ArticlePDF AvailableLiterature Review

Alternative Oral Agents in Prophylaxis and Therapy of Uterine Fibroids-An Up-to-Date Review

Authors:

Abstract and Figures

Uterine fibroids (UFs) are the most common tumors of the female genital tract. The effect of UFs on the quality of life and the overall cost of treatment are significant issues worldwide. Tumor size and location are the two specific factors which influence the occurrence of symptoms, the need for, and method of, treatment (some tumors require surgery while some can be treated with selected drugs). Primary prevention and treatment of early UF disease are worthy goals that might have a great impact on health care systems. Several treatments and prophylactic methods can be used in this endeavor. This publication presents current data about lesser-known substances which may have a beneficial effect on the treatment or prophylaxis of UFs and can be administered orally, serving as an alternative to (or complement of) surgery or selective progesterone receptor modulators (SPRMs). Early prevention and treatment of UFs in women from high-risk groups should be our priority. Innovative forms of UF management are under intensive investigation and may be promising options in the near future. Many of them evaluated vitamin D, paricalcitol, epigallocatechin gallate (EGCG), elagolix, aromatase inhibitors (AIs), and cabergoline and deemed them to be safe and effective. The next step in such projects should be properly constructed randomized control trials (RCTs), carried out by successive phases.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Review
Alternative Oral Agents in Prophylaxis and Therapy
of Uterine Fibroids—An Up-to-Date Review
Michał Ciebiera 1, *ID , Krzysztof Łukaszuk 2,3, Bła˙
zej M˛eczekalski 4, Magdalena Ciebiera 5,
Cezary Wojtyła 1, Aneta Słabuszewska-Jó´zwiak 1and Grzegorz Jakiel 1
1Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education,
00-416 Warsaw, Poland; czwo@op.pl (C.W.); as.jozwiak@op.pl (A.S.-J.); grzegorz.jakiel1@o2.pl (G.J.)
2Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences,
Medical University of Gdansk, 80-210 Gdansk, Poland; luka@gumed.edu.pl
3INVICTA Fertility and Reproductive Center, 80-172 Gdansk, Poland
4
Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-513 Poznan, Poland;
blazejmeczekalski@yahoo.com
5Students’ Scientific Association at the I Department of Obstetrics and Gynecology,
Medical University of Warsaw, 02-015 Warsaw, Poland; mciebiera93@gmail.com
*Correspondence: michal.ciebiera@gmail.com; Tel.: +48-607-155-177
Received: 17 October 2017; Accepted: 28 November 2017; Published: 1 December 2017
Abstract:
Uterine fibroids (UFs) are the most common tumors of the female genital tract. The effect of
UFs on the quality of life and the overall cost of treatment are significant issues worldwide. Tumor size
and location are the two specific factors which influence the occurrence of symptoms, the need for,
and method of, treatment (some tumors require surgery while some can be treated with selected
drugs). Primary prevention and treatment of early UF disease are worthy goals that might have a
great impact on health care systems. Several treatments and prophylactic methods can be used in this
endeavor. This publication presents current data about lesser-known substances which may have a
beneficial effect on the treatment or prophylaxis of UFs and can be administered orally, serving as an
alternative to (or complement of) surgery or selective progesterone receptor modulators (SPRMs).
Early prevention and treatment of UFs in women from high-risk groups should be our priority.
Innovative forms of UF management are under intensive investigation and may be promising options
in the near future. Many of them evaluated vitamin D, paricalcitol, epigallocatechin gallate (EGCG),
elagolix, aromatase inhibitors (AIs), and cabergoline and deemed them to be safe and effective.
The next step in such projects should be properly constructed randomized control trials (RCTs),
carried out by successive phases.
Keywords:
uterine fibroid; leiomyoma; vitamin D; paricalcitol; epigallocatechin gallate; elagolix;
aromatase inhibitors; cabergoline; pharmacology
1. Introduction
Uterine fibroids (UFs)—benign monoclonal tumors protruding from myometrial smooth muscle
cells—are the most common pathology of the female genital tract [
1
3
]. The morphology of UFs may
vary greatly. They can be solitary or appear in multiple clusters. Also, their size range is considerable,
too, from miniscule to giant masses of over 20 cm in diameter. UFs affect 25–80% women, depending
on the population and the risk factors [
2
,
4
,
5
]. The majority of UFs are asymptomatic, and menopause
generally results in tumor atrophy, but the symptomatic tumors constitute a major problem for the
vast number of affected women. The wide range of UF-associated symptoms includes iron deficiency
anemia, abdominal and pelvic pain, gastrointestinal disorders, dysuria, female infertility, and severe
obstetric complications [2,4,6].
Int. J. Mol. Sci. 2017,18, 2586; doi:10.3390/ijms18122586 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2017,18, 2586 2 of 19
This pathology varies greatly in relation to age. These tumors are more prevalent among older
populations [2,7,8]. UFs are not observed in pre-pubescent girls and are a rare finding in adolescents,
indicating that they depend on hormonal changes [
7
,
9
]. According to the available data, the growth of
UFs depends mostly on the influence of steroid hormones [
1
,
10
,
11
]. Estrogens have been known to play
an important role in the pathophysiology of UFs, but the latest research has suggested progesterone
as the main factor initiating pathological uterine muscle differentiation and abnormal growth [
1
].
The main mechanism of progesterone-induced UFs tumorigenesis consists of an increase in the
concentration of selected growth factors [
1
,
12
,
13
]. Also, a significant part of UFs occurs due to
a genetic abnormality [
1
]. In patients with positive family history, the risk for developing UFs
approximately 4 times higher than in the general population [
14
]. According to a study by Makinen
et al., specific mutations within the gene encoding the mediator complex subunit 12 (MED12) were
detected in the examined UFs [
15
]. Nowadays, it is a known fact that even up to 80% of UFs have a
mutation in MED12 [1,15,16].
UFs are a major public health problem. By the age of 50, they might develop in almost 80% and
70% of the African-American and the Caucasian women, respectively [
3
,
8
]. The effects of UFs on the
quality of life (QoL) and the overall cost of treatment are significant but often remain unaddressed
or marginalized [
17
]. As far as QoL for women in general is concerned, Soliman et al. have recently
demonstrated that women who rated their UF-related symptoms as “severe” had significantly worse
QoL as compared to their peers with mild symptoms [
18
]. QoL deteriorated considerably with the
increasing number and severity of symptoms [
18
]. A 2015 review of the literature on direct and
indirect costs of UF management revealed that substantial sums of money are generated by UFs [
19
],
and included not only the price of medicines, medical staff salaries, or the cost of surgical treatment,
but also the hidden costs of work absence, hospitalization, control visits, and preoperative diagnostic
tests. The annual direct and indirect costs of UFs in the United States have been estimated to be
between $4.1–$9.4 billion [
3
,
19
,
20
], and $1.6–$17.2 billion, respectively [
20
]. In the United States,
the total cost of treatment of a single patient with UFs ranges from $11,700 to $25,000 per year after the
diagnosis or surgery [
19
,
21
]. According to a well-known study by Cardozo et al., the total annual cost
of UF treatment in the United States has been estimated at $34.4 billion [20].
Tumor size and location determine the occurrence of symptoms, the need for treatment, and the
treatment method. Other important determinants include symptom severity, patient age and
reproductive plans, the risk for malignancy, skills and expertise of the gynecologists and access
to proper medical equipment [
2
,
6
]. Due to the benign nature of UFs, treatment resulting in the
least morbidity and lowest risk should be chosen, if possible [
2
,
6
,
22
]. Multiple UF management
options are currently available but surgery remains the method of choice and is often accompanied by
pharmacological treatment or pretreatment [
2
,
22
24
]. The most common complaint—menorrhagia—is
managed with surgical procedures like ablation, myomectomy or uterine artery embolization or,
more recently, by pharmacotherapy [
6
,
22
]. The available treatments for UFs, including hysterectomy,
myomectomy, embolization, and gonadotropin-releasing hormone (GnRH) agonists, are effective but
are recommended in more advanced stages of the disease, especially since they are neither low-cost
nor free of risk for adverse events [
25
,
26
]. Ulipristal acetate (UPA), a selective progesterone receptor
modulator (SPRM), is the most common UF pharmacological treatment [
6
,
22
,
26
28
]. Clinical trials
have demonstrated that UPA is effective for controlling UF-related excessive uterine bleeding and
reducing fibroid size [
6
,
22
,
26
,
27
]. Treatment schemes with UPA have recently become the gold standard
in modern management of UFs [
27
]. In those schemes, UPA is administered as first-line therapy to
prepare UFs for surgery or, in case of good response, to lead to a condition when surgical treatment
is no longer necessary [
6
,
29
]. However, UPA is relatively expensive and not accessible to everyone,
nor is it a substance which can be widely used in prevention [
2
,
22
]. In spite of the ongoing research,
the currently available pharmacological therapies are short-term, to avoid the risk of chronic hormonal
therapy, and are accompanied by long-term adverse side effects.
Int. J. Mol. Sci. 2017,18, 2586 3 of 19
One of the major problems associated with UFs is that they are understudied, and therefore,
much research is needed in this field [
25
,
30
]. The development of UFs is multi-factorial in origin,
thus specific methods of prophylaxis are currently unavailable. Various recent attempts to create an
inexpensive, safe, and effective drug for the prevention and treatment of UFs are still in the early
stages of the process [
31
]. Still, several treatments and prophylactic methods can be used in this
endeavor [
31
]. Current findings suggest that substances contained in green tea [
32
], vitamin D [
25
,
33
],
elagolix [
34
], paricalcitol [
35
], gestrinone [
36
], and others may become future preparations for chronic
treatment with minimal or moderate side effects. Similar concepts are discussed in an attempt to create
a specific method of prophylaxis of UFs in high-risk subjects [
14
,
25
,
37
]. High costs and low efficacy
are the key points in UF therapy. If more such substances are known, it will be possible to investigate
them further (new dosages and schemes), and combine their effect with the known agents to achieve
better performance. This, in turn, may have a great impact upon the health of millions of women in
the future.
This publication presents current data about lesser-known substances which may have a beneficial
effect on the treatment or prophylaxis of UFs and can be administered orally, serving as an alternative
to (or complement) surgery or SPRMs.
2. Materials and Methods
This article presents an up-to-date review of publications regarding the current role of
alternative agents in UF treatment and prophylaxis. A literature search was conducted in
PubMed of the National Library of Medicine using the following key words: “uterine fibroid”,
“pharmacotherapy”, “vitamin D”, “vitamin D analog”, “paricalcitol”, “gestrinone”, “elagolix”,
“aromatase inhibitor”, “epigallocatechin gallate”, “green tea”, “curcumin”, and “cabergoline”.
The above keywords were selected to reflect possible oral agents in the prophylaxis and therapy
of UFs. During our search, we combined the key words into pairs, which resulted in: “uterine fibroid”
and “pharmacotherapy”—1694 publications; “uterine fibroid” and “vitamin D”—40 publications;
“uterine fibroid” and “vitamin D analog”—2 publications; “uterine fibroid” and “paricalcitol”—
2 publications; “uterine fibroid” and “gestrinone”—19 publications; “uterine fibroid” and “elagolix”—
1 publication; “uterine fibroid” and “aromatase inhibitor”—73 publications; “uterine fibroid”
and “epigallocatechin gallate”—7 publications; “uterine fibroid” and “green tea”—8 publications;
“uterine fibroid” and “curcumin”—4 publications; “uterine fibroid” and “cabergoline”—4 publications.
If the search was duplicated the papers were excluded. The aim of the review was to critically evaluate
the current data about lesser-known substances which might have a serious impact on UFs and
UF-related symptoms and which can be administered orally. The results of the available studies in
English, published up to October 2017, have been discussed in this article. Additional important
and impactful articles and reviews were considered, when relevant. Articles were excluded if
they were published in languages other than English. After reviewing the titles and abstracts,
approximately 150 full articles have been evaluated.
3. Discussion
3.1. Vitamin D
The least studied factors which affect the risk for UF occurrence are related to lifestyle, diet,
nutrition, or place of residence. This can be the gateway to effective prevention of UFs. Vitamin D is
the part of the fat-soluble steroid compound group which exerts comprehensive action on the human
body [
38
]. Sunlight exposure is the main source of vitamin D for humans [
39
]. This vitamin can also
be extracted from diet or food supplements. However, very few natural foods contain vitamin D
in the appropriate amount. Marine fish, fish oils, and fortified food are among the best sources of
vitamin D [40].
Int. J. Mol. Sci. 2017,18, 2586 4 of 19
Vitamin D is believed to reduce the risk for chronic illnesses and malignancies [
38
,
41
,
42
], and has
a potent immunomodulatory function. Vitamin D receptor (VDR) is expressed in almost all cells of
the immune system where their function is regulated [
43
,
44
]. Vitamin D regulates cell proliferation
and differentiation, inhibits angiogenesis, and stimulates apoptosis [
33
,
45
]. Vitamin D deficiency is
believed to be a major risk factor in the development of UFs [
14
,
33
,
37
,
46
]. According to the recent
literature reports, mean 25-hydroxyvitamin D (25(OH)D) levels are significantly lower in women with
UFs as compared to controls [
47
,
48
]. The same was confirmed in African-Americans, who are more
likely to present vitamin D deficiency and UFs [49]. Three main studies on reduced serum vitamin D
levels in women with UFs have revolutionized the approach in this field and have unambiguously
targeted research for the upcoming years [46,50,51].
New research revealed that a protective effect of vitamin D on UFs is highly likely. VDR is
expressed in both, myometrial and tumor tissues [
52
]. A correlation between low 25(OH)D serum
levels and as increased risk for developing UFs was evaluated [
14
,
33
,
46
,
51
]. According to
in vitro
experiments, UF cells are highly sensitive to the growth-inhibiting effect of the active form of vitamin D.
In those studies, vitamin D induced apoptosis in cultured human UF cells through the downregulation
of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase 1 (CDK1), and B-cell lymphoma-2
(BCL2) and suppressed catechol-O-methyltransferase (COMT) expression and activity [
31
,
52
54
]
(Figure 1).
Int. J. Mol. Sci. 2017, 18, 2586 4 of 19
with UFs as compared to controls [47,48]. The same was confirmed in African-Americans, who are
more likely to present vitamin D deficiency and UFs [49]. Three main studies on reduced serum
vitamin D levels in women with UFs have revolutionized the approach in this field and have
unambiguously targeted research for the upcoming years [46,50,51].
New research revealed that a protective effect of vitamin D on UFs is highly likely. VDR is
expressed in both, myometrial and tumor tissues [52]. A correlation between low 25(OH)D serum
levels and as increased risk for developing UFs was evaluated [14,33,46,51]. According to in vitro
experiments, UF cells are highly sensitive to the growth-inhibiting effect of the active form of vitamin
D. In those studies, vitamin D induced apoptosis in cultured human UF cells through the
downregulation of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase 1 (CDK1), and
B-cell lymphoma-2 (BCL2) and suppressed catechol-O-methyltransferase (COMT) expression and
activity [31,52–54] (Figure 1).
Figure 1. Schematic diagram of uterine fibroid pathophysiology with places for potential use of an
alternative agent therapy. Gonadotropin-releasing hormone (GnRH), estrogen receptor (ER),
progesterone receptor (PR), transforming growth factor beta (TGF-β), extracellular matrix (ECM),
epigallocatechin gallate (EGCG).
In a study by Halder et al., UFs in rats have diminished in size under the influence of this vitamin
on the transforming growth factor beta 3 (TGF-β3) pathway, as vitamin D reduces the TGF-β3-
induced fibrosis [55]. In a different study, Halder et al. proved that administration of
1,25-dihydroxyvitamin D reduced the expression of type I collagen and fibronectin in UFs [56]. In
our recent study, we also found that vitamin D might play a role in decreasing TGF-β3 levels in
women [14,21] (Figure 1).
Vitamin D toxicity is very rare [41]. The recommended level of vitamin D is defined as a 25(OH)D
value of more than 30 ng/mL [57]. According to the summarized supplementation guidelines
developed in 2017 (7000 international units (IU)/day or 50,000 IU/week), deficiency may be remedied
by adequate supplementation [58]. Vitamin D supplementation and sunlight exposure are the two
main measure for prevention of a wide spectrum of disorders, including UFs [59,60].
Our study emphasizes the need for new clinical trials to assess the actual effectiveness of vitamin
D in UF therapy. The next step should be a properly constructed, randomized clinical trial. Vitamin
D seems to be a promising, safe, effective, and low-cost treatment of UFs. In cases of positive
observations, vitamin D preparations could become a new generation of anti-UF drugs, with the
additional beneficial pleiotropic effect. Additional skeletal (bones and ligaments) and extra-skeletal
Figure 1.
Schematic diagram of uterine fibroid pathophysiology with places for potential use of
an alternative agent therapy. Gonadotropin-releasing hormone (GnRH), estrogen receptor (ER),
progesterone receptor (PR), transforming growth factor beta (TGF-
β
), extracellular matrix (ECM),
epigallocatechin gallate (EGCG).
In a study by Halder et al., UFs in rats have diminished in size under the influence of this vitamin
on the transforming growth factor beta 3 (TGF-
β
3) pathway, as vitamin D reduces the TGF-
β
3-induced
fibrosis [
55
]. In a different study, Halder et al. proved that administration of 1,25-dihydroxyvitamin
D reduced the expression of type I collagen and fibronectin in UFs [
56
]. In our recent study, we also
found that vitamin D might play a role in decreasing TGF-β3 levels in women [14,21] (Figure 1).
Vitamin D toxicity is very rare [
41
]. The recommended level of vitamin D is defined as a
25(OH)D value of more than 30 ng/mL [
57
]. According to the summarized supplementation guidelines
developed in 2017 (7000 international units (IU)/day or 50,000 IU/week), deficiency may be remedied
by adequate supplementation [
58
]. Vitamin D supplementation and sunlight exposure are the two
main measure for prevention of a wide spectrum of disorders, including UFs [59,60].
Int. J. Mol. Sci. 2017,18, 2586 5 of 19
Our study emphasizes the need for new clinical trials to assess the actual effectiveness of
vitamin D in UF therapy. The next step should be a properly constructed, randomized clinical trial.
Vitamin D seems to be a promising, safe, effective, and low-cost treatment of UFs. In cases of positive
observations, vitamin D preparations could become a new generation of anti-UF drugs, with the
additional beneficial pleiotropic effect. Additional skeletal (bones and ligaments) and extra-skeletal
(other organs and overall homeostasis) advantages support the use of vitamin D as a prophylactic
agent [
25
,
38
,
58
]. Further data are needed to fully comprehend the exact role of vitamin D in the
pathophysiology of UF. To the best of our knowledge, there are still no randomized controlled trials
(RCTs) on this subject, the main reason for that being lack of on the cut-off thresholds for vitamin D
deficiency, optimal levels and dosage. However, it seems that the near future will finally bring the
consensus [58].
3.2. Vitamin D Analogs—Paricalcitol
Due to the potentially adverse or even toxic effects of vitamin D in very high concentrations,
consideration should be given to possible alternatives [
35
]. Vitamin D analogs are already present
on the pharmaceutical market and their possible beneficial effects are intriguing. Paricalcitol (a well-
known vitamin D analog) is a selective VDR activator which is registered and used in secondary
hyperparathyroidism [
61
]. According to Bouillon et al., paricalcitol has less calcemic activity than
vitamin D and might be a new option in modern therapy [
62
]. Paricalcitol can cause electrolyte
abnormalities like hypercalcemia and hyperphosphatemia, and is contraindicated in patients who
use digoxin, thiazide diuretics, and ketoconazole [
63
]. Paricalcitol has a proven inhibitory effect on
cell proliferation and fibrosis [
35
,
64
]. It has also been found to reduce the increase of the extracellular
matrix (ECM) accumulation in the peritoneum of dialyzed patients [
65
]. In a study by Stavenuiter et al.,
the authors concluded that the immunomodulatory effects of paricalcitol have beneficially contributed
to the development of ECM (limiting its thickening), and that VDR activation can partly increase the
kidney filtration rate due to limitation of angiogenesis and thickening of ECM [65].
Vitamin D reduces inflammation and fibrosis by VDR activation [
66
,
67
]. According to a study by
Protic et al., cytokines play a key role in inflammation and tissue remodeling regulation, indicating
that they may be responsible for UF-associated symptoms such as pain or infertility [
45
]. TGF-
β
(mainly TGF-
β
3 isoform) plays one of the major roles in UF-related fibrosis [
14
,
54
,
68
,
69
]. In their study,
Oblak et al. demonstrated a significant decrease in TGF-
β
serum concentrations in transplant recipients
who received paricalcitol as compared to controls [
66
]. Another study by Gonzales-Mateo et al.,
showed that the use of paricalcitol strongly reduced peritoneal interleukin (IL)-17 levels, which might
correlate with lower peritoneal membrane deterioration and lower fibrotic response in the peritoneum
in mice models [
44
]. As for the relevance of the abovementioned studies to UF therapy, UFs consist
largely of ECM with embedded cells and excessive ECM production is considered to be one of the
mechanisms of UF formation [
70
]. The ECM which builds the uterus is much more abundant than
properly functioning myometrial tissue [
71
]. Matrix metalloproteinases (MMPs) are calcium-dependent
endopeptidases which degrade the structure and rebuild the ECM [
72
]. ECM undergoes a continuous
and balanced reconstruction process which contributes to the maintenance of its proper amount and
hardness. Matrix enzymes are regulated by special inhibitors of ECM metalloproteinases (tissue
inhibitor of metalloproteinases (TIMP)) [
73
] (Figure 1). Recent research has demonstrated that vitamin
D increased TIMP expression in uterine myometrium [56].
In conclusion, paricalcitol effectively reduces the proliferation of human leiomyoma cell cultures,
reduces fibroid tumor volumes, and induces apoptosis in UFs [
35
]. In our opinion, paricalcitol may
be an effective agent in UF therapy. It has great potential as an effective drug or co-drug for the
conservative treatment of UFs. However, advanced clinical trials are necessary to confirm its efficacy
and safety [
35
]. To the best of our knowledge, there have been no RCT on the use of paricalcitol in
UF therapy.
Int. J. Mol. Sci. 2017,18, 2586 6 of 19
3.3. Green Tea Extract—Epigallocatechin Gallate
Tea is one of the most popular beverages in the world. Green tea is made from Camellia sinensis
leaves [
74
,
75
]. It originated in China, but later spread to many parts of Asia. Green tea contains
several catechins with treatment potential: epigallocatechin gallate (EGCG), epigallocatechin (EGC),
epicatechin gallate (ECG), epicatechins, and flavonols [76].
Various researches suggested that polyphenols have antioxidative, anticarcinogenic, and
anti-inflammatory effects in humans [
75
,
77
]. Currently, EGCG, an ester of epigallocatechin and gallic
acid, is one of the most investigated green tea polyphenols in medicine. The effect of EGCG was studied
in oncology, where it presented beneficial effects [
78
,
79
]. EGCG is an anti-obesity and anti-adipogenic
agent due to its activity in the adipogenic differentiation of mice mesenchymal stem cell inhibition [
80
].
One of the key pathways in this process is based on the gamma protein blocking activity of EGCG
human peroxisome proliferator activated receptor gamma (PPAR
γ
) [
80
]. In non-acute fatty liver
disease animal models, EGCG reduced the concentration of pro-fibrotic and pro-inflammatory factors
in several different pathways [
81
]. In studies on pulmonary fibrosis, EGCG significantly inhibited
fibroblast activation and ECM accumulation by blocking the TGF-β1 signaling pathway [82].
The question arises whether adequate amounts of green tea might exhibit protective effect against
UFs. In an interesting study by Matsuzaki et al., EGCG was found to inhibit proliferation and invasion
of endometrial implants in animals [
83
]. In molecular findings, EGCG reduced the TGF-
β
-dependent
mRNA expression of fibrosis mediators [
83
] (Figure 1). These authors concluded that EGCG might be
a potential candidate in the treatment of endometriosis [
83
]. The only problem with this agent is its
low bioavailability in natural sources [
83
85
], but several studies regarding the improved derivatives,
analogs, or prodrugs of EGCG have demonstrated positive and promising results [86,87].
The effectiveness of EGCG has been studied in UF pathophysiology, and the results are more
than interesting [
72
]. Some authors believe that it might be a new weapon in the battle against
UFs [
22
,
88
]. EGCG was found to inhibit proliferation and reduce the volume of UF tumors in
mice [
89
]. Various studies have demonstrated its effect on inhibition of proliferation and induction of
apoptosis in human UF cells [
32
]. In those studies, EGCG was a modulation agent in the proliferation,
transformation, and inflammation in UF tumors [
32
,
50
]. The pathophysiological pathways proving
EGCG modulation potential included PCNA, CDK1 and BCL2 (same as in vitamin D) [
31
]. Studies by
Zhang et al., demonstrated that UFs in mice and human UF cell colonies were shrinking under the
influence of EGCG [
32
,
89
] (Figure 1). Roshdy et al. evaluated the oral use of EGCG in women with
symptomatic UFs and the results are highly encouraging. Women who received 800 mg of 45%
EGCG for 4 months had a significant reduction of the total fibroid tumor burden and alleviation of
symptom severity as compared to controls [
88
]. More recently, Ahmed et al. presented interesting
data about potent prodrugs and analogs of EGCG which resulted in enhanced bioavailability, stability,
and antiproliferative and antifibrotic properties. These findings enable us to discern that green tea
extract might have found its place in UF management. Those authors are planning broader studies
regarding these drugs which, in our opinion, might be revolutionary [
87
]. If the results about long-term
safety and efficacy are positive, EGCG might become the new quality treatment in UF management [
87
].
As EGCG is now available as a dietary supplement [
90
], it would also be interesting to investigate the
possible effects of combining it with vitamin D or paricalcitol.
The literature lack data on safe dose levels of pure EGCG. According to the available sources,
hepatotoxicity was observed in animal studies with the use of high doses of EGCG. Ramachandran
estimated the maximum tolerable dose of EGCG at 67.8 mg/kg orally (in the course of two weeks) [
91
].
In our opinion, available data suggest that EGCG has a very high potential to become an
alternative agent in the prophylaxis or anti-UF therapy, but RCTs on this subject are unavailable.
3.4. Elagolix
GnRH agonists are highly effective drugs in the management of symptomatic
endometriosis [92,93]
,
as well as in UFs [
2
,
6
]. Alas, they can cause severe hypoestrogenic effects, such as vasomotor symptoms
Int. J. Mol. Sci. 2017,18, 2586 7 of 19
(e.g., hot flashes), which will limit the treatment duration or cause the patient to discontinue the therapy
due to the reduced QoL [
93
]. The treatment of benign gynecological diseases does not require full
estrogen suppression to partly or even fully reduce the symptoms [93].
Elagolix is a potent and selective non-peptide antagonist of the GnRH receptor [
94
,
95
]. It is one of
very few (with relugolix [
96
], and OBE2109 [
97
]) GnRH antagonists which can be administered
orally, and its administration in proper doses suppresses the reproductive endocrine axis in
healthy premenopausal women [
98
]. According to several studies, elagolix is superior to the
currently available GnRH agonists and antagonists due to its manageability, rapid onset of action,
good bioavailability, rapid reversibility, and minor side effects [
93
,
98
]. Very recently Ng et al. published
their data supporting elagolix administration in premenopausal women with sex-hormone dependent
diseases [
93
]. In their opinion, elagolix might be useful in modulating the pituitary–ovarian axis by
dose-dependent partial or full suppression [
93
] (Figure 1). Studies about the intense pain associated
with endometriosis demonstrated that elagolix has good efficacy and tolerability. Very recently,
Taylor et al. studied 872 women and found that elagolix has great potential as an important treatment
option for women suffering from endometriosis. In this study, women who received elagolix had
significantly lower scores for dysmenorrhea and non-menstrual pelvic pain than placebo controls [
99
].
Even if its activity and influence have been tested mostly in endometriosis [
95
,
99
], there are
also studies about the use of elagolix in UF management. Recently, Archer et al. presented their
results regarding different doses of elagolix in heavy menstrual bleeding [
34
]. Elagolix reduced heavy
menstrual bleeding symptoms in women with UFs. Optimal results were obtained using the dose of
300 mg twice per day [
34
]. These authors observed hypoestrogenic side effects such hot flashes (most
frequent), nausea, or headache in some patients, but they were significantly reduced by the use of
low-dose estrogen add-back therapy [
34
]. They concluded that elagolix is a candidate for becoming
the new medication in the chronic treatment of clinically symptomatic UFs and should be moved
to the next phase of the clinical trials [
34
]. Phase III studies determined that elagolix achieves its
primary endpoints in endometriosis, while adverse events during its use were described as mild to
moderate (the most common ones are headache, nausea and anxiety) [
100
]. Data about its toxicity
remains limited.
The pharmaceutical company currently undertaking elagolix clinical trials intends to submit
elagolix as a drug in endometriosis therapy to the United States Food and Drug Administration in
2017 [
101
]. Phase III clinical trials of elagolix in UF therapy are underway [
101
]. Even if this new GnRH
antagonist is well tolerated, much remains to be discovered. We need further studies in larger numbers
for longer time periods to gain additional information about the efficacy, tolerability, and compliance
of elagolix [34,100].
3.5. Aromatase Inhibitors
Aromatase is one of the major enzymes responsible for estrogen synthesis. It can be found
in various tissues, including gonads, adipocytes, blood vessels, and the nervous, integumentary,
and skeletal systems, as well as endometrial implants, UF tumors, or gynecological cancers [102,103].
Aromatase inhibitors (AIs) decrease the production of estrogens by blocking or inactivating aromatase.
AIs are a class of drugs which demonstrate the antiestrogenic effect (Figure 1). The most well-known
AIs are anastrozole, letrozole, and fadrozole. They are mainly used in the treatment of gynecological
cancers (breast or endometrial cancer) [
104
], to suppress estrogen production [
104
], and to treat
endometriosis [
105
107
] or infertility (induction of the ovulation) [
107
]. AIs have good efficacy,
but they can cause common side effects, such hot flashes, bone loss, mood swings, vaginal dryness,
ovarian cyst formation, or body pain [
108
]. The use of AIs in premenopausal women can rise the
plasma estrogens levels by the stimulation of gonadotropins [
109
,
110
]. In peri- or post-menopausal
women, estrogens are mainly produced in the peripheral tissues (e.g., adipocytes). Using AIs in this
patient group would decrease estrogen levels in both, plasma and tissues [
109
,
110
]. Postmenopausal
Int. J. Mol. Sci. 2017,18, 2586 8 of 19
women are already exposed to a low-hormone environment, and the use of AIs can lead to better
results in the treatment of hormone-dependent diseases [111].
There are several studies about the effect of AIs on UF and UF-related symptoms [108]. Notably,
UF tissue expresses aromatase in higher amounts than normal myometrial tissue [
112
], and aromatase
has decreased expression in Japanese and Caucasian subjects as compared to African-Americans,
who have a higher rate of UFs [
113
]. On the basis of these findings, we can expect better results
with the use of AIs in African-American populations [
114
]. Anastrozole has been described as an
agent which shrinks UF tumors and reduces the clinical symptoms in post- [
115
] and peri-menopausal
women without serious adverse events [
116
]. AIs were also found to have an advantage in the rapid
onset of action [
115
,
116
]. In the study by Varelas et al., 3 month-use of anastrazole reduced mean UF
volume by almost two-thirds, and these authors concluded that the use of AIs should be proposed
to all women who want to avoid risky surgery [
117
]. In 2003, Shozu et al. published studies where
AIs were shown to reduce UF size even up to 70% in just a 2-month period, with fewer side effects
than GnRH analog therapy [
22
,
118
]. In an RCT comparing the effect of letrozole and triptorelin by
Parsanezhad et al., letrozole presented better uterine volume reduction than the GnRH analog (45.6%
vs. 33.2%, respectively) [
109
]. In this study, approximately 96% of patients in the triptorelin group
reported related vasomotor symptoms, whereas in the AI group it was a rare finding [
109
]. In their
review article, Shozu et al. concluded that certain doses of AIs can fully block the estrogen production
in UF tumors, whereas ovarian production of estrogen would continue, but at reduced levels [
118
].
In this situation, UF tumors would shrink, and estrogen deficiency symptoms would be mild and
tolerable [
118
]. In the case of concomitant symptoms during AI therapy, some authors advise the use of
add-back treatment of GnRH analogs, estrogens, or progestins to prevent the increase in gonadotropin
secretion and related symptoms [119].
A Cochrane review by Song et al., failed to present significant data which would support a wider
clinical use of AIs in UF-related bleeding [
120
]. In 2013, reviewers concluded that the evidence of
AI effectiveness and safety was insufficient to allow any conclusions to be drawn. In recent years,
more studies about the role of AIs in the management of UFs have been published, and we hope that
future systematic reviews and meta-analysis will gave us more answers about the use of AI in UF
therapy [
120
]. In a study conducted by Duhan et al., a 12-week letrozole therapy reduced the mean
UF volume by 52.5% in premenopausal women with clinically symptomatic tumors [
121
]. What is
even more interesting, the side effects were mild (mostly hot flashes), and no significant effects were
observed on lipid profiles and steroid serum levels [
121
]. Similar results were obtained by Sayyah-Melli
et al. [
122
], who mixed letrozole with cabergoline. In their study, 12 weeks of treatment with letrozole
with and without cabergoline improved the size and volume of UFs without significant differences
between the study groups. Both groups were comparable for the remaining minor side effects [
122
].
In an interesting study from 2014, administration of letrozole and norethindrone acetate in patients
with large UFs decreased the mean operative time by 13 min, intraoperative blood loss by 190 mL,
and suturing time by 10 min during laparoscopic myomectomy as compared to placebo controls [
123
].
More recently, in another Italian study, the authors compared preoperative administration of triptorelin,
letrozole, and UPA [
124
]. All medications caused a significant reduction in UF tumor volume, but the
highest percentages were observed in the triptorelin and letrozole groups. The use of triptorelin
and letrozole significantly decreased the mean hysteroscopy time and absorbed fluid, whereas these
variables were insignificant in UPA group and controls [124].
Data about AI use in UF treatment are incomplete and demonstrate the need for more studies
using larger sample size, longer time periods, and different doses. These studies will further our
knowledge about the potential use of AIs in the treatment and prophylaxis of uterine leiomyomas.
As estrogenic pathways are complicated and complex, patients may respond differently. We need
to improve the efficacy or safety, perhaps by individualizing the doses and add-backs [
9
]. In recent
studies, the AIs presented themselves as potent drugs in UF management which reduced UF volume
Int. J. Mol. Sci. 2017,18, 2586 9 of 19
and improved associated symptoms. Yet, due to limited data and lack of blinded trials, the use of AIs
in prevention or early therapy of UFs remains debatable.
3.6. Cabergoline
Cabergoline is an ergot derivative and is a potent dopamine receptor agonist. Its overdose might
cause nasal congestion, syncope, or hallucinations [
125
]. Cabergoline has an inhibitory effect on the
secretion of GnRH, which may be the basis of its anti-UF effect [
114
,
126
] (Figure 1). Clinical data
regarding the use of cabergoline in UF therapy are very limited. In a study by Sayyah-Melli et al.,
0.5 mg of cabergoline per week had the same effect as 3.75 mg of diphereline (GnRH analog) per month
on the reduction of UF volume [
127
]. Additionally, cabergoline presented a safer pharmacological
profile, with fewer adverse drug reactions [
127
]. In another study, also conducted by Sayyah-Melli,
cabergoline was added as a co-drug to letrozole to evaluate its synergic effect in UF therapy [
122
].
In this study, the addition of cabergoline did not significantly change the drop in UF volume,
but caused more women to report a headache as an adverse reaction [
101
]. Finally, an Iranian
study on 51 women showed that administration of cabergoline significantly reduced UF-related
symptoms. In this study, women who received 0.5 mg of cabergoline daily for 3 months had decreased
pelvic pain, reduced bleeding, and a lower volume of UFs as compared to controls [128].
According to abovementioned data, cabergoline might find its place in the treatment of specific
groups of women with clinically symptomatic UFs, but we need more studies which would examine its
safety and efficacy [
126
]. No current RCTs about the use of cabergoline in UF management are available.
3.7. Others
We mentioned the top six current alternative agents in UF therapy. There are also other substances
which might have an impact in this field. Unfortunately, their trial data and available evidence are
limited. We will attempt to briefly explain each of these substances.
Combined oral contraception (COC) has a well-documented efficacy in case of excessive uterine
bleeding [
129
]. The bleeding often accompanies UFs and is in fact UF-dependent [
130
]. COC is a
common first-choice therapy among the gynecologists in women with such complaints. The results
of the ESHRE (European Society of Human Reproduction and Embryology) group demonstrated
that the use of COC before the age of 17 might have a weak association with UF occurrence [
129
].
According to a study by Qin et al., COC does not have a significant influence on UF tumor growth
and occurrence [
131
]. Some clinical trials have confirmed that COCs containing estradiol valerate
and dienogest are in fact effective in treating excessive bleeding. However, they investigated patients
without organic pathologies such as UFs [
132
]. Current data on the efficacy of COC in UF-therapy
are limited.
Gestrinone is a synthetic steroid with mixed progestogenic and antiprogestogenic effects,
with some androgenic and antiestrogenic activity, used in gynecology [
133
]. It has an inhibitory
effect on the pituitary gland and is comparable to danazol in its function [
134
]. According to Zhu et al.,
it inhibits growth of UFs via its antagonistic effect on the estrogen and progesterone receptors [
36
].
In various studies by Coutinho et al., administration of gestrinone reduced UF tumor volume and
uterine bleeding [
135
137
]. The side effects of gestrinone were as follows: weight gain and androgenic
features such as decreased breast size, acne, seborrhea, and hirsutism [
136
]. In light of rather limited
beneficial effects of using gestrinone in UF therapy, no RCTs comparing gestrinone and other agents or
even a placebo are currently available [114].
Curcumin is a yellow substance produced by some plants. It is used as food seasoning, cosmetic
ingredient, or herbal supplement. Curcumin is a diarylheptanoid belonging to the curcuminoid
family (natural phenols), which has anti-inflammatory, antioxidative [
138
], and anti-cancer activities,
including the inhibition of initiation, progression, invasion, and metastasis formation [
139
].
According to the available data, curcumin might be applicable in therapy but its low bioavailability
and solubility, as well as its rapid metabolism create a challenge [
139
]. A study by Shishodia et al.,
Int. J. Mol. Sci. 2017,18, 2586 10 of 19
revealed that curcumin might inhibit proliferation and fibrosis and regulate apoptosis in mantle cell
lymphoma [
90
,
140
]. In a study by Malik et al., curcumin demonstrated an inhibitory effect on UF cell
proliferation and production of ECM [
141
]. This inhibitory effect on UF cells was also described by
Tsuiji et al., who presented data that curcumin suppresses UF cell proliferation via the activation of
PPAR
γ
and decreased proteoglycan expression in ECM [
142
]. In our opinion, the available data do not
support the wider use of curcumin in UF therapy and additional evidence is needed.
SB525334 is a potent and selective inhibitor of TGF-
β
receptor I (ALK5) [
143
]. This agent has been
found to prevent pulmonary and renal fibrosis [
143
]. The TGF-
β
pathway is one of the most important
pathways in UF biology [
1
]. Some authors believe that SB525334 might be a new high-quality treatment
in UF therapy [
108
]. According to a study by Laping et al., treatment with this agent decreases the
incidence, number, and size of UF tumors in a mutant rodent model [
144
]. Due to incomplete data,
further studies are necessary to prove that SB525334 might be useful in UF therapy.
Various literature reports also demonstrated that non-steroidal anti-inflammatory drugs (NSAIDs)
can be effective in reducing heavy menstrual bleeding, due to their influence on the endometrial
cyclooxygenase levels [
114
,
145
]. These drugs were widely tested in abnormal uterine bleeding,
but there is no sufficient data to recommend them in women with UFs and other symptoms as
they do not exert any influence on fibroid volume [
114
]. Very recently, Gao et al. published their data
about the effect of acetylsalicylic acid on UF cells. Their results suggested that aspirin inhibits UF cell
growth by the regulation of K-Ras pathways [
146
]. While interesting, nothing else is known in this area.
We hope that we will receive more data about the potential usefulness of aspirin in the prophylaxis or
UF therapy very soon.
One of the last substances of note is pirfenidone, a pyridine molecule used as an antifibrotic
agent [
147
,
148
] in the treatment of pulmonary fibrosis [
148
]. Studies about pirfenidone and UFs
demonstrated that pirfenidone was an effective inhibitor of myometrial and UF cell proliferation,
and that it reduced ECM mRNA levels [
147
]. We cannot recommend it as a standard treatment due to
lack of published data.
Finally, there are several studies regarding the potential role of Ro 41-0960, a synthetic
COMT inhibitor [
149
] and tocopherol analogs in the treatment of UFs [
150
] but these agents need
more evidence.
4. Conclusions
UFs constitute a serious health problem for many women of reproductive age, as well as those
approaching, or actively in, menopause. Prophylaxis of UFs is practically non-existent, while treatment
is often costly and expensive. Surgery is standard in symptomatic UF treatment. UPA has been
available for several years. Other forms of treatment are not accepted by patients. In the case of more
effective agents, the main problem is the chronic use of drugs and the related side effects. In the case of
other agents, their poor effectiveness remains the greatest issue. In our opinion, additional solutions
are necessary to create appropriate schemes of pharmacological treatment for different groups of
women (obese or non-obese, Caucasian or African-American, pre- or post-menopausal, etc.).
Early prevention and treatment of UFs in women from high-risk groups should be our priority.
Innovative forms of UF management are under intensive investigation and may be promising options
in the near future. There are several studies about the role of the abovementioned agents in UF
prophylaxis and therapy in humans. Many of them evaluated vitamin D, paricalcitol, EGCG, elagolix,
AIs, and cabergoline and deemed them safe and effective. The next step in such projects should be
properly constructed RCTs, carried out by successive phases. However, the agents on the current
list had only one registered trial [
101
], or none at all. In the case of further positive observations,
these agents could become the new generation of drugs in the treatment of UFs, even in the era of UPA.
Perhaps the future solution will be to identify high-risk groups before the onset of UFs and
implement preventive methods on the basis of the presented literature. Vitamin D and green tea extract
might be optimal in such cases. Recent attempts to create a new, cheap, safe, and effective drug for
Int. J. Mol. Sci. 2017,18, 2586 11 of 19
the treatment of UFs remain in the very early stages, and their success has not been yet determined.
Recent findings suggest that substances such as paricalcitol and elagolix may be the formulas for the
future as they have minimal or moderate side effects and high levels of efficacy in UF therapy.
Acknowledgments:
This study was funded by The Centre of Postgraduate Medical Education.
Grant number 501-1-21-27-17.
Author Contributions:
Michał Ciebiera, Krzysztof Łukaszuk, Bła ˙
zej M˛eczekalski and G.J. designed the
review. Michał Ciebiera, Krzysztof Łukaszuk, Bła˙
zej M˛eczekalski, Magdalena Ciebiera, Cezary Wojtyła,
Aneta Słabuszewska-Jó´zwiak and Grzegorz Jakiel analyzed the data and wrote the paper. Michał Ciebiera and
Magdalena Ciebiera draw the figure. Michał Ciebiera Krzysztof Łukaszuk, Bła ˙
zej M˛eczekalski, and Grzegorz Jakiel
supervised the work and accepted the final version of the manuscript.
Conflicts of Interest:
Michał Ciebiera, Aneta Słabuszewska-Jó´zwiak and Grzegorz Jakiel have received personal
fees from Gedeon Richter not related to this work. The Gedeon Richter had no role in the design of the study;
in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish
the results.
Abbreviations
25(OH)D 25-hydroxyvitamin D
AI aromatase inhibitor
ALK5 transforming growth factor-βtype 1 receptor
BCL B-cell lymphoma
CDK cyclin-dependent kinase
COMT catechol-O-methyltransferase
ECG epicatechin gallate
EGC epigallocatechin
EGCG epigallocatechin gallate
ER estrogen receptor
GnRH gonadotropin-releasing hormone
MMP matrix metalloproteinase
mRNA messenger RNA
PCNA proliferating cell nuclear antigen
PPAR peroxisome proliferator-activated receptor
PR progesterone receptor
QoL quality of life
RCT randomized control trial
SPRM selective progesterone receptor modulator
TGF-βtransforming growth factor beta
TIMP tissue inhibitor of metalloproteinase
UF uterine fibroid
UPA ulipristal acetate
VDR vitamin D receptor
References
1. Bulun, S.E. Uterine fibroids. N. Engl. J. Med. 2013,369, 1344–1355. [CrossRef] [PubMed]
2.
Stewart, E.A.; Laughlin-Tommaso, S.K.; Catherino, W.H.; Lalitkumar, S.; Gupta, D.; Vollenhoven, B.
Uterine fibroids. Nat. Rev. Dis. Prim. 2016,2, 16043. [CrossRef] [PubMed]
3.
Al-Hendy, A.; Myers, E.R.; Stewart, E. Uterine fibroids: Burden and unmet medical need.
Semin. Reprod. Med.
2017,35, 473–480. [CrossRef] [PubMed]
4.
Parker, W.H. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil. Steril.
2007
,87, 725–736.
[CrossRef] [PubMed]
5.
Wise, L.A.; Palmer, J.R.; Stewart, E.A.; Rosenberg, L. Age-specific incidence rates for self-reported uterine
leiomyomata in the black women’s health study. Obstet. Gynecol. 2005,105, 563–568. [CrossRef] [PubMed]
6.
Donnez, J.; Dolmans, M.M. Uterine fibroid management: From the present to the future.
Hum. Reprod. Update
2016,22, 665–686. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2017,18, 2586 12 of 19
7. Stewart, E.A. Uterine fibroids. Lancet 2001,357, 293–298. [CrossRef]
8.
Baird, D.D.; Dunson, D.B.; Hill, M.C.; Cousins, D.; Schectman, J.M. High cumulative incidence of uterine
leiomyoma in black and white women: Ultrasound evidence. Am. J. Obstet. Gynecol.
2003
,188, 100–107.
[CrossRef] [PubMed]
9.
Borahay, M.A.; Asoglu, M.R.; Mas, A.; Adam, S.; Kilic, G.S.; Al-Hendy, A. Estrogen receptors and signaling
in fibroids: Role in pathobiology and therapeutic implications. Reprod. Sci.
2017
,24, 1235–1244. [CrossRef]
[PubMed]
10.
Ishikawa, H.; Ishi, K.; Serna, V.A.; Kakazu, R.; Bulun, S.E.; Kurita, T. Progesterone is essential for maintenance
and growth of uterine leiomyoma. Endocrinology 2010,151, 2433–2442. [CrossRef] [PubMed]
11.
Maruo, T.; Ohara, N.; Wang, J.; Matsuo, H. Sex steroidal regulation of uterine leiomyoma growth and
apoptosis. Hum. Reprod. Update 2004,10, 207–220. [CrossRef] [PubMed]
12.
Ciarmela, P.; Islam, M.S.; Reis, F.M.; Gray, P.C.; Bloise, E.; Petraglia, F.; Vale, W.; Castellucci, M. Growth factors
and myometrium: Biological effects in uterine fibroid and possible clinical implications.
Hum. Reprod. Update
2011,17, 772–790. [CrossRef] [PubMed]
13.
Tal, R.; Segars, J.H. The role of angiogenic factors in fibroid pathogenesis: Potential implications for future
therapy. Hum. Reprod. Update 2014,20, 194–216. [CrossRef] [PubMed]
14.
Ciebiera, M.; Wlodarczyk, M.; Slabuszewska-Jozwiak, A.; Nowicka, G.; Jakiel, G. Influence of vitamin D
and transforming growth factor
β
3 serum concentrations, obesity, and family history on the risk for uterine
fibroids. Fertil. Steril. 2016,106, 1787–1792. [CrossRef] [PubMed]
15.
Makinen, N.; Mehine, M.; Tolvanen, J.; Kaasinen, E.; Li, Y.; Lehtonen, H.J.; Gentile, M.; Yan, J.; Enge, M.;
Taipale, M.; et al. Med12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine
leiomyomas. Science 2011,334, 252–255. [CrossRef] [PubMed]
16.
Heinonen, H.R.; Sarvilinna, N.S.; Sjoberg, J.; Kampjarvi, K.; Pitkanen, E.; Vahteristo, P.; Makinen, N.;
Aaltonen, L.A. Med12 mutation frequency in unselected sporadic uterine leiomyomas. Fertil. Steril.
2014
,
102, 1137–1142. [CrossRef] [PubMed]
17.
Soave, I.; Marci, R. Uterine leiomyomata: The snowball effect. Curr. Med. Res. Opin.
2017
,33, 1909–1911.
[CrossRef] [PubMed]
18.
Soliman, A.M.; Margolis, M.K.; Castelli-Haley, J.; Fuldeore, M.J.; Owens, C.D.; Coyne, K.S. Impact of uterine
fibroid symptoms on health-related quality of life of US women: Evidence from a cross-sectional survey.
Curr. Med. Res. Opin. 2017,33, 1971–1978. [CrossRef] [PubMed]
19.
Soliman, A.M.; Yang, H.; Du, E.X.; Kelkar, S.S.; Winkel, C. The direct and indirect costs of uterine fibroid
tumors: A systematic review of the literature between 2000 and 2013. Am. J. Obstet. Gynecol.
2015
,213,
141–160. [CrossRef] [PubMed]
20.
Cardozo, E.R.; Clark, A.D.; Banks, N.K.; Henne, M.B.; Stegmann, B.J.; Segars, J.H. The estimated annual cost
of uterine leiomyomata in the United States. Am. J. Obstet. Gynecol.
2012
,206, 211.e1–211.e9. [CrossRef]
[PubMed]
21.
Lee, D.W.; Ozminkowski, R.J.; Carls, G.S.; Wang, S.; Gibson, T.B.; Stewart, E.A. The direct and indirect cost
burden of clinically significant and symptomatic uterine fibroids. J. Occup. Environ. Med.
2007
,49, 493–506.
[CrossRef] [PubMed]
22.
Faustino, F.; Martinho, M.; Reis, J.; Aguas, F. Update on medical treatment of uterine fibroids. Eur. J. Obstet.
Gynecol. Reprod. Biol. 2017,216, 61–68. [CrossRef] [PubMed]
23.
Singh, S.S.; Belland, L. Contemporary management of uterine fibroids: Focus on emerging medical treatments.
Curr. Med. Res. Opin. 2015,31, 1–12. [CrossRef] [PubMed]
24.
Stewart, E.A. Clinical practice. Uterine fibroids. N. Engl. J. Med.
2015
,372, 1646–1655. [CrossRef] [PubMed]
25.
Wu, J.L.; Segars, J.H. Is vitamin D the answer for prevention of uterine fibroids? Fertil. Steril.
2015
,104,
559–560. [CrossRef] [PubMed]
26.
Donnez, J.; Donnez, O.; Dolmans, M.M. Safety of treatment of uterine fibroids with the selective progesterone
receptor modulator, ulipristal acetate. Expert Opin. Drug Saf. 2016,15, 1679–1686. [CrossRef] [PubMed]
27.
Donnez, J.; Donnez, O.; Courtoy, G.E.; Dolmans, M.M. The place of selective progesterone receptor
modulators in myoma therapy. Minerva Ginecol. 2016,68, 313–320. [PubMed]
28.
Biglia, N.; Carinelli, S.; Maiorana, A.; D’Alonzo, M.; Lo Monte, G.; Marci, R. Ulipristal acetate: A novel
pharmacological approach for the treatment of uterine fibroids. Drug Des. Dev. Ther.
2014
,8, 285–292.
[CrossRef]
Int. J. Mol. Sci. 2017,18, 2586 13 of 19
29.
Donnez, J.; Arriagada, P.; Donnez, O.; Dolmans, M.M. Current management of myomas: The place of medical
therapy with the advent of selective progesterone receptor modulators. Curr. Opin. Obstet. Gynecol.
2015
,
27, 422–431. [CrossRef] [PubMed]
30.
Commandeur, A.E.; Styer, A.K.; Teixeira, J.M. Epidemiological and genetic clues for molecular mechanisms
involved in uterine leiomyoma development and growth. Hum. Reprod. Update
2015
,21, 593–615. [CrossRef]
[PubMed]
31.
Sabry, M.; Al-Hendy, A. Innovative oral treatments of uterine leiomyoma. Obstet. Gynecol. Int.
2012
,
2012, 943635. [CrossRef] [PubMed]
32.
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.
2010,94, 1887–1893. [CrossRef] [PubMed]
33.
Brakta, S.; Diamond, J.S.; Al-Hendy, A.; Diamond, M.P.; Halder, S.K. Role of vitamin D in uterine fibroid
biology. Fertil. Steril. 2015,104, 698–706. [CrossRef] [PubMed]
34.
Archer, D.F.; Stewart, E.A.; Jain, R.I.; Feldman, R.A.; Lukes, A.S.; North, J.D.; Soliman, A.M.; Gao, J.; Ng, J.W.;
Chwalisz, K. Elagolix for the management of heavy menstrual bleeding associated with uterine fibroids:
Results from a phase 2a proof-of-concept study. Fertil. Steril. 2017,108, 152–160. [CrossRef] [PubMed]
35.
Halder, S.K.; Sharan, C.; Al-Hendy, O.; Al-Hendy, A. Paricalcitol, a vitamin D receptor activator,
inhibits tumor formation in a murine model of uterine fibroids. Reprod. Sci.
2014
,21, 1108–1119. [CrossRef]
[PubMed]
36.
Zhu, Y.; Zhang, T.; Xie, S.; Tu, R.; Cao, Y.; Guo, X.; Zhou, J.; Zhou, X.; Cao, L. Gestrinone inhibits growth of
human uterine leiomyoma may relate to activity regulation of ER
α
, Src and P38 MAPK. Biomed. Pharmacother.
2012,66, 569–577. [CrossRef] [PubMed]
37.
Halder, S.; Al-Hendy, A. Hypovitaminosis D and high serum transforming growth factor
β
-3:
Important biomarkers for uterine fibroids risk. Fertil. Steril. 2016,106, 1648–1649. [CrossRef] [PubMed]
38.
Holick, M.F. The vitamin D deficiency pandemic: Approaches for diagnosis, treatment and prevention.
Rev. Endocr. Metab. Disord. 2017,18, 153–165. [CrossRef] [PubMed]
39.
Holick, M.F. Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers,
and cardiovascular disease. Am. J. Clin. Nutr. 2004,80, 1678S–1688S. [PubMed]
40.
Parazzini, F.; Di Martino, M.; Candiani, M.; Vigano, P. Dietary components and uterine leiomyomas: A review
of published data. Nutr. Cancer 2015,67, 569–579. [CrossRef] [PubMed]
41. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007,357, 266–281. [CrossRef] [PubMed]
42.
Nair, R.; Maseeh, A. Vitamin D: The “sunshine” vitamin. J. Pharmacol. Pharmacother.
2012
,3, 118–126.
[CrossRef] [PubMed]
43.
Bikle, D.D. Vitamin D and immune function: Understanding common pathways. Curr. Osteoporos. Rep.
2009
,
7, 58–63. [CrossRef] [PubMed]
44.
Gonzalez-Mateo, G.T.; Fernandez-Millara, V.; Bellon, T.; Liappas, G.; Ruiz-Ortega, M.; Lopez-Cabrera, M.;
Selgas, R.; Aroeira, L.S. Paricalcitol reduces peritoneal fibrosis in mice through the activation of regulatory T
cells and reduction in IL-17 production. PLoS ONE 2014,9, e108477. [CrossRef] [PubMed]
45.
Protic, O.; Toti, P.; Islam, M.S.; Occhini, R.; Giannubilo, S.R.; Catherino, W.H.; Cinti, S.; Petraglia, F.;
Ciavattini, A.; Castellucci, M.; et al. Possible involvement of inflammatory/reparative processes in the
development of uterine fibroids. Cell Tissue Res. 2016,364, 415–427. [CrossRef] [PubMed]
46.
Paffoni, A.; Somigliana, E.; Vigano, P.; Benaglia, L.; Cardellicchio, L.; Pagliardini, L.; Papaleo, E.; Candiani, M.;
Fedele, L. Vitamin D status in women with uterine leiomyomas. J. Clin. Endocrinol. Metab.
2013
,98,
E1374–E1378. [CrossRef] [PubMed]
47.
Lerchbaum, E.; Rabe, T. Vitamin D and female fertility. Curr. Opin. Obstet. Gynecol.
2014
,26, 145–150.
[CrossRef] [PubMed]
48.
Oskovi Kaplan, Z.A.; Tasci, Y.; Topcu, H.O.; Erkaya, S. 25-Hydroxy vitamin D levels in premenopausal
turkish women with uterine leiomyoma. Gynecol. Endocrinol. 2017, 1–4. [CrossRef] [PubMed]
49.
Zhao, G.; Ford, E.S.; Tsai, J.; Li, C.; Croft, J.B. Factors associated with vitamin D deficiency and inadequacy
among women of childbearing age in the United States. ISRN Obstet. Gynecol.
2012
,2012, 691486. [CrossRef]
[PubMed]
Int. J. Mol. Sci. 2017,18, 2586 14 of 19
50.
Sabry, M.; Halder, S.K.; Allah, A.S.; Roshdy, E.; Rajaratnam, V.; Al-Hendy, A. Serum vitamin D3 level
inversely correlates with uterine fibroid volume in different ethnic groups: A cross-sectional observational
study. Int. J. Womens Health 2013,5, 93–100. [CrossRef] [PubMed]
51.
Baird, D.D.; Hill, M.C.; Schectman, J.M.; Hollis, B.W. Vitamin D and the risk of uterine fibroids. Epidemiology
2013,24, 447–453. [CrossRef] [PubMed]
52.
Blauer, M.; Rovio, P.H.; Ylikomi, T.; Heinonen, P.K. Vitamin D inhibits myometrial and leiomyoma cell
proliferation in vitro. Fertil. Steril. 2009,91, 1919–1925. [CrossRef] [PubMed]
53.
Sharan, C.; Halder, S.K.; Thota, C.; Jaleel, T.; Nair, S.; Al-Hendy, A. Vitamin D inhibits proliferation of
human uterine leiomyoma cells via catechol-O-methyltransferase. Fertil. Steril.
2011
,95, 247–253. [CrossRef]
[PubMed]
54.
Halder, S.K.; Goodwin, J.S.; Al-Hendy, A. 1,25-Dihydroxyvitamin D3 reduces TGF-
β
3-induced
fibrosis-related gene expression in human uterine leiomyoma cells. J. Clin. Endocrinol. Metab.
2011
,
96, E754–E762. [CrossRef] [PubMed]
55.
Halder, S.K.; Sharan, C.; Al-Hendy, A. 1,25-Dihydroxyvitamin D3 treatment shrinks uterine leiomyoma
tumors in the eker rat model. Biol. Reprod. 2012,86, 116. [CrossRef] [PubMed]
56.
Halder, S.K.; Osteen, K.G.; Al-Hendy, A. 1,25-Dihydroxyvitamin D3 reduces extracellular matrix-associated
protein expression in human uterine fibroid cells. Biol. Reprod. 2013,89, 150. [CrossRef] [PubMed]
57.
Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.;
Weaver, C.M.; Endocrine, S. Evaluation, treatment, and prevention of vitamin D deficiency: An endocrine
society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011,96, 1911–1930. [CrossRef] [PubMed]
58.
Pludowski, P.; Holick, M.F.; Grant, W.B.; Konstantynowicz, J.; Mascarenhas, M.R.; Haq, A.; Povoroznyuk, V.;
Balatska, N.; Barbosa, A.P.; Karonova, T.; et al. Vitamin D supplementation guidelines. J. Steroid Biochem.
Mol. Biol. 2017. [CrossRef] [PubMed]
59.
Pludowski, P.; Holick, M.F.; Pilz, S.; Wagner, C.L.; Hollis, B.W.; Grant, W.B.; Shoenfeld, Y.; Lerchbaum, E.;
Llewellyn, D.J.; Kienreich, K.; et al. Vitamin D effects on musculoskeletal health, immunity, autoimmunity,
cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality—A review of recent evidence.
Autoimmun. Rev. 2013,12, 976–989. [CrossRef] [PubMed]
60.
Pludowski, P.; Karczmarewicz, E.; Bayer, M.; Carter, G.; Chlebna-Sokol, D.; Czech-Kowalska, J.; Debski, R.;
Decsi, T.; Dobrzanska, A.; Franek, E.; et al. Practical guidelines for the supplementation of vitamin D and
the treatment of deficits in central europe-recommended vitamin D intakes in the general population and
groups at risk of vitamin D deficiency. Endokrynol. Pol. 2013,64, 319–327. [CrossRef] [PubMed]
61.
Al-Baaj, F.; Yadav, P.; Al-Rifai, A. Paricalcitol in secondary hyperparathyroidism and the survival benefit in
patients with chronic kidney disease. J. Ren. Care 2011,37, 75–79. [CrossRef] [PubMed]
62.
Bouillon, R.; Verstuyf, A.; Verlinden, L.; Allewaert, K.; Branisteanu, D.; Mathieu, C.; van Baelen, H.
Non-hypercalcemic pharmacological aspects of vitamin D analogs. Biochem. Pharmacol.
1995
,50, 577–583.
[CrossRef]
63.
EMC. Zemplar Soft Capsules 1 mcg. Available online: https://www.Medicines.Org.Uk/emc/medicine/
20464 (accessed on 16 November 2017).
64.
Zhang, Y.; Kong, J.; Deb, D.K.; Chang, A.; Li, Y.C. Vitamin D receptor attenuates renal fibrosis by suppressing
the renin-angiotensin system. J. Am. Soc. Nephrol. 2010,21, 966–973. [CrossRef] [PubMed]
65.
Stavenuiter, A.W.; Farhat, K.; Vila Cuenca, M.; Schilte, M.N.; Keuning, E.D.; Paauw, N.J.; ter Wee, P.M.;
Beelen, R.H.; Vervloet, M.G. Protective effects of paricalcitol on peritoneal remodeling during peritoneal
dialysis. Biomed. Res. Int. 2015,2015, 468574. [CrossRef] [PubMed]
66.
Oblak, M.; Mlinsek, G.; Kandus, A.; Buturovic-Ponikvar, J.; Arnol, M. Effects of paricalcitol on biomarkers
of inflammation and fibrosis in kidney transplant recipients: Results of a randomized controlled trial.
Clin. Nephrol. 2017,88, 119–125. [CrossRef] [PubMed]
67.
Freundlich, M.; Quiroz, Y.; Zhang, Z.; Zhang, Y.; Bravo, Y.; Weisinger, J.R.; Li, Y.C.; Rodriguez-Iturbe, B.
Suppression of renin-angiotensin gene expression in the kidney by paricalcitol. Kidney Int.
2008
,74,
1394–1402. [CrossRef] [PubMed]
68.
Arici, A.; Sozen, I. Transforming growth factor-
β
3 is expressed at high levels in leiomyoma where it stimulates
fibronectin expression and cell proliferation. Fertil. Steril. 2000,73, 1006–1011. [CrossRef]
Int. J. Mol. Sci. 2017,18, 2586 15 of 19
69.
Lee, B.S.; Nowak, R.A. Human leiomyoma smooth muscle cells show increased expression of transforming
growth factor-
β
3 (TGF
β
3) and altered responses to the antiproliferative effects of TGF
β
.J. Clin.
Endocrinol. Metab. 2001,86, 913–920. [CrossRef] [PubMed]
70.
Leppert, P.C.; Baginski, T.; Prupas, C.; Catherino, W.H.; Pletcher, S.; Segars, J.H. Comparative ultrastructure of
collagen fibrils in uterine leiomyomas and normal myometrium. Fertil. Steril.
2004
,82, 1182–1187. [CrossRef]
[PubMed]
71.
Sozen, I.; Arici, A. Interactions of cytokines, growth factors, and the extracellular matrix in the cellular
biology of uterine leiomyomata. Fertil. Steril. 2002,78, 1–12. [CrossRef]
72.
Dou, Q.; Tarnuzzer, R.W.; Williams, R.S.; Schultz, G.S.; Chegini, N. Differential expression of matrix
metalloproteinases and their tissue inhibitors in leiomyomata: A mechanism for gonadotrophin releasing
hormone agonist-induced tumour regression. Mol. Hum. Reprod. 1997,3, 1005–1014. [CrossRef] [PubMed]
73.
Brew, K.; Dinakarpandian, D.; Nagase, H. Tissue inhibitors of metalloproteinases: Evolution, structure and
function. Biochim. Biophys. Acta 2000,1477, 267–283. [CrossRef]
74.
Beresniak, A.; Duru, G.; Berger, G.; Bremond-Gignac, D. Relationships between black tea consumption and
key health indicators in the world: An ecological study. BMJ Open 2012,2. [CrossRef] [PubMed]
75.
Khan, N.; Mukhtar, H. Tea and health: Studies in humans. Curr. Pharm. Des.
2013
,19, 6141–6147. [CrossRef]
[PubMed]
76.
Forester, S.C.; Lambert, J.D. The role of antioxidant versus pro-oxidant effects of green tea polyphenols in
cancer prevention. Mol. Nutr. Food Res. 2011,55, 844–854. [CrossRef] [PubMed]
77.
Kuriyama, S.; Shimazu, T.; Ohmori, K.; Kikuchi, N.; Nakaya, N.; Nishino, Y.; Tsubono, Y.; Tsuji, I. Green tea
consumption and mortality due to cardiovascular disease, cancer, and all causes in Japan: The ohsaki study.
JAMA 2006,296, 1255–1265. [CrossRef] [PubMed]
78.
Gupta, S.; Ahmad, N.; Nieminen, A.L.; Mukhtar, H. Growth inhibition, cell-cycle dysregulation,
and induction of apoptosis by green tea constituent (
)-epigallocatechin-3-gallate in androgen-sensitive and
androgen-insensitive human prostate carcinoma cells. Toxicol. Appl. Pharmacol.
2000
,164, 82–90. [CrossRef]
[PubMed]
79.
Tang, Y.; Zhao, D.Y.; Elliott, S.; Zhao, W.; Curiel, T.J.; Beckman, B.S.; Burow, M.E. Epigallocatechin-3
gallate induces growth inhibition and apoptosis in human breast cancer cells through survivin suppression.
Int. J. Oncol. 2007,31, 705–711. [CrossRef] [PubMed]
80.
Javaid, M.S.; Latief, N.; Ijaz, B.; Ashfaq, U.A. Epigallocatechin gallate as an anti-obesity therapeutic
compound: An in silico approach for structure-based drug designing. Nat. Prod. Res.
2017
, 1–5. [CrossRef]
[PubMed]
81.
Xiao, J.; Ho, C.T.; Liong, E.C.; Nanji, A.A.; Leung, T.M.; Lau, T.Y.; Fung, M.L.; Tipoe, G.L. Epigallocatechin
gallate attenuates fibrosis, oxidative stress, and inflammation in non-alcoholic fatty liver disease rat model
through TGF/SMAD, PI3 K/Akt/FoxO1, and NF-
κ
B pathways. Eur. J. Nutr.
2014
,53, 187–199. [CrossRef]
[PubMed]
82.
Sriram, N.; Kalayarasan, S.; Manikandan, R.; Arumugam, M.; Sudhandiran, G. Epigallocatechin gallate
attenuates fibroblast proliferation and excessive collagen production by effectively intervening TGF-
β
1
signalling. Clin. Exp. Pharmacol. Physiol. 2015,42, 849–859. [CrossRef] [PubMed]
83.
Matsuzaki, S.; Darcha, C. Antifibrotic properties of epigallocatechin-3-gallate in endometriosis. Hum. Reprod.
2014,29, 1677–1687. [CrossRef] [PubMed]
84.
Yang, C.S.; Lambert, J.D.; Hou, Z.; Ju, J.; Lu, G.; Hao, X. Molecular targets for the cancer preventive activity
of tea polyphenols. Mol. Carcinog. 2006,45, 431–435. [CrossRef] [PubMed]
85.
Kanwar, J.; Taskeen, M.; Mohammad, I.; Huo, C.; Chan, T.H.; Dou, Q.P. Recent advances on tea polyphenols.
Front. Biosci. 2012,4, 111–131. [CrossRef]
86.
Chen, D.; Wan, S.B.; Yang, H.; Yuan, J.; Chan, T.H.; Dou, Q.P. Egcg, green tea polyphenols and their synthetic
analogs and prodrugs for human cancer prevention and treatment. Adv. Clin. Chem.
2011
,53, 155–177.
[PubMed]
87.
Ahmed, R.S.; Liu, G.; Renzetti, A.; Farshi, P.; Yang, H.; Soave, C.; Saed, G.; El-Ghoneimy, A.A.; El-Banna, H.A.;
Foldes, R.; et al. Biological and mechanistic characterization of novel prodrugs of green tea polyphenol
epigallocatechin gallate analogs in human leiomyoma cell lines. J. Cell. Biochem.
2016
,117, 2357–2369.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2017,18, 2586 16 of 19
88.
Roshdy, E.; Rajaratnam, V.; Maitra, S.; Sabry, M.; Allah, A.S.; Al-Hendy, A. Treatment of symptomatic uterine
fibroids with green tea extract: A pilot randomized controlled clinical study. Int. J. Womens Health
2013
,5,
477–486. [CrossRef] [PubMed]
89.
Zhang, D.; Al-Hendy, M.; Richard-Davis, G.; Montgomery-Rice, V.; Sharan, C.; Rajaratnam, V.; Khurana, A.;
Al-Hendy, A. Green tea extract inhibits proliferation of uterine leiomyoma cells
in vitro
and in nude mice.
Am. J. Obstet. Gynecol. 2010,202, 289.e1–289.e9. [CrossRef] [PubMed]
90.
Bartels, C.B.; Cayton, K.C.; Chuong, F.S.; Holthouser, K.; Arian, S.E.; Abraham, T.; Segars, J.H.
An evidence-based approach to the medical management of fibroids: A systematic review.
Clin. Obstet. Gynecol. 2016,59, 30–52. [CrossRef] [PubMed]
91.
Ramachandran, B.; Jayavelu, S.; Murhekar, K.; Rajkumar, T. Repeated dose studies with pure
epigallocatechin-3-gallate demonstrated dose and route dependant hepatotoxicity with associated
dyslipidemia. Toxicol. Rep. 2016,3, 336–345. [CrossRef] [PubMed]
92.
Maggi, R.; Cariboni, A.M.; Marelli, M.M.; Moretti, R.M.; Andre, V.; Marzagalli, M.; Limonta, P. GnRH and
GnRH receptors in the pathophysiology of the human female reproductive system. Hum. Reprod. Update
2016,22. [CrossRef] [PubMed]
93.
Ng, J.; Chwalisz, K.; Carter, D.C.; Klein, C.E. Dose-dependent suppression of gonadotropins and ovarian
hormones by elagolix in healthy premenopausal women. J. Clin. Endocrinol. Metab.
2017
,102, 1683–1691.
[CrossRef] [PubMed]
94.
Chen, C.; Wu, D.; Guo, Z.; Xie, Q.; Reinhart, G.J.; Madan, A.; Wen, J.; Chen, T.; Huang, C.Q.; Chen, M.; et al.
Discovery of sodium R-(+)-4-{2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-[trifluoromethyl]benzyl)-
4-methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino}butyrate (elagolix), a potent and
orally available nonpeptide antagonist of the human gonadotropin-releasing hormone receptor. J. Med. Chem.
2008,51, 7478–7485. [CrossRef] [PubMed]
95.
Ezzati, M.; Carr, B.R. Elagolix, a novel, orally bioavailable GnRH antagonist under investigation for the
treatment of endometriosis-related pain. Womens Health 2015,11, 19–28. [CrossRef] [PubMed]
96.
Nakata, D.; Masaki, T.; Tanaka, A.; Yoshimatsu, M.; Akinaga, Y.; Asada, M.; Sasada, R.; Takeyama, M.;
Miwa, K.; Watanabe, T.; et al. Suppression of the hypothalamic-pituitary-gonadal axis by TAK-385 (relugolix),
a novel, investigational, orally active, small molecule gonadotropin-releasing hormone (GnRH) antagonist:
Studies in human gnrh receptor knock-in mice. Eur. J. Pharmacol. 2014,723, 167–174. [CrossRef] [PubMed]
97.
ObsEva. OBE2109—Uterine Fibroids. Available online: http://www.Obseva.Com/pipeline/obe2109
(accessed on 16 November 2017).
98.
Struthers, R.S.; Nicholls, A.J.; Grundy, J.; Chen, T.; Jimenez, R.; Yen, S.S.; Bozigian, H.P. Suppression of
gonadotropins and estradiol in premenopausal women by oral administration of the nonpeptide
gonadotropin-releasing hormone antagonist elagolix. J. Clin. Endocrinol. Metab.
2009
,94, 545–551. [CrossRef]
[PubMed]
99.
Taylor, H.S.; Giudice, L.C.; Lessey, B.A.; Abrao, M.S.; Kotarski, J.; Archer, D.F.; Diamond, M.P.; Surrey, E.;
Johnson, N.P.; Watts, N.B.; et al. Treatment of endometriosis-associated pain with elagolix, an oral GnRH
antagonist. N. Engl. J. Med. 2017,377, 28–40. [CrossRef] [PubMed]
100.
Perricos, A.; Wenzl, R. Efficacy of elagolix in the treatment of endometriosis. Expert Opin. Pharmacother.
2017
,
18, 1391–1397. [CrossRef] [PubMed]
101.
Abbvie. AbbVie Presents Pivotal Phase 3 Data on Investigational Treatment Elagolix at the World
Congress on Endometriosis. Available online: https://news.Abbvie.Com/news/abbvie-presents-pivotal-
phase-3-data-on-investigational-treatment-elagolix-at-world-congress-on- endometriosis.Htm (accessed on
16 November 2017).
102.
Shozu, M.; Murakami, K.; Inoue, M. Aromatase and leiomyoma of the uterus. Semin. Reprod. Med.
2004
,
22, 51–60. [CrossRef] [PubMed]
103.
Czajka-Oraniec, I.; Simpson, E.R. Aromatase research and its clinical significance. Endokrynol. Pol.
2010
,
61, 126–134. [PubMed]
104.
American College of Obstetricians and Gynecologists’ Committee on Gynecologic Practice. Committee
opinion No. 663: Aromatase inhibitors in gynecologic practice. Obstet. Gynecol.
2016
,127, e170–e174.
[CrossRef]
105.
Attar, E.; Bulun, S.E. Aromatase inhibitors: The next generation of therapeutics for endometriosis?
Fertil. Steril. 2006,85, 1307–1318. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2017,18, 2586 17 of 19
106.
Slopien, R.; Meczekalski, B. Aromatase inhibitors in the treatment of endometriosis. Prz. Menopauzalny
2016
,
15, 43–47. [CrossRef] [PubMed]
107.
Diamond, M.P.; Legro, R.S.; Coutifaris, C.; Alvero, R.; Robinson, R.D.; Casson, P.; Christman, G.M.; Ager, J.;
Huang, H.; Hansen, K.R.; et al. Letrozole, gonadotropin, or clomiphene for unexplained infertility. N. Engl.
J. Med. 2015,373, 1230–1240. [CrossRef] [PubMed]
108.
Islam, M.S.; Protic, O.; Giannubilo, S.R.; Toti, P.; Tranquilli, A.L.; Petraglia, F.; Castellucci, M.; Ciarmela, P.
Uterine leiomyoma: Available medical treatments and new possible therapeutic options. J. Clin.
Endocrinol. Metab. 2013,98, 921–934. [CrossRef] [PubMed]
109.
Parsanezhad, M.E.; Azmoon, M.; Alborzi, S.; Rajaeefard, A.; Zarei, A.; Kazerooni, T.; Frank, V.; Schmidt, E.H.
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, 192–198. [CrossRef] [PubMed]
110.
Giannubilo, S.R.; Ciavattini, A.; Petraglia, F.; Castellucci, M.; Ciarmela, P. Management of fibroids in
perimenopausal women. Curr. Opin. Obstet. Gynecol. 2015,27, 416–421. [CrossRef] [PubMed]
111.
Davis, S.R.; Lambrinoudaki, I.; Lumsden, M.; Mishra, G.D.; Pal, L.; Rees, M.; Santoro, N.; Simoncini, T.
Menopause. Nat. Rev. Dis. Prim. 2015,1, 15004. [CrossRef] [PubMed]
112.
Sumitani, H.; Shozu, M.; Segawa, T.; Murakami, K.; Yang, H.J.; Shimada, K.; Inoue, M. In situ estrogen
synthesized by aromatase P450 in uterine leiomyoma cells promotes cell growth probably via an
autocrine/intracrine mechanism. Endocrinology 2000,141, 3852–3861. [CrossRef] [PubMed]
113.
Ishikawa, H.; Reierstad, S.; Demura, M.; Rademaker, A.W.; Kasai, T.; Inoue, M.; Usui, H.; Shozu, M.;
Bulun, S.E. High aromatase expression in uterine leiomyoma tissues of african-american women. J. Clin.
Endocrinol. Metab. 2009,94, 1752–1756. [CrossRef] [PubMed]
114.
Kashani, B.N.; Centini, G.; Morelli, S.S.; Weiss, G.; Petraglia, F. Role of medical management for uterine
leiomyomas. Best Pract. Res. Clin. Obstet. Gynaecol. 2016,34, 85–103. [CrossRef] [PubMed]
115.
Kaunitz, A.M. Aromatase inhibitor therapy for uterine bleeding in a postmenopausal woman with
leiomyomata. Menopause 2007,14, 941–943. [CrossRef] [PubMed]
116.
Hilario, S.G.; Bozzini, N.; Borsari, R.; Baracat, E.C. Action of aromatase inhibitor for treatment of uterine
leiomyoma in perimenopausal patients. Fertil. Steril. 2009,91, 240–243. [CrossRef] [PubMed]
117.
Varelas, F.K.; Papanicolaou, A.N.; Vavatsi-Christaki, N.; Makedos, G.A.; Vlassis, G.D. The effect of anastrazole
on symptomatic uterine leiomyomata. Obstet. Gynecol. 2007,110, 643–649. [CrossRef] [PubMed]
118.
Shozu, M.; Murakami, K.; Segawa, T.; Kasai, T.; Inoue, M. Successful treatment of a symptomatic uterine
leiomyoma in a perimenopausal woman with a nonsteroidal aromatase inhibitor. Fertil. Steril.
2003
,79,
628–631. [CrossRef]
119.
Doherty, L.; Mutlu, L.; Sinclair, D.; Taylor, H. Uterine fibroids: Clinical manifestations and contemporary
management. Reprod. Sci. 2014,21, 1067–1092. [CrossRef] [PubMed]
120.
Song, H.; Lu, D.; Navaratnam, K.; Shi, G. Aromatase inhibitors for uterine fibroids. Cochrane Database
Syst. Rev. 2013, CD009505. [CrossRef] [PubMed]
121.
Duhan, N.; Madaan, S.; Sen, J. Role of the aromatase inhibitor letrozole in the management of uterine
leiomyomas in premenopausal women. Eur. J. Obstet. Gynecol. Reprod. Biol.
2013
,171, 329–332. [CrossRef]
[PubMed]
122.
Sayyah-Melli, M.; Mobasseri, M.; Gharabaghi, P.M.; Ouladsahebmadarek, E.; Rahmani, V. Comparing the
effect of aromatase inhibitor (letrozole) + cabergoline (dostinex) and letrozole alone on uterine myoma
regression, a randomized clinical trial. Eur. J. Obstet. Gynecol. Reprod. Biol.
2017
,210, 257–264. [CrossRef]
[PubMed]
123.
Leone Roberti Maggiore, U.; Scala, C.; Venturini, P.L.; Ferrero, S. Preoperative treatment with letrozole in
patients undergoing laparoscopic myomectomy of large uterine myomas: A prospective non-randomized
study. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014,181, 157–162. [CrossRef] [PubMed]
124. Bizzarri, N.; Ghirardi, V.; Remorgida, V.; Venturini, P.L.; Ferrero, S. Three-month treatment with triptorelin,
letrozole and ulipristal acetate before hysteroscopic resection of uterine myomas: Prospective comparative
pilot study. Eur. J. Obstet. Gynecol. Reprod. Biol. 2015,192, 22–26. [CrossRef] [PubMed]
125.
Rains, C.P.; Bryson, H.M.; Fitton, A. Cabergoline. A review of its pharmacological properties and therapeutic
potential in the treatment of hyperprolactinaemia and inhibition of lactation. Drugs
1995
,49, 255–279.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2017,18, 2586 18 of 19
126.
Sankaran, S.; Manyonda, I.T. Medical management of fibroids. Best Pract. Res. Clin. Obstet. Gynaecol.
2008
,
22, 655–676. [CrossRef] [PubMed]
127.
Melli, M.S.; Farzadi, L.; Madarek, E.O. Comparison of the effect of gonadotropin-releasing hormone analog
(diphereline) and cabergoline (dostinex) treatment on uterine myoma regression. Saudi Med. J.
2007
,
3, 445–450.
128.
Vahdat, M.; Kashanian, M.; Ghaziani, N.; Sheikhansari, N. Evaluation of the effects of cabergoline (dostinex)
on women with symptomatic myomatous uterus: A randomized trial. Eur. J. Obstet. Gynecol. Reprod. Biol.
2016,206, 74–78. [CrossRef] [PubMed]
129.
ESHRE Capri Workshop Group. Noncontraceptive health benefits of combined oral contraception.
Hum. Reprod. Update 2005,11, 513–525. [CrossRef]
130.
Munro, M.G.; Critchley, H.O.; Fraser, I.S.; FIGO Menstrual Disorders Working Group. The figo classification
of causes of abnormal uterine bleeding in the reproductive years. Fertil. Steril.
2011
,95, 2204–2208. [CrossRef]
[PubMed]
131.
Qin, J.; Yang, T.; Kong, F.; Zhou, Q. Oral contraceptive use and uterine leiomyoma risk: A meta-analysis
based on cohort and case-control studies. Arch. Gynecol. Obstet. 2013,288, 139–148. [CrossRef] [PubMed]
132.
Fraser, I.S.; Romer, T.; Parke, S.; Zeun, S.; Mellinger, U.; Machlitt, A.; Jensen, J.T. Effective treatment of heavy
and/or prolonged menstrual bleeding with an oral contraceptive containing estradiol valerate and dienogest:
A randomized, double-blind phase III trial. Hum. Reprod. 2011,26, 2698–2708. [CrossRef] [PubMed]
133.
Bromham, D.R.; Booker, M.W.; Rose, G.L.; Wardle, P.G.; Newton, J.R. A multicentre comparative study of
gestrinone and danazol in the treatment of endometriosis. J. Obstet. Gynaecol.
1995
,15, 188–194. [CrossRef]
134.
Selak, V.; Farquhar, C.; Prentice, A.; Singla, A. Danazol for pelvic pain associated with endometriosis.
Cochrane Database Syst. Rev. 2007, CD000068. [CrossRef]
135.
Coutinho, E.M. Gestrinone in the treatment of myomas. Acta Obstet. Gynecol. Scand. Suppl.
1989
,150, 39–46.
[CrossRef] [PubMed]
136.
Coutinho, E.M. Treatment of large fibroids with high doses of gestrinone. Gynecol. Obstet. Investig.
1990
,
30, 44–47. [CrossRef]
137.
Coutinho, E.M.; Goncalves, M.T. Long-term treatment of leiomyomas with gestrinone. Fertil. Steril.
1989
,
51, 939–946. [CrossRef]
138.
Aggarwal, B.B.; Kumar, A.; Bharti, A.C. Anticancer potential of curcumin: Preclinical and clinical studies.
Anticancer Res. 2003,23, 363–398. [PubMed]
139.
Feng, T.; Wei, Y.; Lee, R.J.; Zhao, L. Liposomal curcumin and its application in cancer. Int. J. Nanomed.
2017
,
12, 6027–6044. [CrossRef] [PubMed]
140.
Shishodia, S.; Amin, H.M.; Lai, R.; Aggarwal, B.B. Curcumin (diferuloylmethane) inhibits constitutive NF-
κ
B
activation, induces G1/S arrest, suppresses proliferation, and induces apoptosis in mantle cell lymphoma.
Biochem. Pharmacol. 2005,70, 700–713. [CrossRef] [PubMed]
141.
Malik, M.; Mendoza, M.; Payson, M.; Catherino, W.H. Curcumin, a nutritional supplement with
antineoplastic activity, enhances leiomyoma cell apoptosis and decreases fibronectin expression. Fertil. Steril.
2009,91, 2177–2184. [CrossRef] [PubMed]
142.
Tsuiji, K.; Takeda, T.; Li, B.; Wakabayashi, A.; Kondo, A.; Kimura, T.; Yaegashi, N. Inhibitory effect of
curcumin on uterine leiomyoma cell proliferation. Gynecol. Endocrinol.
2011
,27, 512–517. [CrossRef]
[PubMed]
143.
Grygielko, E.T.; Martin, W.M.; Tweed, C.; Thornton, P.; Harling, J.; Brooks, D.P.; Laping, N.J. Inhibition of
gene markers of fibrosis with a novel inhibitor of transforming growth factor-
β
type I receptor kinase in
puromycin-induced nephritis. J. Pharmacol. Exp. Ther. 2005,313, 943–951. [CrossRef] [PubMed]
144.
Laping, N.J.; Everitt, J.I.; Frazier, K.S.; Burgert, M.; Portis, M.J.; Cadacio, C.; Gold, L.I.; Walker, C.L.
Tumor-specific efficacy of transforming growth factor-
β
RI inhibition in Eker rats. Clin. Cancer Res.
2007
,13,
3087–3099. [CrossRef] [PubMed]
145.
Chwalisz, K.; Taylor, H. Current and emerging medical treatments for uterine fibroids. Semin. Reprod. Med.
2017,35, 510–522. [CrossRef] [PubMed]
146.
Gao, M.; Guo, K.M.; Wei, Y.M.; Ma, M.M.; Cai, J.R.; Xia, T.T.; Ye, Q.J. Aspirin inhibits the proliferation
of human uterine leiomyoma cells by downregulation of k-Ras-p110
α
interaction. Oncol. Rep.
2017
,38,
2507–2517. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2017,18, 2586 19 of 19
147.
Lee, B.S.; Margolin, S.B.; Nowak, R.A. Pirfenidone: A novel pharmacological agent that inhibits leiomyoma
cell proliferation and collagen production. J. Clin. Endocrinol. Metab.
1998
,83, 219–223. [CrossRef] [PubMed]
148.
Hayton, C.; Chaudhuri, N. Managing idiopathic pulmonary fibrosis: Which drug for which patient?
Drugs Aging 2017. [CrossRef] [PubMed]
149.
Hassan, M.H.; Fouad, H.; Bahashwan, S.; Al-Hendy, A. Towards non-surgical therapy for uterine fibroids:
Catechol-O-methyl transferase inhibitor shrinks uterine fibroid lesions in the Eker rat model. Hum. Reprod.
2011,26, 3008–3018. [CrossRef] [PubMed]
150.
Young, M.V.; Schwartz, G.G.; Wang, L.; Jamieson, D.P.; Whitlatch, L.W.; Flanagan, J.N.; Lokeshwar, B.L.;
Holick, M.F.; Chen, T.C. The prostate 25-hydroxyvitamin D-1
α
-hydroxylase is not influenced by parathyroid
hormone and calcium: Implications for prostate cancer chemoprevention by vitamin D. Carcinogenesis
2004
,
25, 967–971. [CrossRef] [PubMed]
©
2017 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Gestrinone, a synthetic steroid with anti-progestin and anti-estrogenic properties, has been studied for its effects on fibroid volume and overall uterine size, although data are limited. Its usage is associated with adverse effects such as acne, seborrhea, hirsutism, hot flushes, weight gain, breast size reduction, and headaches [28,29]. In meta-analysis of de Souza Pinto et al on endometriosis treatment, gestrinone showed a favorable safety profile; however, the evidence quality was low [30]. ...
... This study was not chosen because its population did not represent that of Ms. Borgias because the patients were asymptomatic, and the custom symptomology test may have been a confounding variable. Ciebiera M et al. 4 was an excellent review article that summarized the current medications and alternative therapies being explored for uterine fibroids, but it was not chosen because it was not the most current research (published in 2017) and because it did not offer specific treatment regimens unlike the chosen article by Tinelli A et al. 1 Included in the articles concerning traditional Western medicine was the 12-month trial by Neri M et al. 5 that examined the use of ulipristal acetate (UPA) in women with heavy menstrual bleeding. The benefits of the article were both the population and its length. ...
Article
Full-text available
A clinical decision report using: Tinelli A, Gustapane S, D’Oria O, Licchelli M, Panese G. Nutraceuticals in fibroid management after ulipristal acetate administration: An observational study on patients’ compliance. International Journal of Gynecology and Obstetrics; 2022;156(1):133-138. https://doi.org/10.1002/ijgo.13692 for a patient with chronic, symptomatic anemia with menorrhagia and uterine fibroid.
... In the same study, vitamin D demonstrated a proapoptotic effect by reducing the expression of B-cell lymphoma 2. 22 In recent years, EGCG also proved effective in the treatment of UFs. 8,[23][24][25][26] Its activity has been demonstrated both in vitro and in vivo, ...
Article
Full-text available
Uterine fibroids (UF) represent the most common benign tumours in females of reproductive age, and can negatively affect fertility. Patients with UFs need to reduce the tumour size with pharmacological treatments or surgically remove the fibroid before using assisted reproductive technology (ART). On the other hand, surgery implies long waiting times before ART to avoid the risk of rupture of the uterus. Long waiting periods are often unacceptable for older individuals who want to undergo ART procedures. Unfortunately, no specific and safe treatment for UFs is currently available. Here the author reports two cases of patients with UFs and associated heavy menstrual bleeding who seek pregnancy through ART. Both underwent a daily treatment with epigallocatechin gallate, vitamin D, vitamin B6, and D-chiro-inositol for 3 months. The patients showed a volume reduction of 73.8% and 68.4%, respectively. This was associated with decreased blood loss (42.1% and 48.7%, respectively). After 3 months from the end of the treatment, both patients underwent ART procedure without the need for surgical intervention.
Article
Full-text available
Uterine fibroids are benign tumors that arise from the smooth muscle tissue of the uterus and are the most common tumors in women. Due to their high prevalence, costs for the health care system and the substantial impact on women’s quality of life, they are a significant public health concern. Previous literature on the impact of diet on the occurrence, growth and symptoms of fibroids is limited. Recently, many papers have been written on this topic. A scoping review of PubMed and Cochrane databases was performed using the following keywords: uterine fibroids, antioxidants, diet, diet, vegetarian, vegetables, fruits, meat and soy foods, dairy products, tea, vitamin D, vitamin C, ascorbic acid. Preliminary research has shown a beneficial effect of vegetable and fruit consumption on the occurrence of fibroids. A relationship between hypovitaminosis D and an increased risk of fibroids has also been demonstrated. Studies on epigallocatechin gallate showed its apoptosis-promoting and antifibrinolytic effect in fibroid cells. Initial results are promising, but further randomized trials are needed to draw firm conclusions about the effects of diet and nutrients on uterine fibroids.
Article
Full-text available
Background; Uterine fibroids (UFs) are benign tumors that develop from the uterus' smooth muscle cells and are mostly found in the pelvic. Aim of the work; we aimed to compare serum vitamin D level between patients, have uterine fibroids and those without. Subjects and methods; our current study included 90 females and was conducted as a case control study; 50 were diagnosed with uterine fibroid and 40 were healthy volunteers (control) during the period from July 2019 to July 2020. they were recruited from Al Zahra’a University Hospital. Serum vitamin D level was measured by ELISA. Results; Vitamin D serum concentrations were significantly lower in the studied females with UFs compared to healthy controls (mean 19.24 ±16.0 vs. 33.83 ±17.5) ng/mL in cases and controls, respectively, with a statistically significant p-value <0.001. Serum vitamin D levels have a significant, linear, negative, moderate association with uterine fibroids size. Conclusion; in our study group, reduced serum vitamin D levels were found to be inversely related to UF burden. A lack of vitamin D may be a risk factor for the incidence of uterine fibroids.
Article
Full-text available
One of the many factors involved in the development of uterine fibroids is vitamin D deficiency. One aspect of this deficiency is decreased serum concentration of calcidiol-25(OH)D, a metabolite of D3 vitamin. The active form of vitamin D3, which arises after numerous enzymatic reactions, is calcitriol-1,25(OH)2D3; this compound is transported to various body tissues. Vitamin D possesses extra-genomic effects due to its influence on various signaling pathways, i.e., through activating tyrosine kinases and by genomic effects via binding to a specific nuclear receptor, vitamin D receptor (VDR). The vitamin D/VDR complex regulates the expression of genes and is involved in the pathogenesis of fibroids. Numerous studies have shown that vitamin D supplementation reduces fibroid size. It has also been shown that the expression of VDR in myoma tissue is significantly lower than in the uterine muscle tissue at the tumor periphery. However, the expression of VDR in non-myoma uterine muscle has not previously been investigated. Our VDR expression studies were performed immunohistochemically with tissue microarrays (TMA) in three tissue groups: 98 uterine myoma tissues, 98 uterine tissues (tumor margin), and 12 tissues of normal uterine muscle (i.e., without fibroids). A statistical analysis showed significantly lower VDR expression in uterine muscle at the periphery of the fibroid than in healthy uterine muscle. Lower expression of VDR at the periphery of the myoma compared to that in normal uterine muscle may indicate potential for new myomas. This observation and the described reduction in the size of fibroids after vitamin D supplementation supports the hypothesis of causal development of uterine fibroids and may be useful for the prevention of re-development in the event of their excision from the uterus.
Article
Uterine fibroids are the most common tumor of reproductive-age women worldwide and cause significant morbidity in affected women. Vitamin D has emerged as a potential therapy for uterine fibroids based on experimental and epidemiologic evidence. The objective of this systematic review was to evaluate the role of vitamin D in the pathophysiology of uterine fibroids and its efficacy for prevention and treatment of fibroids. A comprehensive search was conducted of Cochrane Library, Embase, PubMed, Scopus, and Web of Science from inception to March 2022. English-language publications that evaluated vitamin D and uterine fibroids in humans, whether experimental or clinical, were considered. The search yielded 960 publications, and 89 publications met inclusion criteria: 23 preclinical studies, 25 clinical studies, and 41 review articles. Preclinical studies indicated that the vitamin D receptor was decreased in fibroid cells. Vitamin D treatment of fibroid cells decreased proliferation, extracellular matrix protein expression, and Wnt/β-catenin signaling. Fourteen clinical studies (n = 3535 participants) assessed serum vitamin D level in women with ultrasound-proven fibroids, and all found an inverse correlation between serum vitamin D level and presence of fibroids. Five clinical studies (n = 472 patients) evaluated treatment of fibroids with vitamin D. Four of five studies showed vitamin D significantly inhibited fibroid growth. One pilot study (n = 109 patients) of vitamin D for secondary prevention of fibroids demonstrated smaller recurrent fibroids in the treated group. These studies provide evidence for vitamin D as a therapy for uterine fibroids and underscore the need for well-designed, randomized, placebo-controlled clinical trials.
Article
Full-text available
According to recent studies and observations in clinical practice, uterine fibroids increase the risk of preterm birth. There are several theories on the pathogenesis of preterm birth in the presence of fibroids. One theory proclaims that fibroid necrosis leads to preterm birth, though pathophysiological mechanisms have not been described. Necrotic tissue secretes specific cytokines and proteins and we suggest these to be comparable to the inflammatory response leading to spontaneous preterm birth. We hypothesize that fibroid necrosis could induce preterm parturition through a similar inflammatory response. This new hypothesis generates novel perspectives for future research and the development of preventative strategies for preterm birth. Moreover, we emphasize the importance of the recognition of fibroids and especially fibroid necrosis by clinicians during pregnancy.
Article
Full-text available
Babunashvili E. L., Buyanova S. N., Logutova L. S., Schukina N. A., Shuginin I. O., Kovalenko T. S., Yudina N.V., Shcherbatykh M. G. VAGINAL DELIVERY AFTER GIANT FIBROID MYOMECTOMY IN A YOUNG PREGNANT WOMAN (CASE STUDY) Moscow Regional Research Institute of Obstetrics and Gynecology GBUZ MO MONIIAG, Moscow, Russia, st. Pokrovka, 22A Director: Doctor of Medical Sciences, Professor V.A. Petrukhin Summary. Hysteromyoma is one of the most common gynecopathies; its incidence in women of childbearing age reaches 40%. Large and giant tumours can cause fatal complications in mothers and feti. Abdominal myomectomy in pregnancy is an extremely rare practice featuring very few positive results. GBUZ MO MONIIAG has 20 years of experience laparotomic myomectomy of giant fibroids during gestation. We have described a unique case in this paper: myomectomy of a giant conglomerate mass of myoma nodules measuring 20x18x20 cm during pregnancy and successful vaginal delivery in an 18-year-old patient. Medical guidelines that include an improved pregnancy and childbirth algorithms for patients with large and giant uterine fibroids should be developed to reduce the number of abdominal births and perinatal complications thus improving mothers and their unborn children's health. Keywords: pregnancy and hysteromyoma; large, giant uterine fibroid; laparotomic myomectomy; vaginal birth with uterine fibroids.
Article
Ethnopharmacological relevance Uterine leiomyoma (UL) is a common severe gynecological issue. In China, Xuefu Zhuyu Decoction (XFZYD), combined with Mifepristone, is widely used in the treatment of UL. However, their combined effectiveness and safety for this purpose have not yet been explored. Aim of the study: This systematic review aims to evaluate the effectiveness and safety of XFZYD combined with Mifepristone as a method of treatment for UL. Materials and methods We searched the following 7 databases: 3 English medical databases (PubMed, EMBASE, Cochrane Library), and 4 Chinese medical databases (Chinese Biomedical Literature Database (CBM), Chinese National Knowledge Infrastructure (CNKI), Chinese Scientific Journal Database (VIP), and the Wanfang database). The primary outcome was the effect of XFZYD combined with Mifepristone on the effective rate, uterine leiomyoma volume (ULV), and uterine volume (UV) of uterine leiomyoma. Bias risk was assessed using the Cochrane risk of bias tool. The software RevMan5 was used to evaluate the quality of the included studies and process the data. Results The 11 studies included in this systematic review were all undertaken in China, with a total of 902 participants. The meta-analysis of XFZYD combined with Mifepristone compared with Mifepristone alone showed that the effective rate (RR 1.20, 95% confidence interval (CI): 1.14–1.27, P < 0.00001), ULV (SMD -1.60, 95% CI: 2.11 to −1.08, P < 0.00001), and UV (SMD -1.65, 95% CI: 1.85 to −1.44, P < 0.00001) in the primary outcomes, and estradiol (E2) (MD -51.81, 95% CI: 69.68 to −33.94, P < 0.00001), luteinizing hormone (LH) (MD -3.09, 95% CI: 3.58 to −2.60, P < 0.00001), follicle stimulating hormone (FSH) (MD -1.09, 95% CI: 1.86 to −0.31, P = 0.006), progesterone (P) (MD -3.55, 95% CI: 4.54 to −2.55, P < 0.00001), and adverse events (RR 0.55, 95% CI: 0.34–0.89), P = 0.01) in the secondary outcomes were significantly reduced, and the data were statistically significant. The subgroups of ULV, E2, and FSH showed that the treatment time might not have been the heterogeneous source of ULV and FSH, but was the heterogeneous source of E2. Sensitivity analysis was carried out on the 3 outcome indicators, and the results were relatively stable after excluding one reference for each indicator. Conclusion There is some encouraging evidence that the combination of XFZYD and Mifepristone can benefit patients by treating UL. However, because of research shortcomings such as lacking allocation concealment and blindness, this study's results should be treated with caution. In order to verify the advantages of this method, it is necessary to carry out further large-scale randomized controlled trials.
Article
Full-text available
Uterine fibroids affect a wide cross-section of the population, with prevalence, symptom severity, and overall disease burden generally higher among black women, likely due to both genetic and environmental factors. Potential symptoms of uterine fibroids include painful and excessive uterine bleeding, interference with everyday life and self-image, and impaired fertility. Because of the high estimated prevalence and costs associated with treatments, the direct and indirect costs of uterine fibroids are substantial for both the health care system and the individual patient. Special patient populations—such as black women, women seeking to retain fertility, and women with asymptomatic fibroids—have particular treatment needs that require a variety of diagnostic methods and treatment options. Despite the widespread occurrence of uterine fibroids and newer treatment options, little high-quality data are available to formulate evidence-based guidelines that address these unmet patient needs. Specific areas in need of attention include improving diagnostic techniques, increasing patient access to early treatment, and identifying best practices for this diverse patient population.
Article
Full-text available
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with high mortality. Two novel antifibrotic agents, pirfenidone and nintedanib, have received licences for use in IPF in recent years. Phase III, multinational, randomised control trials have provided evidence that both drugs reduce decline in forced vital capacity (FVC) over time, while further post hoc studies have suggested that both pirfenidone and nintedanib can be efficacious, regardless of age and severity of baseline lung function. Both therapeutic agents have manageable side effect profiles. In the absence of head-to-head data, decisions regarding which agent to choose when starting treatment for IPF should take into consideration joint decision making between patients and clinicians based on accurate information in the decision-making process. Questions remain as to the role of combination antifibrotic therapy as a future treatment option.
Article
Full-text available
Curcumin (CUR) is a yellow polyphenolic compound derived from the plant turmeric. It is widely used to treat many types of diseases, including cancers such as those of lung, cervices, prostate, breast, bone and liver. However, its effectiveness has been limited due to poor aqueous solubility, low bioavailability and rapid metabolism and systemic elimination. To solve these problems, researchers have tried to explore novel drug delivery systems such as liposomes, solid dispersion, microemulsion, micelles, nanogels and dendrimers. Among these, liposomes have been the most extensively studied. Liposomal CUR formulation has greater growth inhibitory and pro-apoptotic effects on cancer cells. This review mainly focuses on the preparation of liposomes containing CUR and its use in cancer therapy.
Article
Uterine fibroids (leiomyomata) are common uterine neoplasms that are often associated with detrimental symptoms, such as heavy menstrual bleeding, dysmenorrhea, and pressure-related symptoms, which often lead to surgical intervention, including hysterectomy. Although our understanding of the pathophysiology of uterine fibroids, particularly their dependency on progesterone and the role of growth factors, has increased during the past two decades, the medical options currently approved by the United States Food and Drug Administration are limited and are only used for short-term symptom management. A long-term, safe, and effective treatment is not yet available in the United States. However, emerging medical treatments that are currently under clinical development, including progesterone receptor modulators and oral gonadotropin-releasing hormone antagonists, offer hope of new long-term medical therapies for symptomatic uterine fibroids as an alternative to surgery. Herein, we discuss the current and emerging medical therapies for fibroids based on evidence from randomized clinical trials. © 2017 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA.
Article
We aimed to evaluate the relationship between serum 25-hydroxy vitamin D levels and the risk of uterine fibroids in premenopausal women in Turkish population in this prospective observational cross-sectional study. Sixty-eight women with at least one uterine leiomyoma ≥10 mm were compared with 56 healthy controls. Serum 25-(OH) vitamin D3 levels were measured by electrochemiluminescence immunoassay. The groups were similar in terms of age, BMI, gravidity and parity numbers. The mean level of 25-(OH) D3 were 7.28 ± 4.94 ng/ml and 78% of patients (n = 97) had severe vitamin D deficiency ( < 10 ng/mL). Vitamin D levels were significantly lower in the study group (6.54 ± 4.66 ng/ml vs. 8.18 ± 5.16 ng/ml, respectively; p = .009). Vitamin D levels were not correlated with size, volume, localization and number of leiomyomas. Traditional covered clothing style, low education level and being housewife were risk factors for Vitamin D deficiency. This is the first study that investigates the vitamin D levels in women with leiomyomas in Turkish population. Influence of vitamin D on uterine leiomyoma formation to may lead to new preventive strategies in the future.
Article
Background: Uterine fibroids (UF) are associated with significant health-related quality of life (HRQL) impact. This study examined the impact of UF symptoms on HRQL. Methods: An online cross-sectional survey of 18 to 49 year US women was conducted and collected demographics, UF prevalence, symptoms, and HRQL using the UFS-QOL. Descriptive statistics were used to examine the impact of symptom presence, severity, bothersomeness, and number of UF symptoms on HRQL. Analyses were weighted to match the US female population distribution. Multivariate regressions were performed with each subscale as a dependent variable to examine the impact of individual UF symptoms on HRQL. Results: A total of 59,411 (15.5%) panel members completed the prevalence screener; 4,848 met inclusion criteria; 955 had UF and no hysterectomy. Mean age was 40.3; 58% were white; 63% were married/civil union. Common UF symptoms were: lower back pain (65%), fatigue/weariness (63%), bloating (61%), pelvic pain/cramping during menses (63%), and heavy bleeding during menses (54%). Mean UFS-QoL subscale scores were significantly (p < 0.05) worse among women with a UF symptom versus women without the symptom. Women who rated their UF symptoms as severe had significantly (p < 0.001) worse UFS-QoL scores than women with mild or moderate symptoms. UFS-QoL subscale scores worsened as the number of symptoms increased. In the regressions, the presence of bleeding and non-bleeding symptoms were related to worse UFS-QoL subscale scores. Conclusion: HRQL among women with UF was significantly impacted by UF-related symptoms. Greater impact was observed as the number and severity of symptoms increased.
Article
Aspirin has been confirmed as an effective antitumor drug in various cancers. However, the relationship between aspirin and uterine leiomyoma is still underexplored. Here, we explored the effects of aspirin on human uterine leiomyoma cells and provide insights into the underlying mechanisms. Cell Counting Kit-8 (CCK-8) and flow cytometry analysis showed that aspirin treatment inhibited cell proliferation and promoted cell cycle arrest at G0/G1 phase in a dose- and time‑dependent manner of human uterine leiomyoma cells. Further studies revealed that aspirin blocked the interaction between K-Ras and p110α by co-immunoprecipitation and immunofluorescence. Western blotting demonstrated K‑Ras‑p110α interaction was required for the effects of aspirin‑induced inhibition on cell growth and cell cycle transition via cell cycle regulators, including cyclin D1 and cyclin-dependent kinase 2 (CDK2). PI3K/Akt/caspase signaling pathway was involved in human uterine leiomyoma cell growth under aspirin treatment. Taken together, these results suggest that aspirin inhibited human uterine leiomyoma cell growth by regulating K‑Ras‑p110α interaction. Aspirin which targeting on interaction between K-Ras and p110α may serve as a new therapeutic drug for uterine leiomyoma treatment.
Article
Epigallocatechin gallate is a polyphenol of tea plants. Other than tea its trace amounts are found in apple skin, onions and plums. It has anti-adipogenic and anti-oxidant potential. It was investigated that epigallocatechin gallate stopped the adipogenic differentiation of mice mesenchymal stem cells but its underlying mechanism is not well understood. Different proteins and transcription factors responsible for differentiation of adipocytes could be its targets. This study was designed to determine the potential target of epigallocatechin gallate in human. Human Peroxisome Proliferator-Activated Receptors (PPAR) gamma protein was selected as the potential target as it is a key transcription factor for differentiation of adipose cells. Docking analysis of PPAR gamma and epigallocatechin gallate demonstrated that epigallocatechin gallate binds with PPAR gamma at its active site and blocks its activity. This study helps in understanding the mode of action of epigallocatechin gallate that would help for anti-obesity drug development.
Article
Introduction: Much research has gone into developing medications that can be used to alleviate endometriosis-associated symptoms. In addition to already established medications, a new GnRH antagonist, elagolix, is in development. The novelty of this drug compared to other GnRH antagonists, is its nonpeptide structure, allowing it to be administered orally. Areas covered: We analyzed several Phase I, II and III clinical trials that have evaluated the safety and efficacy of this new medication. Expert opinion: Since many medications have been put on the market and have gained popularity for the treatment of endometriosis-associated symptoms, the demonstration of equality or superiority of effect, tolerability, as well as patient compliance should be assessed when introducing a new drug. While elagolix may have an advantage over established GnRH agonists, in that it does not lead to a ‘flare-up’ effect, it too, takes a toll on bone mineral density. Nevertheless, studies have shown that this new oral GnRH antagonist is well tolerated, and the side effects have been described as ‘mild or moderate’. However, in order to examine whether elagolix can compete with or even surpass established gold-standard medical treatments in this field, further studies that directly compare elagolix to said treatments, might be necessary.