You are on page 1of 7

Middle East Fertility Society Journal 23 (2018) 1–7

Contents lists available at ScienceDirect

Middle East Fertility Society Journal


journal homepage: www.sciencedirect.com

Review

The thin endometrium in assisted reproductive technology: An ongoing


challenge
Maryam Eftekhar a, Nasim Tabibnejad a,⇑, Afsar Alsadat Tabatabaie b
a
Department of Obstetrics & Gynecology, Shahid Sadoughi University of Medical Science, Yazd, Iran
b
Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Science, Yazd, Iran

a r t i c l e i n f o a b s t r a c t

Article history: Thin endometrium is identified to adversely affect reproductive success rates after assisted reproductive
Received 3 December 2017 technology (ART). Several treatment modalities have been presented to patients with thin endometrium,
Accepted 15 December 2017 to improve endometrial thickness and the subsequent endometrial receptivity. These approaches com-
Available online 21 December 2017
prising hormonal management by estradiol, tamoxifen, human chorionic gonadotropin (hCG) and
gonadotropin-releasing hormone (GnRH) agonist, vasoactive agents such as aspirin, vitamin E, pentoxi-
Keywords: fylline, nitroglycerin and sildenafil, intra-uterine infusion of growth factor such as Granulocyte Colony
Thin endometrium
Stimulating Factor (G-CSF) and the latest application of platelet-rich plasma, electrical stimulation, regen-
ART
Treatment, endometrial receptivity
erative medicine and presentation of endometrial receptivity array. In spite of the large variety of treat-
ment, most of the choices achieve only minor modification in the endometrium thickness and have not
been validated so far. Treatment of thin endometrium remains a challenge and future enormous investi-
gations are required to further clarification and ideal management of patients with thin endometrium.
Ó 2017 Middle East Fertility Society. Production and hosting by Elsevier B.V. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Pathophysiology of thin endometrium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Treatment of thin endometrium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.1. Estradiol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.2. Human chorionic gonadotropin (hCG) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.3. Midluteal GnRH-agonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.4. Tamoxifen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.5. Aspirin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.6. Sildenafil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.7. Vitamin E and pentoxifylline . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.8. Nitroglycerin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.9. Granulocyte colony stimulating factor (G-CSF) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.10. Platelet-rich plasma (PRP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.11. Neuromuscular electrical stimulation (NMES) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.12. Stem cell therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.13. Endometrial receptivity array (ERA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5

Peer review under responsibility of Middle East Fertility Society.


⇑ Corresponding author at: Yazd Reproductive Sciences Institute, Shahid
Sadoughi University of Medical Science, Bouali Ave, Safayieh, Yazd, Iran.
E-mail address: nasimtabibnejad@ssu.ac.ir (N. Tabibnejad).

https://doi.org/10.1016/j.mefs.2017.12.006
1110-5690/Ó 2017 Middle East Fertility Society. Production and hosting by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
2 M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7

1. Introduction 3. Treatment of thin endometrium

Despite recent developments in assisted reproductive tech- Since a thin endometrium is a multifactorial condition, its man-
niques (ART), the implantation rates still remain relatively low. agement should be cause-related, with the aim of increasing
Successful implantation requires high quality embryo, receptive endometrial receptivity and simplifying implantation [30,31].
endometrium, and perfect embryo transfer technique [1]. The However, improving endometrial growth in patients with thin
receptive endometrium define as a healthy uterine milieu contain- endometrium is very challenging; several regimens have been
ing the transformation of endometrial cells into decidua cells tried in the literatures [21,29,30].
appropriate for implantation of blastocysts, and rapid growth of
placenta [2]. Therefore, endometrial assessment is routinely per-
3.1. Estradiol
formed during in vitro fertilization (IVF) and intracytoplasmic
sperm injection (ICSI). In addition endometrial thickness (EMT)
Infertile patients, who display inadequate endometrium thick-
have been considered as a marker of endometrium receptivity [3]
ness, will be initially treated with oral estradiol. As the endome-
and a prognostic factors for embryo transfers during IVF/ICSI treat-
trium is a hormone dependent tissue estrogen supports
ment [4,5]. It is known that appropriate endometrial thickness is
endometrial proliferation by causing spiral artery contraction and
essential for a successful pregnancy [6,7] and several studies have
decreasing oxygen tension in the functional layer, that simplifies
stated low pregnancy rates in the presence of thin endometrium
embryo implantation [32,33]. The best method for prescription of
[8–11]. The optimal endometrial thickness for conception remains
estradiol is oral administration, and there are no significant differ-
controversial among clinicians. EMT less than 7 mm on ultrasound
ences between micronized estradiol or estradiol valerate [34]. The
is generally considered sub-optimal for embryo transfer and is cor-
extended administration of estradiol valerate during controlled
related to a decreased probability of pregnancy. Thin endometrium
ovarian hyperstimulation (COH) cycles for 14–82 days, improve
remains a challenge in gynecology and reproductive science with
the mean endometrial thickness from 6.7 to 8.6 mm as well as sig-
only slight enhancements attained with the currently available
nificant increase of pregnancy rate (38.5 vs. 4.3%) [35]. Similarly a
treatment. In this review, we focus on developing and promising
case of repeated implantation failure due to unreceptive thin endo-
treatment options for refractory thin endometrium.
metrium received extended estrogen supplementation for an
extended period, using 16 mg/day of estradiol valerate from the
third day of her menstrual cycle for 9 days before COH. IVF in this
patient led to a twin pregnancy and delivery at term [36]. In con-
2. Pathophysiology of thin endometrium
trast Demir et al. reported that estradiol supplementation with
estradiol hemihydrate 4 mg/day started on the day of hCG among
It has been supposed that a thin endometrium is caused by a
women with thin endometrium undergoing ICSI, did not improve
diminished normal endometrial growth. However, little evidence
clinical pregnancy rate, implantation rate (16% vs. 10.4%), and
is presented with regards to reasons for impaired endometrial
endometrial thickness [37]. According to a meta-analysis results,
growth in patients with thin endometrium. Acute or chronic infec-
administration of pure ethinyl estradiol (EE) for treatment of thin
tion like genital tuberculosis may result in destruction of endome-
endometrium, increase the endometrial thickness in comparison
trium basal layer due to replacing healthy endometrium by fibroid
to patients whom used placebo. Moreover, the best result were
tissue after healing [12]. Iatrogenic thin endometrium can be
achieved while EE administration is starting on 7th–10th day of
occurred by surgical procedures such as repeated curettage,
menstrual cycle with the dose of 0.02–0.05 mg/day for 5 days
polypectomy, laparoscopic or hysteroscopic myomectomy as well
[38]. As the highest serum and endometrial level of estrogen occur
as immethodical drug usage such as clomiphene citrate [12]. It is
after vaginal administration, it is considered as the desirable route
also indicated that thin endometrium may be a result of individual
in cases that do not response to the other ways. Therefore, Cetin-
uterine structural pattern [13].
kaya and colleagues administered estrogen vaginally 25 mg daily
Angiogenesis plays a key role in different female reproductive
from 4th day of the cycle for 15 days in Clomiphene citrate induced
developments, including growth of dominant follicles, formation
cycle. They reported significant increase in endometrial thickness
of a corpus luteum, and endometrial pattern [14–18]. Endometrial
on the day of ovulation in estrogen + clomiphene citrate group
angiogenesis is vital for regeneration endometrium after menstru-
compare to the group where only Clomiphene citrate was used,
ation and to provide a vascularized receptive endometrium for
but there was no difference in pregnancy rate [39]. In the same
implantation [14,17]. The majority of studies has focused on vascu-
way another study compared oral and vaginal administration of
lar endothelial growth factor (VEGF) as a regulator for endometrial
estradiol among donor oocytes recipients. The results showed an
angiogenesis, and a number of works stated that VEGF is expressed
increase in endometrial thickness as well as ongoing pregnancy
differentially in the uterine with thin endometrium [17,19–21].
rate when vaginal estradiol administration extended to 4–6 weeks
Uterine blood flow is an essential marker regulating endometrial
in women who failed to achieve acceptable endometrial thickness
growth and is narrowly related to vascular development of the
after oral estradiol administration [40]. Oral estrogens were also
endometrium [22–25]. Recent improvements in ultrasonography
used for endometrium preparation in FET, where prior IVF failure
have provided new opportunities for noninvasive evaluation of
was believed to be due to thin endometrium. Jimenez and col-
endometrial perfusion. A significant reduced pregnancy rate in
leagues administered oral estradiol 2 mg three times a day from
IVF-ET patients with low uterine blood flow, displayed a close rela-
day 1st for 12 days. They stated satisfactory development of endo-
tionship between uterine blood flow and uterine receptivity
metrium in 67% patients [41].
[26,27]. Asherman’s syndrome is one of the causes of thin endome-
trium. Transvaginal ultrasonography usually reveals minimal
endometrial thickness in this patients [28]. Impaired uterine perfu- 3.2. Human chorionic gonadotropin (hCG)
sion is the reason for atrophy of the remaining endometrium due
to restricted exposure to circulating hormones. This syndrome hCG play a local paracrine role in the endometrium differentia-
may occur followed myometrial fibrosis that is remarkably tion and endometrial receptivity by regulating different cytokines
increased among women with intrauterine adhesions (IUAs) com- and growth factors [42,43]. Papanikolaou and colleagues recruited
pared to healthy controls [29]. seventeen infertile patients with the history of implantation fail-
M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7 3

ures and resistant thin endometrium. On day 8 or 9 of the estrogen more, Clinical pregnancy per cycle, spontaneous abortion or ecto-
administration (8 mg daily), hCG injection was started subcuta- pic pregnancy per Clinical pregnancy and live birth rate per cycle
neously by 150 IU daily for 7 days. After a week on hCG priming, or embryo transfer were not significantly significant between the
on the day 14 or 15 of cycle, the mean endometrial thickness mentioned groups [54]. On the basis of a Cochrane study, low-
was improved by 0.8 mm. Such as 35.3% of the patients had more dose aspirin has no substantial positive effect on pregnancy and
than 20% development in their endometrial thickness after hCG endometrial thickness [55].
priming and 17% achieved an endometrial thickness more than
7 mm [44]. Likewise in a nonrandomized clinical trial the effect 3.6. Sildenafil
of adding hCG to frozen thawed embryo transfer cycles with
history of thin endometrium was examined in 28 patients who Sildenafil citrate -an inhibitor of cGMP specific phosphodi-
had two previous failed cycles because of thin endometrium. esterase type 5 (PDE5)- prevents the breakdown of cGMP and
150 IU hCG was administrated intramuscularly from the day 8 of increases the effect of nitric oxide on vascular smooth muscles.
cycle until endometrial thickness reached at least 7 mm. The study Sildenafil citrate administration cause an improvement in uterine
suggested that adding hCG to the conventional method is effective blood flow and, in combination with estrogen, could lead to the
and improved endometrial thickness and pregnancy outcomes estrogen-induced proliferation of the endometrial lining [24]. The
significantly [45]. expression of vascular endothelial growth factor (VEGF) and Tumor
suppressor factor (p53) genes are essential for breaking down the
3.3. Midluteal GnRH-agonist endometrial cellular matrix, normalize cell growth, and induce
angiogenesis. These process are required for adequate endometrial
According to the evidence that mid-luteal GnRH-agonist admin- blood supply and consequent embryo implantation [56,57]. Silde-
istration improve pregnancy rate, one hundred and twenty women nafil citrate causes significant increases in p53 activity and stimu-
with thin endometrium undergoing IVF received either triptorelin lated angiogenic reactions with elevated VEGF [58,59]. It is showed
(0.1 mg) on the day of ovum pickup, day of embryo transfer and previously that vaginal sildenafil citrate is effective for the treat-
three days afterward as case group, or placebo as controls. The ment of women with repeated implantation failure and leads to
results indicated that treatment with GnRH-a increases the increase endometrial thickness and attaining pregnancy in women
implantation and pregnancy rate significantly as the same as E2, with poor endometrial growth [60]. Takasaki et al. evaluated the
progesterone levels and endometrial thickness [46]. effect of sildenafil on improving radial arteries blood flow and
endometrial growth. Sildenafil citrate increased radial artery-
3.4. Tamoxifen resistance index and endometrial thickness in 92% cases [61].
Recently Zinger et al. have reported two infertility patients with
Tamoxifen blocks the actions of estrogen and has a key role in Asherman’s syndrome with successful pregnancy after consuming
endocrine therapy for the treatment of estrogen receptor positive 25 mg vaginal sildenafil citrate, four times a day for 6–14 days dur-
breast cancer. Wang et al. compared reproductive outcome in ovar- ing the first half of their cycle [62]. To evaluate the effects of silde-
ian stimulation cycles using tamoxifen (40 mg/day from day 3 of nafil administration on the thickness of the endometrium and the
the menstrual cycle for 7 days) and clomiphene cycles (100 mg/day implantation rate, Dehghani-Firouzabadi and coworkers conducted
for 5 days) in patients undergoing intrauterine insemination. It was a randomized controlled trial. 80 patients who had a history of
found that tamoxifen-treated patients have a significantly poor endometrial response and frozen embryos included the study.
increased endometrial thickness and pregnancy rate [47]. Simi- 40 patients were given sildenafil citrate tablets (50 mg) daily in
larly, another study showed that switching CC to tamoxifen for addition to the estradiol and compared to 40 women who received
ovulation induction in women with adequate follicular recruit- estradiol only. They concluded that the oral usage of sildenafil
ment and thin endometrium improves endometrial thickness citrate is a good approach to improve the endometrial receptivity
[48]. Ke et al. evaluate the effect of tamoxifen on endometrial [60]. Likewise, a recent study examined the effect of adding silde-
thickness among patients with thin endometrium undergoing fro- nafil vaginal gel to CC in infertile women with thin endometrium
zen–thawed embryo transfer cycles. 226 women who had and previous CC failure (5 cycles). In the patients’ 6th cycle (CC
endometrial thickness less than 7.5 mm in previous cycles were only), women sustained on CC 100 mg/day for 5 days. In the 7th
recruited. Tamoxifen administration improves endometrial thick- cycle, women administered usual dose of CC supplemented in con-
ness in patients after natural cycle as the same as hormone junction with sildenafil vaginal gel (5 gm, containing 50 mg silde-
replacement treatment and ovulation induction cycles during nafil) twice daily from day 8 to day of HCG administration. The
FET. However, Patients with PCOS got the most advantage finding indicated that sildenafil vaginal gel considerably increased
from tamoxifen and achieve higher pregnancy outcomes [49]. endometrial thickness and uterine blood flow, and may improve
pregnancy rate [63].
3.5. Aspirin
3.7. Vitamin E and pentoxifylline
The effect of low dose aspirin on pregnancy rate is still debated
in the literatures. Even though some studies reported the positive The combination of pentoxifylline (PTX) as a derivative of
impact of low dose aspirin on endometrial thickness, pattern, and methylxanthine that induce vasodilatation and vitamin E as an
endometrial blood flow [50,51], others claimed that aspirin do not antioxidant was used for treatment of thin endometrium. This
increase success of implantation [25,52]. Hsieh et al. found signif- combination was given to oocyte recipients with inadequate
icantly higher percentages of trilaminar endometrium (46.5% vs. endometrial thickness after vaginal E2 administraion. This method
26.2%) and pregnancy rates (18.4% vs. 9.0%) following low-dose results in significantly improved endometrial thickness, pregnancy
aspirin (100 mg) therapy non-aspirin group [53]. In a systematic and delivery rates [3]. In the same way, Acharya et al. found an
review and meta-analysis the effect of low-dose aspirin on likeli- improvement in endometrial thickness and pregnancy rate of
hood of pregnancy was studied among women undergoing IVF/ 40% in treated patients [64]. Similarly, it is reported that use of
ICSI. Clinical pregnancy rate per embryo transfer was similar vitamin E cause adequate ET in 52% of patients as well as growth
between patients who received low-dose aspirin and those who of the glandular epithelium, number of blood vessels and VEGF
used placebo or no treatment (RR 1.09, 95% CI 0.92–1.29). Further- protein expression [3]. Also, Cicek and colleagues found that
4 M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7

Vitamin E administration could improve the endometrial thickness endometrium. Five women with thin endometrium after standard
but not implantation and ongoing pregnancy rate in women with hormone replacement therapy (HRT) who were candidate for IVF
unexplained infertility [65]. Pentoxifylline has been used clinically included the study. PRP was prepared from autologous blood by
in the treatment of vascular abnormalities, and is also a good centrifugation, and infusion of 0.5–1 ml of PRP into the uterine cav-
choice for radiation-induced fibrosis. However the precise mecha- ity on the 10th day of HRT cycle was performed. If endometrial
nism has not yet been known, the influence is improved while it thickness failed to improve 72 h later, PRP infusion was done 1–2
use in combination with vitamin E [66]. One study assessed the times in each cycle. Embryos were transferred while the endome-
antifibrotic effect of PTX by a combination of PTX and tocopherol trium thickness reached more than 7 mm. Endometrial growth as
(vitamin E) in patients with a thin endometrium who were joined well as successful pregnancy was occurred in all the patients after
an oocyte donation program. Eighteen oocyte recipients who failed PRP infusion [77]. In the same way, ten patients with the history of
to improve endometrial thickness after receiving vaginal micro- inadequate endometrial growth in FET cycles were enrolled in
nized estradiol were included the study. The patients received a another study. Intrauterine infusion of PRP was performed for all
combination of PTX (800 mg/day) and vitamin E (1000 IU/day) patients. According to the results endometrial thickness increased
for 6 months. Pregnancy rate, endometrial thickness and ovarian and 50% of patients achieved pregnancy [78].
function have been developed [3].
3.11. Neuromuscular electrical stimulation (NMES)
3.8. Nitroglycerin
NMES defined as electrical stimulation of a group of muscles
Nitric oxide (NO) is a vasodilator that involved in endometrium through electrodes located on the skin. It is demonstrated that
reception and regulation of reproductive cycle. NO administration NMES in an effective method for treatment of pelvic pain, low back
may have beneficial effects on in women with a thin endometrium. pain, stress urinary incontinence, sexual dysfunction, and constipa-
However there is no publication on this issue, two studies reported tion [79]. A total of 40 patients with thin endometrium undergoing
that usage of glyceryl trinitrate patch induces a reversible rise in FET cycle divided to NMES-treated and control group. Endometrial
subendometrial blood flow velocity during days 9–12 of the men- thickness was improved in 60% of patients after the NMES therapy
strual cycle [67] with no positive effect on implantation or preg- and the difference was statistically significant. Moreover, NMES
nancy rates among patients with implantation failure [68]. therapy resulted in higher pregnancy [80].

3.9. Granulocyte colony stimulating factor (G-CSF) 3.12. Stem cell therapy

It is stated that G-CSF may increase the endometrial thickness It is indicated that endometrial stem cells are present in both
and the chance of pregnancy in the patient with extremely thin basalis and functionalis layers of the human endometrium, and
endometrium [69]. In the same way, Tehraninejad and coworkers these cells play a key role in regenerating endometrial lining dur-
showed that infusion of G-CSF in endometrial cavity is safe and ing each menstrual cycle [81,82]. It is also showed that human
possibly effective to increase endometrial growth in patient with endometrial adult stem cells are capable to produce human endo-
thin and unresponsive endometrium [70]. Endometrial perfusion metrium after transplantation in NOD-SCID mice renal capsules
with G-CSF is effective in expanding unresponsive thin endome- [83]. Other studies claimed that transplanted BMSCs into rats’ uter-
trium to at least minimal thickness of 7 mm, which was resistant ine cavity [84] as well as transplantation by tail vein IV injection
to estrogen and vasodilators. A growth occurred in endometrial [85] resulted in a remarkable thicker endometrial lining and higher
thickness can be detected within 48–72 h of G-CSF administration expression of cytokeratin, vimentin, integrin acb3, and leukaemia
[71]. This approach in infertile women on the day of hCG adminis- inhibitor factor. Likewise it is revealed by Taylor and colleagues
tration and in combination with second fusion resulted in low but that non-endometrial stem cells such as hematopoietic and non-
very reasonable clinical pregnancy rate [72]. In contrast, a random- hematopoietic bone marrow-derived stem cells (BMDSCs) could
ized clinical trial in normal IVF patients, G-CSF did not increase induce endometrial regeneration [86]. Interestingly male origin
endometrial thickness, implantation rates, or clinical pregnancy BMDSCs were found in endometrium of female bone marrow
rates [73]. Some other studies confirmed these results. Eftekhar transplant recipients [87]. Moreover, it is demonstrated that
et al. failed to demonstrate that G-CSF improve endometrial thick- BMDSCs induce regeneration of endometrial cells by paracrine sig-
ness but they reported G-CSF positive effect on chemical and clin- naling [88] and this effect is twofold higher during uterine injury
ical pregnancy rate of the infertile women with thin endometrium rather than monthly cyclic regeneration of the endometrium
in frozen-thawed embryo transfer cycle [74]. Otherwise, Li and col- [89]. Furthermore the influence of BMDSC on endometrial renewal
leagues stated effectiveness of G-CSF in developing embryo after injury was confirmed in several animal studies [90,91]. On the
implantation and clinical pregnancy rate among infertile women other hand Hunter et al. did not found any significant improve-
with thin endometrium [75]. ments in endometrial lining following stromal vascular fraction
cells (SVF) treatment [92].
3.10. Platelet-rich plasma (PRP) Based on aforementioned researches, the effects of bone mar-
row stem cells in the treatment of Asherman’s syndrome and thin
PRP is blood plasma prepared from fresh whole blood that has endometrium were evaluated in murine model. The numbers of
been supplemented with platelets. It is collected from peripheral conceived mice were 9 of the 10 in the BMDSC transplant group
veins and contains several growth factors and cytokines [76]. As in comparison with 3 of 10 in the non-transplanted group [93].
soon as platelets are motivated in PRP, cytokines and growth fac- Similarly, Kilic and coworkers treated Asherman’s syndrome
tors turn out to be active and are secreted within 10 min. Recently, among Wistar albino rats using either mesenchymal stem cells
PRP has been commonly applied in different clinical situations; (MSCs) or oral estrogen or combination of these two methods.
however, little is identified about the application of PRP in the Fibrosis decreased but vascularization and immunehistochemical
treatment of thin endometrium. For the first time, Chang and staining increased in all treated groups compare to controls [94].
coworkers introduced intrauterine infusion of PRP as a new In a recent study, seven types of BMDCs in the endometrial
approach for the treatment of thin endometrium. They evaluated renewal in mice were investigated and results showed that MSCs
the efficacy of PRP in the therapy of infertile women with thin and EPCs together with mOct4(+) BM-HypoSCs prompted the high-
M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7 5

est degree of regeneration [95]. There are few reports regarding [3] N. Ledee-Bataille, F. Olivennes, J.L. Lefaix, G. Chaouat, R. Frydman, S. Delanian,
Combined treatment by pentoxifylline and tocopherol for recipient women
human application of stem cells for the treatment of thin endome-
with a thin endometrium enrolled in an oocyte donation programme, Hum.
trium. In a plan of autologous stem cells therapy, endometrial Reprod. 17 (5) (2002) 1249–1253.
angiogenic stem cells were isolated from bone marrow of a patient [4] C. De Geyter, M. Schmitter, M. De Geyter, E. Nieschlag, W. Holzgreve, H.P.
with Asherman’s syndrome. These cells were infused into the Schneider, Prospective evaluation of the ultrasound appearance of the
endometrium in a cohort of 1,186 infertile women, Fertil. Steril. 73 (1)
endometrial cavity and the results were endometrial thickness of (2000) 106–113.
8 mm and a confirmed clinical pregnancy following IVF [96]. Sim- [5] X. Yuan, S.H. Saravelos, Q. Wang, Y. Xu, T.C. Li, C. Zhou, Endometrial thickness
ilarly six cases of refractory Asherman’s syndrome with the history as a predictor of pregnancy outcomes in 10787 fresh IVF-ICSI cycles, Reprod.
Biomed. Online 33 (2) (2016) 197–205.
of failed standard treatment received sub-endometrial autologous [6] T. El-Toukhy, A. Coomarasamy, M. Khairy, K. Sunkara, P. Seed, Y. Khalaf, et al.,
stem cell implantation followed by exogenous oral estrogen ther- The relationship between endometrial thickness and outcome of medicated
apy. The endometrial thickness was significantly improved in com- frozen embryo replacement cycles, Fertil. Steril. 89 (4) (2008) 832–839.
[7] P. Kovacs, S. Matyas, K. Boda, S.G. Kaali, The effect of endometrial thickness on
parison with pretreatment condition. Additionally five women of IVF/ICSI outcome, Hum. Reprod. 18 (11) (2003) 2337–2341.
six patients resumed menstruation [97]. [8] A. Glissant, J. de Mouzon, R. Frydman, Ultrasound study of the endometrium
In a most recent experimental study, 16 patients with refrac- during in vitro fertilization cycles, Fertil. Steril. 44 (6) (1985) 786–790.
[9] Y. Gonen, R.F. Casper, W. Jacobson, J. Blankier, Endometrial thickness and
tory Asherman’s syndrome or endometrial atrophy received autol- growth during ovarian stimulation: a possible predictor of implantation in
ogous CD133 + BMDSCs through the spiral arterioles by in vitro fertilization, Fertil. Steril. 52 (3) (1989) 446–450.
catheterization. Endometrial thickness increased about 2.4 mm [10] J.H. Check, K. Nowroozi, J. Choe, C. Dietterich, Influence of endometrial
thickness and echo patterns on pregnancy rates during in vitro fertilization,
and 1.5 mm in Asherman’s syndrome and endometrial atrophy
Fertil. Steril. 56 (6) (1991) 1173–1175.
patients respectively. Three patients obtained spontaneous preg- [11] R.P. Dickey, T.T. Olar, D.N. Curole, S.N. Taylor, P.H. Rye, Endometrial pattern
nancy as well as seven pregnancies which attained after ART [98]. and thickness associated with pregnancy outcome after assisted reproduction
technologies, Hum. Reprod. 7 (3) (1992) 418–421.
[12] N. Mahajan, S. Sharma, The endometrium in assisted reproductive
3.13. Endometrial receptivity array (ERA) technology: how thin is thin?, J Hum. Reprod. Sci. 9 (1) (2016) 3–8.
[13] M. Scioscia, G. Lamanna, F. Lorusso, G. Serrati, L.E. Selvaggi, R. Depalo,
Characterization of endometrial growth in proliferative and early luteal phase
Challenges to catch an appropriate marker for endometrial in IVF cycles, Reprod. Biomed. Online 18 (1) (2009) 73–78.
receptivity led to definition of a molecular test; the endometrial [14] S.K. Smith, Angiogenesis and implantation, Hum. Reprod. 15 (Suppl 6) (2000)
receptivity array (ERA). It is a molecular diagnostic tool based on 59–66.
[15] C.E. Gargett, P.A. Rogers, Human endometrial angiogenesis, Reproduction 121
the transcriptomic signature of human endometrial receptivity. (2) (2001) 181–186.
The test comprises 238 genes which are differentially expressed [16] T. Shimizu, J.Y. Jiang, H. Sasada, E. Sato, Changes of messenger RNA expression
between varied genetic profiles. This array is able to predict per- of angiogenic factors and related receptors during follicular development in
gilts, Biol. Reprod. 67 (6) (2002) 1846–1852.
sonalized window of implantation irrespective of her endometrial [17] N. Sugino, S. Kashida, A. Karube-Harada, S. Takiguchi, H. Kato, Expression of
histology. The bioinformatics predictor categorizes an endometrial vascular endothelial growth factor (VEGF) and its receptors in human
sample as ‘‘receptive” or ‘‘non-receptive” [99]. Sensitivity and endometrium throughout the menstrual cycle and in early pregnancy,
Reproduction 123 (3) (2002) 379–387.
specificity of ERA are 0.99 and 0.88 respectively [100]. Cruz and [18] A. Matsuoka-Sakata, H. Tamura, H. Asada, I. Miwa, T. Taketani, Y. Yamagata,
Bellver reported a healthy term live birth in a 35-year-old cancer- et al., Changes in vascular leakage and expression of angiopoietins in the
ous woman following treatment with chemotherapy and radio- corpus luteum during pregnancy in rats, Reproduction 131 (2) (2006) 351–
360.
therapy. The woman suffers ovarian failure, hypoplastic uterus [19] S. Kashida, N. Sugino, S. Takiguchi, A. Karube, H. Takayama, Y. Yamagata,
and atrophic endometrium. The recognition of a receptive endome- et al., Regulation and role of vascular endothelial growth factor in the
trium using ERA and ovum donation resulted in a successful preg- corpus luteum during mid-pregnancy in rats, Biol. Reprod. 64 (1) (2001)
317–323.
nancy in this poor prognosis case [101]. Another study used
[20] A.M. Sharkey, K. Day, A. McPherson, S. Malik, D. Licence, S.K. Smith, et al.,
personalized embryo transfer on the day elected by ERA in the Vascular endothelial growth factor expression in human endometrium is
Indian population. After personalized ET in patients with persis- regulated by hypoxia, J. Clin. Endocrinol. Metab. 85 (1) (2000) 402–409.
tently thin endometrium, the pregnancy rate of 66.7% was attained [21] I. Miwa, H. Tamura, A. Takasaki, Y. Yamagata, K. Shimamura, N. Sugino,
Pathophysiologic features of ‘‘thin” endometrium, Fertil. Steril. 91 (4) (2009)
in this group. Application of ERA revealed that the endometrium in 998–1004.
75% of these patients was receptive regardless of its thickness of 6 [22] B.S. Hurst, J.T. Bhojwani, P.B. Marshburn, M.A. Papadakis, T.A. Loeb, M.L.
mm or less [12]. Matthews, Low-dose aspirin does not improve ovarian stimulation,
endometrial response, or pregnancy rates for in vitro fertilization, J. Exp.
In conclusion, a receptive endometrium is an essential part of Clin. Assist. Reprod. 2 (2005) 8.
embryo implantation process, and inadequate endometrial growth [23] G. Sher, J.D. Fisch, Vaginal sildenafil (Viagra): a preliminary report of a novel
is associated with lower possibility of pregnancy. Many treatment method to improve uterine artery blood flow and endometrial development
in patients undergoing IVF, Hum. Reprod. 15 (4) (2000) 806–809.
modalities have been applied to improve endometrial receptivity, [24] G. Sher, J.D. Fisch, Effect of vaginal sildenafil on the outcome of in vitro
but their efficacy remains controversial. Large randomized trials fertilization (IVF) after multiple IVF failures attributed to poor endometrial
should be conducted to evaluate method, dosage, timing of admin- development, Fertil. Steril. 78 (5) (2002) 1073–1076.
[25] B. Urman, R. Mercan, C. Alatas, B. Balaban, A. Isiklar, A. Nuhoglu, Low-dose
istration and outcomes of these treatment approaches. Stem cell aspirin does not increase implantation rates in patients undergoing
therapy is considered as a very favorable option for the most cases, intracytoplasmic sperm injection: a prospective randomized study, J. Assist.
even though, more investigation is necessary to evaluate the safety Reprod. Genet. 17 (10) (2000) 586–590.
[26] L.W. Chien, H.K. Au, P.L. Chen, J. Xiao, C.R. Tzeng, Assessment of uterine
and cost-effectiveness of this technique. ERA is an accurate test for
receptivity by the endometrial-subendometrial blood flow distribution
diagnosing endometrial receptivity and could establish a personal- pattern in women undergoing in vitro fertilization-embryo transfer, Fertil.
ized window of implantation in cases with thin endometrium. Steril. 78 (2) (2002) 245–251.
[27] S. Friedler, J.G. Schenker, A. Herman, A. Lewin, The role of ultrasonography in
the evaluation of endometrial receptivity following assisted reproductive
References treatments: a critical review, Hum. Reprod. Update 2 (4) (1996) 323–335.
[28] W.D. Schlaff, B.S. Hurst, Preoperative sonographic measurement of
[1] M. Eftekhar, R. Hosseinisadat, R. Baradaran, E. Naghshineh, Effect of endometrial pattern predicts outcome of surgical repair in patients with
granulocyte colony stimulating factor (G-CSF) on IVF outcomes in infertile severe Asherman’s syndrome, Fertil. Steril. 63 (2) (1995) 410–413.
women: an RCT, Int. J. Reprod. Biomed. (Yazd) 14 (5) (2016) 341–346. [29] L.M. Senturk, C.T. Erel, Thin endometrium in assisted reproductive
[2] A.C. Zenclussen, G.J. Hammerling, Cellular regulation of the uterine technology, Curr. Opin. Obstet. Gynecol. 20 (3) (2008) 221–228.
microenvironment that enables embryo implantation, Front. Immunol. 6 [30] O. Lebovitz, R. Orvieto, Treating patients with ‘‘thin” endometrium - an
(2015) 321. ongoing challenge, Gynecol. Endocrinol. 30 (6) (2014) 409–414.
6 M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7

[31] M. Eftekhar, E. Rahmani, S. Pourmasumi, Evaluation of clinical factors frozen embryos or embryos derived from donor oocytes, Cochrane
influencing pregnancy rate in frozen embryo transfer, Iran J. Reprod. Med. Database Syst. Rev. (1) (2010) CD006359.
12 (7) (2014) 513–518. [56] C. Goodman, R.S. Jeyendran, C.B. Coulam, P53 tumor suppressor factor,
[32] M.A. Bedaiwy, M.A. Abdelaleem, M. Hussein, N. Mousa, L.N. Brunengraber, R. plasminogen activator inhibitor, and vascular endothelial growth factor gene
F. Casper, Hormonal, follicular and endometrial dynamics in letrozole-treated polymorphisms and recurrent implantation failure, Fertil. Steril. 92 (2) (2009)
versus natural cycles in patients undergoing controlled ovarian stimulation, 494–498.
Reprod. Biol. Endocrinol. 9 (2011) 83. [57] K.S. Richter, K.R. Bugge, J.G. Bromer, M.J. Levy, Relationship between
[33] S.L. Young, Oestrogen and progesterone action on endometrium: a endometrial thickness and embryo implantation, based on 1,294 cycles of
translational approach to understanding endometrial receptivity, Reprod. in vitro fertilization with transfer of two blastocyst-stage embryos, Fertil.
Biomed. Online 27 (5) (2013) 497–505. Steril. 87 (1) (2007) 53–59.
[34] J.A. Garcia-Velasco, B. Acevedo, C. Alvarez, M. Alvarez, J. Bellver, J. Fontes, [58] X. Di, C. Gennings, H.D. Bear, L.J. Graham, C.M. Sheth, K.L. White Jr, et al.,
et al., Strategies to manage refractory endometrium: state of the art in 2016, Influence of the phosphodiesterase-5 inhibitor, sildenafil, on sensitivity to
Reprod. Biomed. Online 32 (5) (2016) 474–489. chemotherapy in breast tumor cells, Breast Cancer Res. Treat. 124 (2) (2010)
[35] M.J. Chen, J.H. Yang, F.H. Peng, S.U. Chen, H.N. Ho, Y.S. Yang, Extended 349–360.
estrogen administration for women with thin endometrium in frozen-thawed [59] A. Pyriochou, Z. Zhou, V. Koika, C. Petrou, P. Cordopatis, W.C. Sessa, et al., The
in-vitro fertilization programs, J. Assist. Reprod. Genet. 23 (7–8) (2006) 337– phosphodiesterase 5 inhibitor sildenafil stimulates angiogenesis through a
342. protein kinase G/MAPK pathway, J. Cell. Physiol. 211 (1) (2007) 197–204.
[36] M.S. Shen, C.W. Wang, C.H. Chen, C.R. Tzeng, New horizon on successful [60] R. Dehghani Firouzabadi, R. Davar, F. Hojjat, M. Mahdavi, Effect of sildenafil
management for a woman with repeated implantation failure due to citrate on endometrial preparation and outcome of frozen-thawed embryo
unresponsive thin endometrium: use of extended estrogen transfer cycles: a randomized clinical trial, Iran J. Reprod. Med. 11 (2) (2013)
supplementation, J. Obstet. Gynaecol. Res. 39 (5) (2013) 1092–1094. 151–158.
[37] B. Demir, S. Dilbaz, O. Cinar, O. Ozdegirmenci, S. Dede, B. Dundar, et al., [61] A. Takasaki, H. Tamura, I. Miwa, T. Taketani, K. Shimamura, N. Sugino,
Estradiol supplementation in intracytoplasmic sperm injection cycles with Endometrial growth and uterine blood flow: a pilot study for improving
thin endometrium, Gynecol. Endocrinol. 29 (1) (2013) 42–45. endometrial thickness in the patients with a thin endometrium, Fertil. Steril.
[38] R.F. Torres, A.E. Habana, L.G. Tansengco, The effect of estrogen 93 (6) (2010) 1851–1858.
supplementation on the endometrium and pregnancy rate among infertile [62] M. Zinger, J.H. Liu, M.A. Thomas, Successful use of vaginal sildenafil citrate in
women treated with clomifene citrate: a meta-analysis, Fertil. Steril. 84 two infertility patients with Asherman’s syndrome, J. Womens Health
(2005) S162–S163. (Larchmt) 15 (4) (2006) 442–444.
[39] K. Cetinkaya, S. Kadanali, The effect of administering vaginal estrogen to [63] A.N. Fetih, D.M. Habib, I.I. Abdelaal, M. Hussein, G.N. Fetih, E.R. Othman,
clomiphene citrate stimulated cycles on endometrial thickness and Adding sildenafil vaginal gel to clomiphene citrate in infertile women with
pregnancy rates in unexplained infertility, J. Turk. Ger. Gynecol. Assoc. 13 prior clomiphene citrate failure due to thin endometrium: a prospective self-
(3) (2012) 157–161. controlled clinical trial, Facts Views Vis. Obgyn 9 (1) (2017) 21–27.
[40] D.E. Tourgeman, C.C. Slater, F.Z. Stanczyk, R.J. Paulson, Endocrine and clinical [64] S. Acharya, E. Yasmin, A.H. Balen, The use of a combination of pentoxifylline
effects of micronized estradiol administered vaginally or orally, Fertil. Steril. and tocopherol in women with a thin endometrium undergoing assisted
75 (1) (2001) 200–202. conception therapies–a report of 20 cases, Hum. Fertil. (Camb). 12 (4) (2009)
[41] P.T. Jimenez, S.B. Schon, R.R. Odem, V.S. Ratts, E.S. Jungheim, A retrospective 198–203.
cross-sectional study: fresh cycle endometrial thickness is a sensitive [65] N. Cicek, O.G. Eryilmaz, E. Sarikaya, C. Gulerman, Y. Genc, Vitamin E effect on
predictor of inadequate endometrial thickness in frozen embryo transfer controlled ovarian stimulation of unexplained infertile women, J. Assist.
cycles, Reprod. Biol. Endocrinol. 11 (2013) 35. Reprod. Genet. 29 (4) (2012) 325–328.
[42] P. Licht, H. Fluhr, J. Neuwinger, D. Wallwiener, L. Wildt, Is human chorionic [66] T.B. Chiao, A.J. Lee, Role of pentoxifylline and vitamin E in attenuation of
gonadotropin directly involved in the regulation of human implantation?, radiation-induced fibrosis, Ann. Pharmacother. 39 (3) (2005) 516–522.
Mol Cell. Endocrinol. 269 (1–2) (2007) 85–92. [67] U. Zackrisson, M. Brannstrom, S. Granberg, P.O. Janson, W.P. Collins, T.H.
[43] M. Eftekhar, E. Rahmani, T. Eftekhar, Effect of adding human chorionic Bourne, Acute effects of a transdermal nitric oxide donor on perifollicular and
gonadotropin to the endometrial preparation protocol in frozen embryo intrauterine blood flow, Ultrasound Obstet. Gynecol. 12 (1) (1998) 50–55.
transfer cycles, Int. J. Fertil. Steril. 6 (3) (2012) 175–178. [68] J. Ohl, C. Lefebvre-Maunoury, C. Wittemer, G. Nisand, M.C. Laurent, P.
[44] E.G. Papanikolaou, D. Kyrou, G. Zervakakou, E. Paggou, P. Humaidan, Follicular Hoffmann, Nitric oxide donors for patients undergoing IVF. A prospective,
HCG endometrium priming for IVF patients experiencing resisting thin double-blind, randomized, placebo-controlled trial, Hum. Reprod. 17 (10)
endometrium. A proof of concept study, J. Assist. Reprod. Genet. 30 (10) (2002) 2615–2620.
(2013) 1341–1345. [69] W. Wurfel, C. Santjohanser, K. Hirv, M. Buhl, O. Meri, I. Laubert, et al., High
[45] R. Davar, S. Miraj, Mojtahedi M. Farid, Effect of adding human chorionic pregnancy rates with administration of granulocyte colony-stimulating factor
gonadotropin to frozen thawed embryo transfer cycles with history of thin in ART-patients with repetitive implantation failure and lacking killer-cell
endometrium, Int. J. Reprod. Biomed. (Yazd) 14 (1) (2016) 53–56. immunglobulin-like receptors, Hum. Reprod. 25 (8) (2010) 2151–2152,
[46] H. Qublan, Z. Amarin, M. Al-Qudah, F. Diab, M. Nawasreh, S. Malkawi, et al., author reply 2.
Luteal phase support with GnRH-a improves implantation and pregnancy [70] E. Tehraninejad, F. Davari Tanha, E. Asadi, K. Kamali, E. Aziminikoo, E. Rezayof,
rates in IVF cycles with endometrium of < or = 7 mm on day of egg retrieval, G-CSF Intrauterine for thin endometrium, and pregnancy outcome, J. Family
Hum. Fertil. (Camb). 11 (1) (2008) 43–47. Reprod. Health 9 (3) (2015) 107–112.
[47] C.W. Wang, S.G. Horng, C.K. Chen, H.S. Wang, H.Y. Huang, C.L. Lee, et al., [71] N. Gleicher, A. Vidali, D.H. Barad, Successful treatment of unresponsive thin
Ovulation induction with tamoxifen and alternate-day gonadotrophin in endometrium, Fertil. Steril. 95 (6) (2011) 2123, e13-7.
patients with thin endometrium, Reprod. Biomed. Online 17 (1) (2008) 20– [72] N. Gleicher, A. Kim, T. Michaeli, H.J. Lee, A. Shohat-Tal, E. Lazzaroni, et al., A
26. pilot cohort study of granulocyte colony-stimulating factor in the treatment
[48] K. Reynolds, J. Khoury, J. Sosnowski, J. Thie, G. Hofmann, Comparison of the of unresponsive thin endometrium resistant to standard therapies, Hum.
effect of tamoxifen on endometrial thickness in women with thin Reprod. 28 (1) (2013) 172–177.
endometrium (<7 mm) undergoing ovulation induction with clomiphene [73] D.H. Barad, Y. Yu, V.A. Kushnir, A. Shohat-Tal, E. Lazzaroni, H.J. Lee, et al., A
citrate, Fertil. Steril. 93 (6) (2010) 2091–2093. randomized clinical trial of endometrial perfusion with granulocyte colony-
[49] H. Ke, J. Jiang, M. Xia, R. Tang, Y. Qin, Z.J. Chen, The effect of tamoxifen on thin stimulating factor in in vitro fertilization cycles: impact on endometrial
endometrium in patients undergoing frozen-thawed embryo transfer ( thickness and clinical pregnancy rates, Fertil. Steril. 101 (3) (2014) 710–715.
Reprod. Sci. (2017), 1933719117698580). [74] M. Eftekhar, M. Sayadi, F. Arabjahvani, Transvaginal perfusion of G-CSF for
[50] H.C. Kuo, C.C. Hsu, S.T. Wang, K.E. Huang, Aspirin improves uterine blood flow infertile women with thin endometrium in frozen ET program: a non-
in the peri-implantation period, J. Formos. Med. Assoc. 96 (4) (1997) 253– randomized clinical trial, Iran J. Reprod. Med. 12 (10) (2014) 661–666.
257. [75] Y. Li, P. Pan, X. Chen, L. Li, Y. Li, D. Yang, Granulocyte colony-stimulating factor
[51] L.N. Weckstein, A. Jacobson, D. Galen, K. Hampton, J. Hammel, Low-dose administration for infertile women with thin endometrium in frozen embryo
aspirin for oocyte donation recipients with a thin endometrium: prospective, transfer program, Reprod. Sci. 21 (3) (2014) 381–385.
randomized study, Fertil. Steril. 68 (5) (1997) 927–930. [76] P.R. Amable, R.B. Carias, M.V. Teixeira, I. da Cruz Pacheco, R.J. Correa do
[52] J.H. Check, C. Dietterich, D. Lurie, A. Nazari, J. Chuong, A matched study to Amaral, J.M. Granjeiro, et al., Platelet-rich plasma preparation for
determine whether low-dose aspirin without heparin improves pregnancy regenerative medicine: optimization and quantification of cytokines and
rates following frozen embryo transfer and/or affects endometrial growth factors, Stem Cell Res. Ther. 4 (3) (2013) 67.
sonographic parameters, J. Assist. Reprod. Genet. 15 (10) (1998) 579–582. [77] Y. Chang, J. Li, Y. Chen, L. Wei, X. Yang, Y. Shi, et al., Autologous platelet-rich
[53] Y.Y. Hsieh, H.D. Tsai, C.C. Chang, H.Y. Lo, C.L. Chen, Low-dose aspirin for plasma promotes endometrial growth and improves pregnancy outcome
infertile women with thin endometrium receiving intrauterine insemination: during in vitro fertilization, Int. J. Clin. Exp. Med. 8 (1) (2015) 1286–1290.
a prospective, randomized study, J. Assist. Reprod. Genet. 17 (3) (2000) 174– [78] S. Zadehmodarres, S. Salehpour, N. Saharkhiz, L. Nazari, Treatment of thin
177. endometrium with autologous platelet-rich plasma: a pilot study, JBRA
[54] T.A. Gelbaya, M. Kyrgiou, T.C. Li, C. Stern, L.G. Nardo, Low-dose aspirin for Assist. Reprod. 21 (1) (2017) 54–56.
in vitro fertilization: a systematic review and meta-analysis, Hum. Reprod. [79] M. Fall, A.P. Baranowski, S. Elneil, D. Engeler, J. Hughes, E.J. Messelink, et al.,
Update 13 (4) (2007) 357–364. EAU guidelines on chronic pelvic pain, Eur. Urol. 57 (1) (2010) 35–48.
[55] D. Glujovsky, R. Pesce, G. Fiszbajn, C. Sueldo, R.J. Hart, A. Ciapponi, [80] M.M. Bodombossou-Djobo, C. Zheng, S. Chen, D. Yang, Neuromuscular
Endometrial preparation for women undergoing embryo transfer with electrical stimulation and biofeedback therapy may improve endometrial
M. Eftekhar et al. / Middle East Fertility Society Journal 23 (2018) 1–7 7

growth for patients with thin endometrium during frozen-thawed embryo [92] R.K. Hunter 2nd, C.D. Nevitt, J.T. Gaskins, B.B. Keller, H.C. Bohler Jr., A.J.
transfer: a preliminary report, Reprod. Biol. Endocrinol. 9 (2011) 122. LeBlanc, Adipose-derived stromal vascular fraction cell effects on a rodent
[81] C.E. Gargett, Uterine stem cells: what is the evidence?, Hum Reprod. Update model of thin endometrium, PLoS One 10 (12) (2015) e0144823.
13 (1) (2007) 87–101. [93] F. Alawadhi, H. Du, H. Cakmak, H.S. Taylor, Bone marrow-derived stem cell
[82] C.E. Gargett, H.P. Nguyen, L. Ye, Endometrial regeneration and endometrial (BMDSC) transplantation improves fertility in a murine model of Asherman’s
stem/progenitor cells, Rev. Endocr. Metab. Disord. 13 (4) (2012) 235–251. syndrome, PLoS One 9 (5) (2014) e96662.
[83] I. Cervello, A. Mas, C. Gil-Sanchis, L. Peris, A. Faus, P.T. Saunders, et al., [94] S. Kilic, B. Yuksel, F. Pinarli, A. Albayrak, B. Boztok, T. Delibasi, Effect of stem
Reconstruction of endometrium from human endometrial side population cell application on Asherman syndrome, an experimental rat model, J. Assist.
cell lines, PLoS One 6 (6) (2011) e21221. Reprod. Genet. 31 (8) (2014) 975–982.
[84] J. Zhao, Q. Zhang, Y. Wang, Y. Li, Uterine infusion with bone marrow [95] C. Gil-Sanchis, I. Cervello, S. Khurana, A. Faus, C. Verfaillie, C. Simon,
mesenchymal stem cells improves endometrium thickness in a rat model of Contribution of different bone marrow-derived cell types in endometrial
thin endometrium, Reprod. Sci. 22 (2) (2015) 181–188. regeneration using an irradiated murine model, Fertil. Steril. 103 (6) (2015)
[85] Z. Jing, Z. Qiong, W. Yonggang, L. Yanping, Rat bone marrow mesenchymal 1596–1605, e1.
stem cells improve regeneration of thin endometrium in rat, Fertil. Steril. 101 [96] C.B. Nagori, S.Y. Panchal, H. Patel, Endometrial regeneration using autologous
(2) (2014) 587–594. adult stem cells followed by conception by in vitro fertilization in a patient of
[86] H.S. Taylor, Endometrial cells derived from donor stem cells in bone marrow severe Asherman’s syndrome, J. Hum. Reprod. Sci. 4 (1) (2011) 43–48.
transplant recipients, JAMA 292 (1) (2004) 81–85. [97] N. Singh, S. Mohanty, T. Seth, M. Shankar, S. Bhaskaran, S. Dharmendra,
[87] H. Du, H.S. Taylor, Contribution of bone marrow-derived stem cells to Autologous stem cell transplantation in refractory Asherman’s syndrome: a
endometrium and endometriosis, Stem Cells 25 (8) (2007) 2082–2086. novel cell based therapy, J. Hum. Reprod. Sci. 7 (2) (2014) 93–98.
[88] I. Cervello, C. Gil-Sanchis, A. Mas, A. Faus, J. Sanz, F. Moscardo, et al., Bone [98] X. Santamaria, S. Cabanillas, I. Cervello, C. Arbona, F. Raga, J. Ferro, et al.,
marrow-derived cells from male donors do not contribute to the endometrial Autologous cell therapy with CD133+ bone marrow-derived stem cells for
side population of the recipient, PLoS One 7 (1) (2012) e30260. refractory Asherman’s syndrome and endometrial atrophy: a pilot cohort
[89] H. Du, H. Naqvi, H.S. Taylor, Ischemia/reperfusion injury promotes and study, Hum. Reprod. 31 (5) (2016) 1087–1096.
granulocyte-colony stimulating factor inhibits migration of bone marrow- [99] T. Garrido-Gomez, M. Ruiz-Alonso, D. Blesa, P. Diaz-Gimeno, F. Vilella, C.
derived stem cells to endometrium, Stem Cells Dev. 21 (18) (2012) 3324– Simon, Profiling the gene signature of endometrial receptivity: clinical
3331. results, Fertil. Steril. 99 (4) (2013) 1078–1085.
[90] T. Ikoma, S. Kyo, Y. Maida, S. Ozaki, M. Takakura, S. Nakao, et al., Bone [100] P. Diaz-Gimeno, J.A. Horcajadas, J.A. Martinez-Conejero, F.J. Esteban, P. Alama,
marrow-derived cells from male donors can compose endometrial glands in A. Pellicer, et al., A genomic diagnostic tool for human endometrial
female transplant recipients, Am. J. Obstet. Gynecol. 201 (6) (2009) 608, e1-8. receptivity based on the transcriptomic signature, Fertil. Steril. 95 (1)
[91] S.S. Morelli, P. Rameshwar, L.T. Goldsmith, Experimental evidence for bone (2011) 50–60, e1-15.
marrow as a source of nonhematopoietic endometrial stromal and epithelial [101] F. Cruz, J. Bellver, Live birth after embryo transfer in an unresponsive thin
compartment cells in a murine model, Biol. Reprod. 89 (1) (2013) 7. endometrium, Gynecol. Endocrinol. 30 (7) (2014) 481–484.

You might also like