer Research UK. Soft tissue sarcoma incidence statistics. In, 2013. Ono M, Maruyama T, Masuda H, Kajitani T, Nagashima T, Arase T, et al. Side population in human uterine myometrium displays phenotypic and

VOL. 102 NO. 3 / SEPTEMBER 2014

Fertility and Sterility®

32. 33.

34.

35.

36.

37. 38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

functional characteristics of myometrial stem cells. Proc Natl Acad Sci U S A 2007;104:18700–5. Ono M, Qiang W, Serna VA, Yin P, Coon JS, Navarro A, et al. Role of stem cells in human uterine leiomyoma growth. PLoS One 2012;7:e36935. Mas A, Cervello I, Gil-Sanchis C, Faus A, Ferro J, Pellicer A, et al. Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertil Steril 2012;98:741–51.e6. Makinen N, Heinonen HR, Sjoberg J, Taipale J, Vahteristo P, Aaltonen LA. Mutation analysis of components of the mediator kinase module in MED12 mutation–negative uterine leiomyomas. Br J Cancer 2014;110:2246–9. Rogers R, Norian J, Malik M, Christman G, Abu-Asab M, Chen F, et al. Mechanical homeostasis is altered in uterine leiomyoma. Am J Obstet Gynecol 2008;198:474.e1–11. Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology 2010;151:2433–42. Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev 2013;34:130–62. Stewart EA, Friedman AJ, Peck K, Nowak RA. Relative overexpression of collagen type I and collagen type III messenger ribonucleic acids by uterine leiomyomas during the proliferative phase of the menstrual cycle. J Clin Endocrinol Metab 1994;79:900–6. Morikawa A, Ohara N, Xu Q, Nakabayashi K, DeManno DA, Chwalisz K, et al. Selective progesterone receptor modulator asoprisnil downregulates collagen synthesis in cultured human uterine leiomyoma cells through up-regulating extracellular matrix metalloproteinase inducer. Hum Reprod 2008;23:944–51. Alam NA, Olpin S, Leigh IM. Fumarate hydratase mutations and predisposition to cutaneous leiomyomas, uterine leiomyomas and renal cancer. Br J Dermatol 2005;153:11–7. Navarro A, Yin P, Monsivais D, Lin SM, Du P, Wei JJ, et al. Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS One 2012;7:e33284. Greathouse KL, Bredfeldt T, Everitt JI, Lin K, Berry T, Kannan K, et al. Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis. Mol Cancer Res 2012;10: 546–57. Marsh EE, Lin Z, Yin P, Milad M, Chakravarti D, Bulun SE. Differential expression of microRNA species in human uterine leiomyoma versus normal myometrium. Fertil Steril 2008;89:1771–6. Georgieva B, Milev I, Minkov I, Dimitrova I, Bradford AP, Baev V. Characterization of the uterine leiomyoma microRNAome by deep sequencing. Genomics 2012;99:275–81. Mehine M, Kaasinen E, Makinen N, Katainen R, Kampjarvi K, Pitkanen E, et al. Characterization of uterine leiomyomas by whole-genome sequencing. N Engl J Med 2013;369:43–53. Sweet S, Legro RS, Coney P. A comparison of methods and results in recruiting white and black women into reproductive studies: the MMCPSU Cooperative Center on Reproduction Experience. Contemp Clin Trials 2008;29:478–81. Peddada SD, Laughlin SK, Miner K, Guyon JP, Haneke K, Vahdat HL, et al. Growth of uterine leiomyomata among premenopausal black and white women. Proc Natl Acad Sci U S A 2008;105:19887–92. Radin RG, Rosenberg L, Palmer JR, Cozier YC, Kumanyika SK, Wise LA. Hypertension and risk of uterine leiomyomata in US black women. Hum Reprod 2012;27:1504–9. Wise LA, Palmer JR, Harlow BL, Spiegelman D, Stewart EA, AdamsCampbell LL, et al. Reproductive factors, hormonal contraception, and risk of uterine leiomyomata in African-American women: a prospective study. Am J Epidemiol 2004;159:113–23. Wise LA, Palmer JR, Spiegelman D, Harlow BL, Stewart EA, AdamsCampbell LL, et al. Influence of body size and body fat distribution on risk of uterine leiomyomata in U.S. black women. Epidemiology 2005; 16:346–54. Wise LA, Palmer JR, Stewart EA, Rosenberg L. Age-specific incidence rates for self-reported uterine leiomyomata in the Black Women's Health Study. Obstet Gynecol 2005;105:563–8.

VOL. 102 NO. 3 / SEPTEMBER 2014

52.

53.

54.

55.

56.

57.

58.

59. 60.

61. 62.

63.

64. 65.

66.

67. 68. 69.

70.

71.

72.

73.

74.

Wise LA, Radin RG, Palmer JR, Rosenberg L. Association of intrauterine and early life factors with uterine leiomyomata in black women. Ann Epidemiol 2012;22:847–54. de Oliveira E, de Aquino Castro R, Gomes MT, da Silva ID, Baracat EC, de Lima GR, et al. The catechol-O-methyltransferase (COMT) gene polymorphism and prevalence of uterine fibroids. Maturitas 2008;60:235–8. Gooden KM, Schroeder JC, North KE, Gammon MD, Hartmann KE, Taylor J, et al. Val153Met polymorphism of catechol-O-methyltransferase and prevalence of uterine leiomyomata. Reprod Sci 2007;14:117–20. Catherino WH, Eltoukhi HM, Al-Hendy A. Racial and ethnic differences in the pathogenesis and clinical manifestations of uterine leiomyoma. Semin Reprod Med 2013;31:370–9. Nair S, Al-Hendy A. Adipocytes enhance the proliferation of human leiomyoma cells via TNF-alpha proinflammatory cytokine. Reprod Sci 2011; 18:1186–92. Wakabayashi A, Takeda T, Tsuiji K, Li B, Sakata M, Morishige K, et al. Antiproliferative effect of adiponectin on rat uterine leiomyoma ELT-3 cells. Gynecol Endocrinol 2010;27:33–8. Weuve J, Hauser R, Calafat AM, Missmer SA, Wise LA. Association of exposure to phthalates with endometriosis and uterine leiomyomata: findings from NHANES, 1999–2004. Environ Health Perspect 2010;118:825–32. Baird DD, Newbold R. Prenatal diethylstilbestrol (DES) exposure is associated with uterine leiomyoma development. Reprod Toxicol 2005;20:81–4. Newbold RR, Moore AB, Dixon D. Characterization of uterine leiomyomas in CD-1 mice following developmental exposure to diethylstilbestrol (DES). Toxicol Pathol 2002;30:611–6. McDonough PG. The guinea pig model for uterine leiomyomata: genehormone interaction? Fertil Steril 2004;82:989. Bulun SE, Simpson ER, Word RA. Expression of the CYP19 gene and its product aromatase cytochrome P450 in human uterine leiomyoma tissues and cells in culture. J Clin Endocrinol Metab 1994;78:736–43. Parsanezhad ME, Azmoon M, Alborzi S, Rajaeefard A, Zarei A, Kazerooni T, et al. A randomized, controlled clinical trial comparing the effects of aromatase inhibitor (letrozole) and gonadotropin-releasing hormone agonist (triptorelin) on uterine leiomyoma volume and hormonal status. Fertil Steril 2010;93:192–8. Marsh EE, Bulun SE. Steroid hormones and leiomyomas. Obstet Gynecol Clin North Am 2006;33:59–67. Bakas P, Liapis A, Vlahopoulos S, Giner M, Logotheti S, Creatsas G, et al. Estrogen receptor alpha and beta in uterine fibroids: a basis for altered estrogen responsiveness. Fertil Steril 2008;90:1878–85. Grings AO, Lora V, Ferreira GD, Brum IS, Corleta H, Capp E. Protein expression of estrogen receptors alpha and beta and aromatase in myometrium and uterine leiomyoma. Gynecol Obstet Invest 2012;73:113–7. Cook JD, Walker CL. The Eker rat: establishing a genetic paradigm linking renal cell carcinoma and uterine leiomyoma. Curr Mol Med 2004;4:813–24. Tiltman AJ. The effect of progestins on the mitotic activity of uterine fibromyomas. Int J Gynecol Pathol 1985;4:89–96. Lamminen S, Rantala I, Helin H, Rorarius M, Tuimala R. Proliferative activity of human uterine leiomyoma cells as measured by automatic image analysis. Gynecol Obstet Invest 1992;34:111–4. Palomba S, Sena T, Morelli M, Noia R, Zullo F, Mastrantonio P. Effect of different doses of progestin on uterine leiomyomas in postmenopausal women. Eur J Obstet Gynecol Reprod Biol 2002;102:199–201. West CP, Lumsden MA, Hillier H, Sweeting V, Baird DT. Potential role for medroxyprogesterone acetate as an adjunct to goserelin (Zoladex) in the medical management of uterine fibroids. Hum Reprod 1992;7:328–32. Friedman AJ, Daly M, Juneau-Norcross M, Gleason R, Rein MS, LeBoff M. Long-term medical therapy for leiomyomata uteri: a prospective, randomized study of leuprolide acetate depot plus either oestrogen-progestin or progestin ‘‘add-back’’ for 2 years. Hum Reprod 1994;9:1618–25. Bouchard P, Chabbert-Buffet N, Fauser BC. Selective progesterone receptor modulators in reproductive medicine: pharmacology, clinical efficacy and safety. Fertil Steril 2011;96:1175–89. Donnez J, Tatarchuk TF, Bouchard P, Puscasiu L, Zakharenko NF, Ivanova T, et al. Ulipristal acetate versus placebo for fibroid treatment before surgery. N Engl J Med 2012;366:409–20.

637

VIEWS AND REVIEWS 75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87. 88.

89. 90.

91.

92.

93.

94.

95.

96.

638

Donnez J, Tomaszewski J, Vazquez F, Bouchard P, Lemieszczuk B, Baro F, et al. Ulipristal acetate versus leuprolide acetate for uterine fibroids. N Engl J Med 2012;366:421–32. Giangrande PH, McDonnell DP. The A and B isoforms of the human progesterone receptor: two functionally different transcription factors encoded by a single gene. Recent Prog Horm Res 1999;54:291–313. discussion 313–4. Lora V, Grings AO, Capp E, von Eye Corleta H, Brum IS. Gene and protein expression of progesterone receptor isoforms A and B, p53 and p21 in myometrium and uterine leiomyoma. Arch Gynecol Obstet 2012;286:119–24. Ariga N, Suzuki T, Moriya T, Kimura M, Inoue T, Ohuchi N, et al. Progesterone receptor A and B isoforms in the human breast and its disorders. Jpn J Cancer Res 2001;92:302–8. Arnett-Mansfield RL, DeFazio A, Mote PA, Clarke CL. Subnuclear distribution of progesterone receptors A and B in normal and malignant endometrium. J Clin Endocrinol Metab 2004;89:1429–42. Kowalik MK, Rekawiecki R, Kotwica J. The putative roles of nuclear and membrane-bound progesterone receptors in the female reproductive tract. Reprod Biol 2013;13:279–89. Feng Q, Crochet JR, Dai Q, Leppert PC, Price TM. Expression of a mitochondrial progesterone receptor (PR-M) in leiomyomata and association with increased mitochondrial membrane potential. J Clin Endocrinol Metab 2014;99:E390–9. Chegini N. Proinflammatory and profibrotic mediators: principal effectors of leiomyoma development as a fibrotic disorder. Semin Reprod Med 2010;28:180–203. Ciarmela P, Islam MS, Reis FM, Gray PC, Bloise E, Petraglia F, et al. Growth factors and myometrium: biological effects in uterine fibroid and possible clinical implications. Hum Reprod Update 2011;17:772–90. Ono M, Yin P, Navarro A, Moravek MB, Coon JS 5th, Druschitz SA, et al. Paracrine activation of WNT/beta-catenin pathway in uterine leiomyoma stem cells promotes tumor growth. Proc Natl Acad Sci U S A 2013;110: 17053–8. Sanci M, Dikis C, Inan S, Turkoz E, Dicle N, Ispahi C. Immunolocalization of VEGF, VEGF receptors, EGF-R and Ki-67 in leiomyoma, cellular leiomyoma and leiomyosarcoma. Acta Histochem 2010;113:317–25. Islam MS, Protic O, Stortoni P, Grechi G, Lamanna P, Petraglia F, et al. Complex networks of multiple factors in the pathogenesis of uterine leiomyoma. Fertil Steril 2013;100:178–93. Ke LQ, Yang K, Li J, Li CM. Danazol for uterine fibroids. Cochrane Database Syst Rev 2009:CD007692. Sangkomkamhang US, Lumbiganon P, Laopaiboon M, Mol BW. Progestogens or progestogen-releasing intrauterine systems for uterine fibroids. Cochrane Database Syst Rev 2013:CD008994. Bouchard P. Current and future medical treatments for menometrorrhagia during the premenopause. Gynecol Endocrinol 2012;27(Suppl 1):1120–5. Lethaby A, Vollenhoven B, Sowter M. Pre-operative GnRH analogue therapy before hysterectomy or myomectomy for uterine fibroids. Cochrane Database Syst Rev 2001:CD000547. Lethaby AE, Vollenhoven BJ. An evidence-based approach to hormonal therapies for premenopausal women with fibroids. Best Pract Res Clin Obstet Gynaecol 2008;22:307–31. Gupta J, Kai J, Middleton L, Pattison H, Gray R, Daniels J. Levonorgestrel intrauterine system versus medical therapy for menorrhagia. N Engl J Med 2013;368:128–37. Machado RB, de Souza IM, Beltrame A, Bernardes CR, Morimoto MS, Santana N. The levonorgestrel-releasing intrauterine system: its effect on the number of hysterectomies performed in perimenopausal women with uterine fibroids. Gynecol Endocrinol 2013;29:492–5. Zapata LB, Whiteman MK, Tepper NK, Jamieson DJ, Marchbanks PA, Curtis KM. Intrauterine device use among women with uterine fibroids: a systematic review. Contraception 2010;82:41–55. Palomba S, Orio F Jr, Russo T, Falbo A, Tolino A, Lombardi G, et al. Antiproliferative and proapoptotic effects of raloxifene on uterine leiomyomas in postmenopausal women. Fertil Steril 2005;84:154–61. Palomba S, Russo T, Orio F Jr, Tauchmanova L, Zupi E, Panici PL, et al. Effectiveness of combined GnRH analogue plus raloxifene administration in the

treatment of uterine leiomyomas: a prospective, randomized, single-blind, placebo-controlled clinical trial. Hum Reprod 2002;17:3213–9. 97. Palomba S, Zullo F, Orio F Jr, Lombardi G. Does raloxifene inhibit the growth of uterine fibroids? Fertil Steril 2004;81:1719–20. author reply 1720–1. 98. Liu J, Matsuo H, Xu Q, Chen W, Wang J, Maruo T. Concentration-dependent effects of a selective estrogen receptor modulator raloxifene on proliferation and apoptosis in human uterine leiomyoma cells cultured in vitro. Hum Reprod 2007;22:1253–9. 99. Cuzick J, Sestak I, Bonanni B, Costantino JP, Cummings S, DeCensi A, et al. Selective oestrogen receptor modulators in prevention of breast cancer: an updated meta-analysis of individual participant data. Lancet 2013;381: 1827–34. 100. Deng L, Wu T, Chen XY, Xie L, Yang J. Selective estrogen receptor modulators (SERMs) for uterine leiomyomas. Cochrane Database Syst Rev 2012: CD005287. 101. Ohara N. Selective estrogen receptor modulator and selective progesterone receptor modulator: therapeutic efficacy in the treatment of uterine leiomyoma. Clin Exp Obstet Gynecol 2005;32:9–11. 102. Song H, Lu D, Navaratnam K, Shi G. Aromatase inhibitors for uterine fibroids. Cochrane Database Syst Rev 2013:CD009505. 103. Hilario SG, Bozzini N, Borsari R, Baracat EC. Action of aromatase inhibitor for treatment of uterine leiomyoma in perimenopausal patients. Fertil Steril 2009;91:240–3. 104. Chegini N, Kornberg L. Gonadotropin releasing hormone analogue therapy alters signal transduction pathways involving mitogen-activated protein and focal adhesion kinases in leiomyoma. J Soc Gynecol Investig 2003;10:21–6. 105. di Lieto A, de Falco M, Pollio F, Mansueto G, Salvatore G, Somma P, et al. Clinical response, vascular change, and angiogenesis in gonadotropinreleasing hormone analogue–treated women with uterine myomas. J Soc Gynecol Investig 2005;12:123–8. 106. Wang PH, Lee WL, Cheng MH, Yen MS, Chao KC, Chao HT. Use of a gonadotropin-releasing hormone agonist to manage perimenopausal women with symptomatic uterine myomas. Taiwan J Obstet Gynecol 2009;48:133–7. 107. Luo X, Ding L, Xu J, Williams RS, Chegini N. Leiomyoma and myometrial gene expression profiles and their responses to gonadotropin-releasing hormone analog therapy. Endocrinology 2005;146:1074–96. 108. Chabbert-Buffet N, Meduri G, Bouchard P, Spitz IM. Selective progesterone receptor modulators and progesterone antagonists: mechanisms of action and clinical applications. Hum Reprod Update 2005;11:293–307. 109. Sasaki H, Ohara N, Xu Q, Wang J, DeManno DA, Chwalisz K, et al. A novel selective progesterone receptor modulator asoprisnil activates tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–mediated signaling pathway in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2007;92:616–23. 110. Xu Q, Ohara N, Liu J, Amano M, Sitruk-Ware R, Yoshida S, et al. Progesterone receptor modulator CDB-2914 induces extracellular matrix metalloproteinase inducer in cultured human uterine leiomyoma cells. Mol Hum Reprod 2008;14:181–91. 111. Xu Q, Ohara N, Liu J, Nakabayashi K, DeManno D, Chwalisz K, et al. Selective progesterone receptor modulator asoprisnil induces endoplasmic reticulum stress in cultured human uterine leiomyoma cells. Am J Physiol Endocrinol Metab 2007;293:E1002–11. 112. Chen W, Ohara N, Wang J, Xu Q, Liu J, Morikawa A, et al. A novel selective progesterone receptor modulator asoprisnil (J867) inhibits proliferation and induces apoptosis in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2006;91:1296–304. 113. Luo X, Yin P, Coon VJ, Cheng YH, Wiehle RD, Bulun SE. The selective progesterone receptor modulator CDB4124 inhibits proliferation and induces apoptosis in uterine leiomyoma cells. Fertil Steril 2010;93:2668–73. 114. Maruo T, Ohara N, Matsuo H, Xu Q, Chen W, Sitruk-Ware R, et al. Effects of levonorgestrel-releasing IUS and progesterone receptor modulator PRM CDB-2914 on uterine leiomyomas. Contraception 2007;75:S99–103.

VOL. 102 NO. 3 / SEPTEMBER 2014

Fertility and Sterility® 115.

116.

117.

118.

119.

120.

121.

122. 123.

124.

125.

126.

127.

128.

129.

130.

131.

Wang J, Ohara N, Wang Z, Chen W, Morikawa A, Sasaki H, et al. A novel selective progesterone receptor modulator asoprisnil (J867) downregulates the expression of EGF, IGF-I, TGFbeta3 and their receptors in cultured uterine leiomyoma cells. Hum Reprod 2006;21:1869–77. Xu Q, Ohara N, Chen W, Liu J, Sasaki H, Morikawa A, et al. Progesterone receptor modulator CDB-2914 down-regulates vascular endothelial growth factor, adrenomedullin and their receptors and modulates progesterone receptor content in cultured human uterine leiomyoma cells. Hum Reprod 2006;21:2408–16. Engman M, Varghese S, Lagerstedt Robinson K, Malmgren H, Hammarsjo A, Bystrom B, et al. GSTM1 gene expression correlates to leiomyoma volume regression in response to mifepristone treatment. PLoS One 2013;8:e80114. Kettel LM, Murphy AA, Morales AJ, Rivier J, Vale W, Yen SS. Rapid regression of uterine leiomyomas in response to daily administration of gonadotropin-releasing hormone antagonist. Fertil Steril 1993;60:642–6. Murphy AA, Kettel LM, Morales AJ, Roberts VJ, Yen SS. Regression of uterine leiomyomata in response to the antiprogesterone RU 486. J Clin Endocrinol Metab 1993;76:513–7. Murphy AA, Morales AJ, Kettel LM, Yen SS. Regression of uterine leiomyomata to the antiprogesterone RU486: dose-response effect. Fertil Steril 1995;64:187–90. Donnez J, Vazquez F, Tomaszewski J, Nouri K, Bouchard P, Fauser BC, et al. Long-term treatment of uterine fibroids with ulipristal acetate. Fertil Steril 2014;101:1565–73.e1–18. Molecule of the month: asoprisnil. Drug News Perspect 2005;18:404. Chwalisz K, Larsen L, Mattia-Goldberg C, Edmonds A, Elger W, Winkel CA. A randomized, controlled trial of asoprisnil, a novel selective progesterone receptor modulator, in women with uterine leiomyomata. Fertil Steril 2007;87:1399–412. Nieman LK, Blocker W, Nansel T, Mahoney S, Reynolds J, Blithe D, et al. Efficacy and tolerability of CDB-2914 treatment for symptomatic uterine fibroids: a randomized, double-blind, placebo-controlled, phase IIb study. Fertil Steril 2011;95:767–72.e1–2. Engman M, Granberg S, Williams AR, Meng CX, Lalitkumar PG, GemzellDanielsson K. Mifepristone for treatment of uterine leiomyoma. A prospective randomized placebo controlled trial. Hum Reprod 2009;24:1870–9. Wilkens J, Chwalisz K, Han C, Walker J, Cameron IT, Ingamells S, et al. Effects of the selective progesterone receptor modulator asoprisnil on uterine artery blood flow, ovarian activity, and clinical symptoms in patients with uterine leiomyomata scheduled for hysterectomy. J Clin Endocrinol Metab 2008;93:4664–71. Levens ED, Potlog-Nahari C, Armstrong AY, Wesley R, Premkumar A, Blithe DL, et al. CDB-2914 for uterine leiomyomata treatment: a randomized controlled trial. Obstet Gynecol 2008;111:1129–36. Eisinger SH, Fiscella J, Bonfiglio T, Meldrum S, Fiscella K. Open-label study of ultra low-dose mifepristone for the treatment of uterine leiomyomata. Eur J Obstet Gynecol Reprod Biol 2009;146:215–8. Fiscella K, Eisinger SH, Meldrum S, Feng C, Fisher SG, Guzick DS. Effect of mifepristone for symptomatic leiomyomata on quality of life and uterine size: a randomized controlled trial. Obstet Gynecol 2006;108:1381–7. Feng C, Meldrum S, Fiscella K. Improved quality of life is partly explained by fewer symptoms after treatment of fibroids with mifepristone. Int J Gynaecol Obstet 2010;109:121–4. Eisinger SH, Bonfiglio T, Fiscella K, Meldrum S, Guzick DS. Twelve-month safety and efficacy of low-dose mifepristone for uterine myomas. J Minim Invasive Gynecol 2005;12:227–33.

VOL. 102 NO. 3 / SEPTEMBER 2014

132. 133. 134.

135.

136.

137.

138.

139.

140.

141. 142.

143.

144.

145. 146.

147.

148.

149.

150.

Critchley HO. Three perspectives on progestins: clinical research perspective. Endocr News 2012;February:27–30. Patlak M. Focus on fibroids. Endocr News 2011;December:32–5. Fiscella J, Bonfiglio T, Winters P, Eisinger SH, Fiscella K. Distinguishing features of endometrial pathology after exposure to the progesterone receptor modulator mifepristone. Hum Pathol 2011;42:947–53. Mutter GL, Bergeron C, Deligdisch L, Ferenczy A, Glant M, Merino M, et al. The spectrum of endometrial pathology induced by progesterone receptor modulators. Mod Pathol 2008;21:591–8. Williams AR, Bergeron C, Barlow DH, Ferenczy A. Endometrial morphology after treatment of uterine fibroids with the selective progesterone receptor modulator, ulipristal acetate. Int J Gynecol Pathol 2012;31:556–69. Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Rajaratnam V, Al-Hendy A. Antiproliferative and proapoptotic effects of epigallocatechin gallate on human leiomyoma cells. Fertil Steril 2009;94: 1887–93. Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Sharan C, Rajaratnam V, et al. Green tea extract inhibits proliferation of uterine leiomyoma cells in vitro and in nude mice. Am J Obstet Gynecol 2010;202: 289.e1–9. Lee BS, Margolin SB, Nowak RA. Pirfenidone: a novel pharmacological agent that inhibits leiomyoma cell proliferation and collagen production. J Clin Endocrinol Metab 1998;83:219–23. Paffoni A, Somigliana E, Vigano P, Benaglia L, Cardellicchio L, Pagliardini L, et al. Vitamin D status in women with uterine leiomyomas. J Clin Endocrinol Metab 2013;98:E1374–8. Baird DD, Hill MC, Schectman JM, Hollis BW. Vitamin D and the risk of uterine fibroids. Epidemiology 2013;24:447–53. Halder SK, Goodwin JS, Al-Hendy A. 1,25-Dihydroxyvitamin D3 reduces TGF-beta3–induced fibrosis-related gene expression in human uterine leiomyoma cells. J Clin Endocrinol Metab 2011;96:E754–62. Halder SK, Osteen KG, Al-Hendy A. 1,25-Dihydroxyvitamin D3 reduces extracellular matrix–associated protein expression in human uterine fibroid cells. Biol Reprod 2013;89:150. Halder SK, Osteen KG, Al-Hendy A. Vitamin D3 inhibits expression and activities of matrix metalloproteinase-2 and -9 in human uterine fibroid cells. Hum Reprod 2013;28:2407–16. Blauer M, Rovio PH, Ylikomi T, Heinonen PK. Vitamin D inhibits myometrial and leiomyoma cell proliferation in vitro. Fertil Steril 2009;91:1919–25. Halder SK, Sharan C, Al-Hendy A. 1,25-Dihydroxyvitamin D3 treatment shrinks uterine leiomyoma tumors in the Eker rat model. Biol Reprod 2012;86:116. Scharla SH, Minne HW, Waibel-Treber S, Schaible A, Lempert UG, Wuster C, et al. Bone mass reduction after estrogen deprivation by longacting gonadotropin-releasing hormone agonists and its relation to pretreatment serum concentrations of 1,25-dihydroxyvitamin D3. J Clin Endocrinol Metab 1990;70:1055–61. Waibel-Treber S, Minne HW, Scharla SH, Bremen T, Ziegler R, Leyendecker G. Reversible bone loss in women treated with GnRHagonists for endometriosis and uterine leiomyoma. Hum Reprod 1989;4: 384–8. Kongnyuy EJ, Wiysonge CS. Interventions to reduce haemorrhage during myomectomy for fibroids. Cochrane Database Syst Rev 2009: CD005355. Eisinger SH, Meldrum S, Fiscella K, le Roux HD, Guzick DS. Low-dose mifepristone for uterine leiomyomata. Obstet Gynecol 2003;101:243–50.

639

Fibroid growth and medical options for treatment.

Although fibroids are common benign tumors, their impact on women's quality of life can be considerable. The most frequent symptoms are uterine bleedi...
544KB Sizes 2 Downloads 4 Views