Sunteți pe pagina 1din 12

Middle East Fertility Society Journal (2017) 22, 1–12

Middle East Fertility Society

Middle East Fertility Society Journal


www.mefsjournal.org
www.sciencedirect.com

REVIEW

Modern management of thin lining


Youssef Mouhayar a, Fady I. Sharara b,c,*

a
Dept of OB/GYN, University of Miami/Jackson Memorial Hospital, Miami, FL, United States
b
Virginia Center for Reproductive Medicine, Reston, VA, United States
c
Dept of OB/GYN, George Washington University, Washington, DC, United States

Received 31 May 2016; revised 23 June 2016; accepted 4 September 2016


Available online 4 October 2016

KEYWORDS Abstract Objective: To define ‘‘thin” endometrium in fertility treatment, and to critically explore
Thin endometrium; the available treatment options.
Endometrial thickness; Design: A review of the scientific literature.
Endometrial pattern; Setting: N/A.
Pregnancy rates; Methods: An electronic literature search pertaining to patients with ‘‘thin” endometrium under-
G-CSF; going fertility treatment was performed through April 2016.
Stem cell therapy Results: Adequate endometrial growth is an integral step in endometrial receptivity and embryo
implantation. Whether idiopathic or resulting from an underlying pathology, a thin endometrium
of <7 mm is linked to a lower probability of pregnancy; however, no reported thickness excludes
the occurrence of pregnancy. Several treatment modalities have been studied and include extended
estrogen, gonadotropin therapy, low-dose hCG, tamoxifen, pentoxifylline, tocopherol, l-arginine,
low-dose aspirin, vaginal sildenafil, acupuncture and neuromuscular electric stimulation, intrauter-
ine G-CSF, and stem cell therapy. All treatment modalities except vaginal sildenafil, intrauterine
GCF, and stem cell therapy were inconsistent in showing significant improvement in pregnancy
rates. Early results of stem cell therapy trials seem promising.
Conclusions: EMT <7 mm is associated with lower probability of pregnancy in ART. Vaginal
sildenafil appears to be a reasonable first line therapy option, and G-CSF appears to be a potential
second option, while stem cell therapy seems to be a promising new treatment modality.
Ó 2016 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/).

* Corresponding author at: Virginia Center for Reproductive Medicine, 11150 Sunset Hills Rd, Suite 100, Reston, VA, United States.
E-mail address: fsharara@vcrmed.com (F.I. Sharara).
Peer review under responsibility of Middle East Fertility Society.

Production and hosting by Elsevier

http://dx.doi.org/10.1016/j.mefs.2016.09.001
1110-5690 Ó 2016 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 Y. Mouhayar, F.I. Sharara

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Materials and methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.1. Hormonal adjustment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.2. Pentoxifylline and tocopherol. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.3. Low-dose aspirin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.4. Acupuncture, neuromuscular electric stimulation and electro-acupuncture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.5. Vaginal sildenafil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.6. Granulocyte colony-stimulating factor (G-CSF). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.7. Stem cell therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8

1. Introduction (18,22,35–58). Moreover, most of the reports used a cutoff


EMT of 7 mm while others used 6 mm, 8 mm, 9 mm, or
Embryo implantation is a very delicate and well-orchestrated 10 mm (18,32–34,38,40,44,46–52,56–60).
process that is governed by the interaction between several Several mechanisms have been proposed to explain the
maternal and embryonic factors, ultimately resulting in adher- underlying pathophysiology of thin endometrium. Intrauterine
ence of the blastocyst to the endometrium (1–5). For a short adhesions, ovarian stimulation with clomiphene citrate (CC),
period of time during the normal menstrual cycle, the endome- as well as prolonged use of progesterone, or combined oral
trium represents the fertile ‘‘soil” for the implanting embryo contraceptive pills have been associated with thin endome-
(6). The human endometrium undergoes complex changes, in trium (61–63). Vascular epithelial growth factor (VEGF),
response to circulating estrogen and progesterone, which cul- which plays a critical role in angiogenesis, appears to be an
minate at the mid-luteal phase of the menstrual cycle when it important factor in the pathophysiology of thin endometrium.
becomes suitable to host the blastocyst (7–9). These changes Adequate endometrial development, which is hormonally
occur at the morphological, biochemical, and molecular levels; mediated, is highly dependent on adequate blood supply.
any faux pas may result in failed implantation (10–13). Identi- Miwa et al. elegantly described that with increased impedance
fying a receptive endometrium is essential; however, an ideal across the radial uterine arteries, there is resultant decrease in
method that can reliably predict endometrial receptivity has VEGF expression and subsequent poor vascular development,
yet to be determined. In clinical practice, high-resolution ultra- resulting in thin endometrium (64).
sonography is routinely used to monitor follicular develop- In 2008, Senturk et al. reviewed thin endometrium in ART
ment and endometrial responsiveness during controlled and the available treatment modalities (extended estrogen
ovarian stimulation. Several ultrasound markers, such as administration, vaginal sildenafil, vitamin E, pentoxifylline,
endometrial thickness (EMT), pattern, blood flow impedance, and luteal phase GnRH-a supplementation), and concluded
and uterine volume have been evaluated for their predictive that these were ineffective (65). More recently, Lebovitz
role of endometrial receptivity; however, they had low speci- et al. concluded that the treatment of ‘‘thin” endometrium
ficity and positive predictive value for detecting a receptive remains a challenge, with only minor improvements achieved
endometrium (14,15). Endometrial patterns, such as a triple- with the currently available treatment modalities (66). A recent
layer endometrium on day of human chorionic gonadotropin systematic review and meta-analysis of 10,724 cases showed
(hCG) trigger or oocyte retrieval and embryo transfer might that EMT as an independent variable is not predictive for
be a better predictor of implantation than endometrial thick- the occurrence of pregnancy (67). This meta-analysis however,
ness (16–25). A certain endometrial development however, is found that the most commonly reported cutoff of 7 mm
an integral part of a receptive endometrium. Despite that a occurred in only 2.4% of the cases (260/10,724), and this cutoff
minimal endometrial thickness of 6 mm has been reported to was associated with a significant drop in the probability of
be essential for achieving implantation in assisted reproductive pregnancy (67). We have elected to review this topic as new
technologies (ART), successful pregnancies were documented treatment modalities emerge. Here we provide a more thor-
with a minimum EMT of 4 mm (26–29). In fact and up to this ough review of the available literature to date, aiming to define
date, there is no consensus on a cutoff value of an EMT below ‘‘thin” endometrium in infertile patients, and to critically ana-
which implantation rates even decline in ART. Using receiver lyze the proposed treatment modalities.
operating characteristics (ROC) area under the curve, a cutoff
limit of endometrial thickness (on day of hCG trigger) above 2. Materials and methods
which implantation could be predicted was not detected by
three reports, whereas two studies reported a threshold thick- We performed a review of the literature on thin endometrium,
ness of 8 mm (30–34). While several reports showed that its pathophysiology, and treatment. Abstracts, case reports,
endometrial thickness has a predictive value for successful original, and review articles were considered. A computer-
pregnancies in ART, others have demonstrated the opposite based systematic literature review was performed through
Modern management of thin lining 3

April 2016. The Cochrane database, PubMed, Medline reg- received 0.1 mg of Triptorelin on days of egg retrieval, embryo
istries, and other online sources were searched using the broad transfer, and 3 days after embryo transfer, while the remaining
terms: thin endometrium, thin endometrium and IVF, thin 60 patients served as the control group. EMT increased from
endometrium and ART, and treatment of thin endometrium. 6.89 ± 0.24 mm to 8.92 ± 1.6 mm in the study group which
The titles and abstracts were screened for relevance, and rele- was significantly higher than the increase noted in the control
vant articles were analyzed in detail to determine which studies group which was from 6.83 ± 0.26 to 7.12 ± 0.45 mm (8.92
could be included in the review. Furthermore, the references ± 1.6 mm vs. 7.12 ± 0.45 mm, P < 0.05). Pregnancy rate
cited in relevant studies and review articles were hand searched was also significantly higher in the study group vs. the control
to identify further relevant studies. Studies were considered eli- group (36.6% vs. 13.7%, P < 0.01) (71).
gible if they were conducted to assess or compare different Adding low-dose human chorionic gonadotropin (hCG)
treatment modalities of thin endometrium. Changes in mean during endometrial preparation with estradiol was investigated
endometrial thickness and pregnancy rates were the two prin- as well. In a pilot study by Papanikolaou et al. seventeen
ciple summary measures, and were independently extracted patients with resistant thin endometrium (<7 mm) during
from individual studies. fresh or frozen donor-embryo cycles were recruited (72). 150
IUs of hCG were administered daily for seven days starting
on days 8 or 9 of estrogen administration. The mean EMT
3. Results increased from an average of 5.18 mm to 6.01 mm
(P = 0.008); however, 5 of 17 patients (29.4%) did not experi-
3.1. Hormonal adjustment ence any improvement in EMT. The pregnancy rate following
treatment was 52.9% (9/17 patients), including pregnancies in
Endometrial proliferation is dependent on serum estrogen two patients who had not improved their EMT after treatment
levels, which results in progressive growth of the functional (72).
endometrium in the proliferative phase. On this basis, patients Clomiphene citrate (CC) has been widely utilized for ovula-
with thin endometrium were offered supplemental exogenous tion induction. Similar to other selective estrogen receptor
estrogen. Studies were mostly in frozen-thawed embryo trans- modulators (SERMs), CC has mixed estrogenic and anti-
fer (FET) cycles and in vitro maturation (IVM). In 2006, Chen estrogenic effects along the hypothalamic-pituitary-gonadal-u
et al. randomized 36 patients with endometrial thickness terine axis (73). In fact, CC’s positive effects in treating infer-
<8 mm to either extend estrogen treatment followed by tility by inducing ovulation are counteracted by its negative
FET, or to proceed directly with embryo transfer (68). The effects on endometrial proliferation (74–76). Tamoxifen
study group received oral estradiol valerate for 14–82 days, fol- (TMX) on the other hand is another SERM that is similar
lowed by embryo transfer when the EMT was greater than to CC but has an estrogen agonist effect at the level of the
8 mm. The endometrial thickness was significantly higher after endometrium, and on this basis it was suggested that TMX
treatment in patients who received the estradiol treatment (8.6 could be used for ovulation induction (77). In a prospective
± 0.7 mm vs. 6.7 ± 0.9 mm, P = 0.031), with a significantly trial, Wang et al. had 131 patients with various infertility diag-
higher pregnancy rate than in patients who proceeded with noses who had failed to develop an EMT of 8 mm (78).
embryo transfer (38.5% vs. 4.3%, P = 0.016). In this study Patients were allocated to either TMX or CC with alternating
however, the EMT was similar in patients who conceived com- gonadotropins. Sixty-one patients (81 cycles) received 40 mg of
pared to those who did not, in both the study and control tamoxifen daily on days 3–9 and 70 patients (82 cycles)
groups (68). Shen et al. presented a case of a 37 year-old received 100 mg CC daily on days 3–7 with alternating hMG
woman with a refractory thin endometrium (<6 mm) who (150 IUs). The TMX group had a significantly thicker EMT
was treated with extended estrogen supplementation for 9 days on day of hCG trigger vs. CC with alternating hMG (10.8
followed by controlled ovarian hyperstimulation, and this ± 2.3 mm vs. 6.7 ± 1.3 mm, P < 0.001), as well as a higher
combination resulted in a live twin birth at 36 weeks (69). clinical pregnancy rate following intrauterine insemination
Poor endometrial development has also been suggested to (32.1% vs. 15.9%, P = 0.015) (78). In 2010, Reynolds and col-
be a reason behind the lower pregnancy rates in IVM cycles. leagues retrospectively reviewed 19 women who had failed to
To improve endometrial development in IVM cycles, Elizur develop an EMT of P7 mm during OI with CC and who were
et al. compared the administration of low-dose human meno- switched to OI with TMX (79). The mean EMT, measured on
pausal gonadotropin (hMG) and micronized 17b-estradiol either day 12 or 14 of the cycle, increased from 5.5 ± 0.8 mm
supplementation in IVM cycles with thin endometrium (70). to 8.8 ± 1.3 mm, P < 0.001 in the TMX treated cycles with a
The authors noted a significant improvement in endometrial pregnancy rate of 42.1% (79). A recent prospective random-
thickness from <6 mm to 7.7 ± 1.8 mm and 7.3 ± 1.7 mm ized trial by Morad et al. compared OI with TMX versus
with low-dose hMG and 17b-estradiol, respectively, without CC plus vaginal sildenafil in 65 patients with EMT < 7 mm
a significant difference between the two groups. There was in a previous OI cycle with CC only (80). Of those, 34 were
no difference in pregnancy rates between the two arms of the randomized to receive TMX 40 mg daily on days 3–7 of the
study, nor was there an untreated control group for compar- cycle, and 31 patients to receive 100 mg of CC daily with
ison (70). 25 mg of vaginal sildenafil four times per day on days 3–7 of
Luteal phase support with gonadotropin-releasing hormone the cycle. EMT, measured on day of hCG trigger, significantly
agonists (GnRH-a) is another form of hormonal manipulation increased in both groups in comparison with the previous CC
that was employed in patients with thin endometrium undergo- only cycles: from 5.7 ± 1.2 to 8.81 ± 1.32, P < 0.05 in TMX
ing IVF. Qublan et al. prospectively randomized 120 patients group and from 5.5 ± 1.4 mm to 9.3 ± 1.2 mm; however,
undergoing IVF with thin endometrium (67 mm), to either there was no difference in pregnancy rates between the two
GnRH-a or placebo during luteal support (71). Sixty patients groups after IUI (18.75% vs. 27.59%, P > 0.05) (80). Most
4 Y. Mouhayar, F.I. Sharara

recently Chen et al. reported three cases of recurrent unrespon- 3.3. Low-dose aspirin
sive thin endometrium (<6 mm) who were successfully treated
with TMX to increase their EMT (81). Patients were undergo- Low-dose aspirin is hypothesized to increase endometrial
ing programmed FET, and TMX was used to simulate blood flow by decreasing impedance across the uterine artery.
endometrial proliferation. Patients were given 20 mg of TMX It has been previously reported that low-dose aspirin results in
daily for 5 days. The endometrial thickness increased to 7.7, lower pulsatility index of the uterine artery and subsequently
7.8, and 8.1 mm in the first, second, and third patient respec- improves pregnancy rates (88). In their report on low-dose
tively, 7–11 days after TMX withdrawal with three clinical aspirin use in frozen-thawed embryo transfers Check and col-
pregnancies and two healthy deliveries (81). leagues noted an increase in mid-luteal phase EMT when com-
pared to controls (89). Weckstein et al. randomized 28 patients
3.2. Pentoxifylline and tocopherol with thin endometrium (<8 mm) in an oocyte donation pro-
gram to either low-dose aspirin (81 mg) with estrogen or estro-
Pentoxifylline (PTX) is a methylxanthine derivative used to gen only (90). There was no significant difference between the
treat vascular diseases. In vivo, it has been reported to increase aspirin group and non-aspirin group (1.6 ± 1.5 mm vs. 0.9
erythrocyte flexibility, to vasodilate, and to inhibit inflamma- ± 0.8, respectively). Despite the increased implantation rate
tory reactions and tumor necrosis factor (TNF) (82). PTX in the aspirin group, there was only a trend for improved clin-
and tocopherol (Vitamin E) combination has been reported ical pregnancy rate in the aspirin treatment group (60% vs.
to improve endometrial thickness in patients with radiation- 31%). Interestingly, in those patients whose endometrial thick-
induced thin endometrium, and was therefore tried in oocyte ness remained less than 8 mm in the two groups, the clinical
recipients with thin endometrium unresponsive to estradiol pregnancy rate was significantly higher for those who received
therapy. In 2002, Lédée-Bataille et al. administered PTX and aspirin (83% vs. 25%, P < 0.05) (90). In another prospective
tocopherol (400 mg and 500 IU respectively twice daily) to randomized trial, patients with thin endometrium (<8 mm)
18 oocyte-donor recipient patients with thin endometria undergoing COH and intrauterine insemination (IUI) were
(66 mm) who had failed to respond to micronized vaginal randomized to either receive 100 mg of aspirin daily starting
estradiol (83). After six months of treatment, the EMT signif- day 1 of the cycle until the pregnancy test, or no aspirin ther-
icantly increased by 1.3 mm ± 1 mm. The pregnancy and apy (91). There were no significant differences in the mean
delivery rates were 33% and 27%, respectively. However, there EMT after treatment between the aspirin group and the non-
was no significant difference in the endometrial thickness aspirin group (7.2 ± 1.8 vs. 5.8 ± 1.4, respectively). The clin-
between those who conceived and those who did not, before ical pregnancy rate however, was significantly higher in the
or after the treatment (83). The mechanism of action of this aspirin group vs. non-aspirin group (18.4% vs. 9.0%,
combined treatment on endometrial thickness and pregnancy P = 0.036) (91).
outcome is unclear; however, it is hypothesized that it inhibits
inflammatory reactions and decreases TNF-alpha levels. In a 3.4. Acupuncture, neuromuscular electric stimulation and
case series by Letur-Konirsch and Delanian, three oocyte- electro-acupuncture
donor recipients with estrogen-resistant thin endometria (mean
EMT 4.9 mm) were treated with combined PTX and toco- Acupuncture, one of the oldest interventions of traditional
pherol (800 mg and 1000 IU respectively) for 9 months (84). Chinese medicine, has been utilized in several obstetrical and
There was an increase in the endometrial thickness to a mean gynecologic conditions, including infertility. Several reports
of 7.4 mm, and 2 of 3 patients conceived (84). Acharya et al. were able to show that acupuncture may improve pregnancy
also administered combined PTX and tocopherol (800 mg rates in ART (92,93). Two other reports were able to show that
and 1000 IU respectively) to 20 infertile patients with thin electro-acupuncture was able to reduce blood flow impedance
endometrium over an average duration of 8.1 months (85). across the uterine artery in infertile patients undergoing IVF
There was a significant increase in EMT at the end of the treat- (94,95). In a prospective, non-randomized trial, Stener-
ment (4.9 ± 1.5 mm vs. 7.4 ± 0.9 mm, P = 0.001) resulting in Victorin was able to demonstrate that electro-acupuncture
a 40% pregnancy rate (85). reduced uterine artery blood flow impedance in 10 infertile
The effects of administering vitamin E alone were also stud- but otherwise healthy patients with baseline impedance (94).
ied. In a prospective observational trial, 600 IU of Vitamin E In a randomized controlled trial (RCT), Ho et al. treated thirty
daily significantly increased the endometrial thickness in infertile patients with electro-acupuncture twice weekly for
52% of patients with an EMT < 8 mm (7.2 mm vs. 8.3 mm, 4 weeks, whereas 14 other patients served as a control group
P < 0.05) (86). In that same trial, L-Arginine (6 grams/day) (95). There was a significant reduction in the uterine artery
treatment was also successful in six out of 9 (67%) patients blood flow impedance in the study compared to the control
(7.4 mm vs. 8.0 mm, P < 0.05). In a prospective randomized group, without a significant difference in pregnancy rates
controlled trial by Cicek et al. 400 IU of vitamin E was admin- between the two groups (95). In another recent single-
istered to patients with unexplained infertility during con- blinded randomized controlled trial, Shuai et al. treated
trolled ovarian hyperstimulation (COH) (87). There was a patients undergoing FET, with either transcutaneous electrical
significant increase in EMT to 9.6 ± 2.1 mm vs. 8.2 acupuncture point stimulation (TEAS) or mock TEAS. EMT
± 2.0 mm in patients who received vitamin E with clomiphene on day of hCG trigger was similar between the two groups
citrate for ovulation induction compared to those who did not (11.26 ± 1.5 mm vs. 10.74 ± 1.54 mm), whereas the study
(P = 0.001). However, there were no significant differences in group had a higher number of triple-line endometrium
implantation rates and ongoing pregnancy rates between the (91.2% vs. 41.2%, P = 0.002), and a higher clinical pregnancy
two groups (87). rate (44.1% vs. 20.6%, P = 0.038) (96). However, as far as we
Modern management of thin lining 5

know, there have not been any studies on improving endome- 8.9 mm, and the second patient from 5.0 mm to 6.6 mm. Both
trial thickness using acupuncture. patients had a full term healthy singleton deliveries (101). In
Pelvic floor neuromuscular electrical stimulation (NMES) another trial by Takasaki et al. vaginal sildenafil significantly
has been used in patients with thin lining. In their preliminary increased the EMT in 11 out of 12 patients (92%) from
report, Bodombossou-Djobo et al. evaluated 41 subjects with 7.1 mm to 9.4 mm (P < 0.01), with a 50% pregnancy rate after
an EMT 6 7 mm who failed to conceive in two prior ART 12 IVF cycles (102). Similar positive effects on improving
cycles (97). Twenty had NMES in their subsequent FET cycle, EMT were also noted by Morad and colleagues where 31
whereas 21 proceeded directly with FET. NMES was applied patients experienced significant improvement in EMT from
daily for a total of 3–4 days starting on day 9 of the stimula- 5.5 ± 1.4 mm to 9.3 ± 1.2 mm (80). Most recently, Eid et al.
tion. EMT significantly increased in the treatment group com- administered vaginal sildenafil to 22 patients with an EMT
pared to the control group (7.93 ± 1.42 mm vs. 6.78 of 7 mm and elevated pulsatility index (PI > 0.3) for a total
± 0.47 mm, P = 0.002) but without a significant difference of 7 days between ovulation trigger and embryo transfer
in the clinical pregnancy rates between the two groups (97). (103). Sixty-eight percent (15/22) of patients experienced
improvement in EMT and a decrease in their PI. Implantation
3.5. Vaginal sildenafil rates and pregnancy rates were higher in patients who
responded to treatment (26% vs. 7%, and 40% vs. 14%,
Sildenafil citrate is phosphodiesterase-5 inhibitor that respectively) (103).
enhances the vasodilatory effects of nitric oxide, which in turn
increases subendometrial blood flow. In 2000, Sher et al. 3.6. Granulocyte colony-stimulating factor (G-CSF)
reported four patients who had previously had an EMT
between 5–7 mm in a prior failed IVF or IUI cycles, and G-CSF, initially described as a hematopoietic growth factor,
who were all given vaginal sildenafil 25 mg four times per has been shown to have important functions in nonhematopoi-
day for 8–12 days during COH (98). The EMT measured on etic cells, including the endometrium, and has a potential role
day of hCG administration and was found to be between 8 in promoting early endometriotic lesions in a murine model
and 12 mm (98). The same four patients had also undergone (104). Gleicher et al. hypothesized that G-CSF might have a
a prior mock cycle with leuprolide acetate and received the direct role promoting endometrial growth, and reported a case
same daily dose of vaginal sildenafil for 7 days, then 7 days series of 4 patients with thin endometria between 3 and 6.5 mm
of placebo followed by vaginal sildenafil and estradiol for who failed to improve with oral and vaginal estrogen as well as
another 7 days. The pulsatility index decreased after treatment with vaginal sildenafil (in one of the patients) (105). All four
with sildenafil, but was back to baseline after 7 days of pla- patients had intrauterine G-CSF infusion (300 lg) 2–9 days
cebo, indicating increased diastolic blood flow after sildenafil before ET, and had a significant increase to at least 7 mm
administration (98). Two years later the same group adminis- within 48 h. All four patients conceived, one of which had an
tered vaginal sildenafil (25 mg, 4 times daily) on days 3 ectopic (105). Two years later, the same group performed a
through 10 in 105 patients undergoing IVF who had failed prospective pilot cohort study of 21 patients undergoing IVF
to develop an EMT of 9 mm in prior failed IVF cycles (99). who had EMT < 7 mm on the day of hCG trigger despite oral
The patients had various causes leading to poor endometrial and vaginal estrogen as well as vaginal sildenafil (106). All
development, including pregnancy-related endometritis, DES patients received 300 lg of G-CSF intrauterine infusion 6–
anomalies, fibroids, adenomyosis, and idiopathic. Seventy- 12 h prior to hCG trigger. A second infusion was needed in
three patients (70%) experienced an increase in their EMT to only 3 of 21 patients (14.3%). Overall, EMT increased from
greater than 9 mm, and had a significantly higher implantation an average of 6.4 ± 1.4 mm to 9.3 ± 2.1 mm (P < 0.001)
and ongoing pregnancy rates when compared to those who after G-CSF treatment, but did not differ between those who
failed to respond (29% vs. 2%, P < 0.01, and 45% vs. 0%, conceived and those who did not. The overall clinical preg-
P < 0.01 respectively). The overall ongoing pregnancy rate nancy rate in the cohort was 19.1% (106).
in that cohort was 31.4% (33/105) (99). In a smaller trial by Lucena et al. reported a patient with thin endometrium
Check et al. 16 patients who had failed to attain an EMT of (<5.7 mm) during an in vitro maturation (IVM) cycle who
8 mm during FET cycles were assigned to either vaginal silde- was given 300 lg of intrauterine G-CSF on day of oocyte
nafil or vaginal estradiol in addition to oral estradiol (100). retrieval (107). The EMT subsequently improved to 8.9 mm,
Nine patients received 25 mg of vaginal sildenafil four times and embryo transfer resulted in a full term delivery (107).
daily from days 3 to 9 with no significant change in their Check and colleagues reported a patient who had multiple
EMT after treatment (6.3–6.4 mm). Of the remaining seven IVF-ET and FET cycle failures due to ‘‘thin” endometrium
patients who received vaginal estradiol, 6 were previously of 6 mm despite treatment with oral and vaginal estrogen as
given vaginal sildenafil without improvement in EMT. The well as vaginal sildenafil (108). The patient was given intrauter-
pregnancy rate was only 16.6% (1 out of 6 attempted FET) ine G-CSF as described by Gleicher et al. (105); however, her
(100). EMT was only 5 mm and had a negative pregnancy test after
Asherman’s syndrome (AS) is another cause of suboptimal embryo transfer (108). Li et al. then reported on 69 patients
endometrial development that can lead to subfertility. Zinger who failed to develop an EMT greater than 7 mm in FET
et al. successfully treated two cases with subfertility and thin cycles despite escalating endometrial preparation protocols
endometrium (<7 mm) during previous stimulated cycles (oral (109). Fifty-nine patients were included in this retrospective
estradiol or clomiphene citrate) (101). Vaginal sildenafil was analysis, of which 34 opted for G-CSF treatment (28 refused),
administered for 8–15 days during the treatment cycles. One and received intrauterine G-CSF (100 lg) on the day of ovula-
patient experienced an increase in EMT from 6.5 mm to tion, or day of progesterone start, or day of hCG administra-
6 Y. Mouhayar, F.I. Sharara

tion (109). The cycle cancelation rate due to thin endometrium interestingly, male origin BMDSCs were capable of composing
was lowest in the G-CSF treatment group (69.39% vs. 48.75% the endometria of female bone marrow transplant recipients,
vs. 17.5%, P < 0.05 self-controlled vs. control vs. treatment further indicating their role in endometrial injury repair
groups, respectively), and there was a trend toward better (119,120). In their elegant study Cervello et al. recently demon-
implantation (15.8% vs. 7.89%), and clinical pregnancy rates strated that BMDSCs do not associate with endometrial side
(30.30% vs. 20.0%) in the G-CSF group compared to both cell population, but rather they exert their regenerative poten-
control groups (109). Kunicki et al. administered intrauterine tial in a paracrine fashion, ultimately stimulating endometrial
G-CSF (100 lg) to 37 patients who failed to develop an side cell population (121). The role of BMDSC in endometrial
EMT of 7 mm on day of hCG trigger during prior IVF cycles. regeneration was further demonstrated in several murine mod-
In this prospective study, intrauterine G-CSF was given 6–12 h els (122–124). In fact, uterine ischemia/reperfusion injury
prior to hCG trigger (based on Gleicher’s earlier report), which results in a 2-fold increase in bone marrow-derived stem cell
resulted in a significant increase in EMT from 6.74 recruitment to the endometrium (125). This recruitment seems
± 1.75 mm, to 8.42 ± 1.73 (P < 0.001) within 72 h (110). to be independent of G-CSF and only serves in uterine repair
The clinical pregnancy rate was 18.9%; however, there was after injury rather than monthly cyclic regeneration of the
no difference in the EMT between patients who conceived endometrium (125).
and those who did not (110). The popularity of the regenerative potential of bone mar-
Recently, Barad et al. tried to expand the use of G-CSF on row stem cells led researchers to investigate their effects in
endometrial thickness for all patients undergoing IVF or FET, the treatment of Asherman’s syndrome and thin endometrium.
regardless of endometrial thickness. The study group received Alwadhi and colleagues administered BMDSCs to female mice
300 lg/cc G-CSF (on the day of HCG trigger), while the con- (via tail veins) with AS which were later bred after 3 estrous
trol group received placebo (saline) (111). The EMT increased cycles (126). 9 of 10 treated mice conceived whereas only 3
significantly in the entire cohort by approximately 1.36 mm of 10 non-treated mice conceived in the non-transplanted mice
without a difference between the G-CSF and control groups, vs. 10 of 10 in the control group (126). In a similar study, Kilic
with similar clinical pregnancy rates (111). et al. induced AS in Wistar albino rats and later treated them
In the most recent trial to date, Xu et al. prospectively ran- with either mesenchymal stem cells (MSCs) or oral estrogen or
domized 30 patients with EMT < 7 mm during frozen-thawed combined MSC and oral estrogen (127). All treatment groups
embryo transfer cycles to either intrauterine G-CSF or G-CSF demonstrated a decrease in fibrosis when compared to control,
with endometrial scratch, with 52 patients serving as the con- mostly noted with combined MSC and estrogen treatment
trol group (112). G-CSF was administered ‘‘on the day that (127).
one follicle became dominant (12 mm)”. The EMT increased To further elucidate the role of BMDSCs in regenerating
significantly after treatment (D 3.9 ± 2.0 mm, P < 0.001) thin endometrium, Zhao et al. compared thin endometria
without a difference between the two subgroups (G-CSF vs. infused with bone marrow mesenchymal stem cells (BMSCs)
G-CSF + scratch), and the implantation and clinical preg- to controls and normal rats (128,129). The treatment groups
nancy rates were significantly higher in both treatment sub- showed significant increase in EMT compared to the control
groups compared to the control group (31.5% vs. 13.9%, groups in both trials respectively (325.35 ± 75.51 lm vs.
P < 0.01, and 48.1% vs. 25.0%, P = 0.038 respectively) (112). 187.53 ± 34.38 lm, P < 0.05 and 359.13 ± 49.70 lm vs.
187.53 ± 34.38 lm, P < 0.05) (128,129).
A recent comparative study analyzed the role of different
3.7. Stem cell therapy BM-derived cell subtypes in endometrial regeneration (130).
Various subtypes of BM-derived cell were injected into tail
Multiple evidence, some dating back to the early 1980s, sup- veins of a total body irradiated murine model. Freshly isolated
ports the presence of endometrial stem/progenitor cells in the unfractionated BM cells, hematopoietic progenitor cells,
basalis and functionalis layers of the human endometrium endothelial progenitor cells (EPCs), mesenchymal stem cells,
(113–116). With the tremendous regenerative capacity of the and in vitro cultured mouse Oct4+ BM-derived hypoplast-
endometrial lining during each estrous cycle, it was hypothe- like stem cells supported endometrial regeneration (130).
sized that these stem/progenitor cells play an important role The first human application was in 2011, in a case of thin
in endometrial regeneration. Despite their discovery over endometrium (3.6 mm) secondary to AS refractory to estradiol
30 years ago, the regenerative capacity of endometrial stem treatment (131). Autologous endometrial angiogenic stem cells
cells was just recently proven when Cervello and colleagues were infused into the uterine cavity and was followed by high
demonstrated that human endometrial adult stem cells are able dose estradiol valerate, aspirin (75 mg PO daily), and four
to generate human endometrium after transplantation in cycles of cyclical estrogen and progesterone therapy to finally
NOD-SCID mice renal capsules (117). The role of stem cells reach an EMT of 7.1 mm. Three donor oocyte embryos were
in endometrial regeneration is not limited to local endometrial transferred resulting in a single viable intrauterine pregnancy
progenitor cells; in fact, hematopoietic and non-hematopoietic at 8 weeks as detected by ultrasonography (131). This was fol-
bone marrow-derived stem cells (BMDSCs) are recruited to lowed by a case series of 6 patients with refractory AS who
the endometrium in response to injury. Taylor showed that were treated with autologous mononuclear stem cells implan-
BMDSCs play a role in the regeneration of endometrial stro- tation. The mean EMT significantly increased from a pretreat-
mal and epithelial layers of bone marrow transplant patients ment measurement of 1.38 ± 0.39 mm to 4.05 ± 1.4, 5.46
(118). These findings were further affirmed by other investiga- ± 1.36, and 5.48 ± 1.14 mm at 3, 6, and 9 months respectively
tors, who showed that CD 45 + hematopoietic progenitor (P < 0.05) (132). Cervello et al. investigated whether human
cells are able to colonize the uterine epithelium and play an CD 133 + BMDSCs would promote endometrial growth in
important role in uterine epithelial regeneration; even more a murine model of Asherman’s syndrome (133). The authors
Modern management of thin lining 7

found that the BMDSCs engrafted around small endometrial with or without Asherman’s syndrome. Extended estrogen
vessels in all damage horns and resulted in a significant was beneficial in improving EMT and implantation rates in
increase in epithelial gland cells proliferation via paracrine some, but not all, patients with thin endometrium due to speci-
fashion by up-regulation of thrombospondin 1 and down- fic etiologies such as endometritis and multiple curettages. This
regulation of insulin-like growth factor 1 (133). was particularly helpful for patients undergoing IVM with thin
In a recent human pilot trial, Santamaria and colleagues endometrium, where extended vaginal estrogen and low dose
infused autologous CD 133 + bone marrow-derived stem cells hMG were beneficial in improving endometrial thickness or
(BMDSCs) into the spiral arterioles of 11 patients with refrac- pregnancy rates. However, these studies were limited by small
tory Asherman’s syndrome and 5 patients with refractory sample size and the fact that EMT was similar between
endometrial atrophy (134). An increase in endometrial thick- patients who conceived and those who did not; thus, it is dif-
ness lasting up to six months was noted in patients with AS ficult to draw a positive conclusion regarding the benefits of
(4.3–6.7 mm) and those with refractory atrophic endometrium the prolonged estrogen treatment in the absence of further sup-
(4.2–5.7 mm), with subsequent conception attempts resulting porting results. Two lone studies, one using luteal GnRH-a
in three spontaneous pregnancies (2, 4, and 19 months after support and another using low-dose HCG in programmed
treatment) and seven positive pregnancies after 14 embryo FET cycles seem promising; however, data are lacking to draw
transfers (134). solid recommendations (71,72). Tamoxifen citrate, as an ovu-
lation induction agent, seems to be a good alternative to clomi-
4. Discussion phene citrate in patients who have poor endometrial
development during COH with IUI or FET (78–81). Treat-
‘‘Thin endometrium” occurs not uncommonly in clinical prac- ment with Tamoxifen however may be accompanied with
tice and poses a frustrating challenge to both patients and undesired side effects such as hot flashes, and the possibility
physicians. It is not exactly clear how thin endometrium results of increased risk of endometrial cancer and adenomyosis
in lower chances of pregnancy; however, hypotheses such the (78). Long courses of PTX and tocopherol or tocopherol-
harmful role of reactive oxygen species (ROS) or possibly only were used in oocyte-donor recipient patients, unexplained
not enough ‘‘soil” to sustain the ‘‘seed” have been suggested. infertility, and patients who received radiotherapy. Although
The functional layer of the endometrium undergoes several some trials noted a significant improvement in EMT, there
changes during the menstrual cycle relative to the basal layer, was no evidence of a significant improvement in pregnancy
which homes the larger-caliber oxygen-rich spiral arteries. It is rates, therefore not justifying the lengthy treatment protocol.
hypothesized that, with thin endometrium, the proximity to Aspirin supplementation on the other hand had no beneficial
the ROS-rich basal layer is detrimental for embryo develop- role in oocyte-donor recipient patients, but it showed improve-
ment and implantation (66). Clinicians are faced with the chal- ment in pregnancy rates without improvement in EMT in
lenge of where to draw the cutoff, and how and whether to patients with thin endometrium undergoing IUI. This
treat it. The most widely accepted measurement is 7 mm; how- improvement in pregnancy rates however, did not reach nor-
ever, this cutoff is not absolute, as pregnancies have been mal expected levels for COH and IUI. Electro-acupuncture
reported at much lower values (27–29). More importantly, significantly decreased blood flow impedance across the uter-
endometrial thickness alone does not seem to have a strong ine arteries but had no effects on pregnancy rates in IVF
predictive capacity for the occurrence of pregnancies in IVF cycles, and none of the conducted trials specifically addressed
cycles, but more so a factor for the assessment of conception patients with thin lining (94–96). While pelvic floor NMES
probability which significantly drops below a thickness of with biofeedback significantly improved EMT, it showed no
7 mm (63). Other key factors such as endometrial pattern play benefit in improving pregnancy rates (97).
a vital role in successful implantation as well, and may be more Vaginal sildenafil administration was beneficial for a signif-
important than thickness. In fact, in the setting of CCS-tested icant percentage of patients with various infertility diagnoses
euploid embryos, different endometrial patterns appear to who had two prior failed IVF cycles or of those who are under-
have a better correlation with the likelihood of pregnancy than going ICSI. The implantation and pregnancy rates were signif-
thin endometrium (25). Specifically, a mid to late secretory icantly better in those patients who responded, indicating that
stage endometrial type (type 3 endometrium) at time of ovula- vaginal sildenafil could be a reasonable first line treatment
tion trigger, had a negative correlation with implantation rates option. However these findings could be limited by the retro-
(25). Furthermore, a trilaminar endometrial pattern on spective nature of the largest study supporting them.
embryo transfer day might be a better prognostic factor of Treatment with intrauterine G-CSF also received its share
cycle outcomes than endometrial thickness in IVF/ICSI cycles of interest despite its significant cost, but the majority of trials
(24). New diagnostic tools are emerging to aid in objectively showed beneficial effects for G-CSF in patients with thin endo-
assessing endometrial receptivity. According to the initial find- metrium. In fact, out of the seven reviewed papers, a case
ings by Mahajan, the endometrial receptivity array (ERA) is report and one retrospective analysis in FET cycles did not
an accurate test in determining the window of implantation. demonstrate a positive effect of G-CSF on improving EMT
In fact 75% of patients (n = 13) with an EMT 6 6 mm in that in patients with thin lining (105–110,112). On the other hand,
study were found to have a receptive endometrium according there is no evidence to support G-CSF administration to all
to the customized endometrial receptivity microarray test patients undergoing IVF or FET (111). While the use of GCSF
(135). ERA however is an invasive and costly test with early seems promising in increasing EMT and possibly pregnancy
results that must be validated with larger studies. rates, most studies suffer from small sample sizes, and different
Over the years, multiple treatment modalities have been studies used different doses and time points when G-CSF was
studied in hopes of improving refractory thin endometrium administered, making interpretation rather difficult. Larger
8 Y. Mouhayar, F.I. Sharara

prospective, randomized, placebo-controlled, trials are there- (6) Salamonsen LA, Nie G, Hannan NJ, Dimitriadis E. Society for
fore sorely needed. reproductive biolosy founders’ lecture 2009. Preparing fertile
The most promising evolving treatment modality in refrac- soil: the importance of endometrial receptivity. Reprod Fertil
tory cases appears to be stem cell therapy, as the administra- Dev 2009;21:923–34.
(7) Strowitzki T, Germeyer A, Popovici R, von Wolff M. The
tion of intrauterine angiogenic endometrial cells improved
human endometrium as a fertility-determining factor. Hum
the endometrial lining of patients with Asherman’s syndrome Reprod Update 2006;12:617–30.
or refractory thin lining. Murine models showed promising (8) Achache H, Revel A. Endometrial receptivity markers, the
results when bone marrow mesenchymal stem cells administra- journey to successful embryo implantation. Hum Reprod
tion restored endometrial thickness to normal after total body Update 2006;12:731–46.
irradiation. Research is also developing to determine what (9) Paulson RJ. Hormonal induction of endometrial receptivity.
specific bone marrow-derived stem cell subtypes will have an Fertil Steril 2011;96:530–5.
impact on endometrial regeneration. In an early human trials, (10) Simón C, Landeras J, Zuzuarregui JL, Martı́n JC, Remohı́ J,
autologous bone marrow-derived stem cells gave promising Pellicer A. Early pregnancy losses in in vitro fertilization and
results in restoring endometrial thickness for a period of oocyte donation. Fertil Steril 1999;72(6):1061–5.
(11) Pellicer A, Rubio C, Vidal F, Minguez Y, Gimenez C, Egozcue
6 months in patients with Asherman’s syndrome or refractory
J, et al. In vitro fertilization plus preimplantation genetic
thin endometrium, with excellent subsequent pregnancy rates, diagnosis in patients with recurrent miscarriage: an analysis of
making stem therapy a potentially valuable option for patients chromosome abnormalities in human preimplantation embryos.
who fail traditional treatment options (132,134). It is impor- Fertil Steril 1999;71(6):1033–9.
tant to note however, that stem cell therapy treatment is inva- (12) Edwards RG. Human implantation: the last barrier in assisted
sive requiring a bone marrow biopsy and interventional reproduction technologies? Reprod Biomed Online 2007;14
radiology assistance for injection into the uterine arterioles. (6):5–22.
In conclusion, a receptive endometrium plays a critical role (13) Revel A. Defective endometrial receptivity. Fertil Steril 2012;97
in embryo implantation, and adequate endometrial growth is (5):1028–32.
essential to this process. Poor endometrial development is (14) Makker A, Singh MM. Endometrial receptivity: clinical assess-
ment in relation to fertility, infertility, and antifertility. Med Res
associated with a decreased probability of pregnancy; yet, it
Rev 2006;26:699–746.
is not the sole predictor of pregnancy occurrence, and (15) Friedler S, Schenker JG, Herman A, Lewin A. The role of
endometrial pattern may be more critical. Among multiple ultrasonography in the evaluation of endometrial receptivity
available treatment options, vaginal sildenafil during the stim- following assisted reproductive treatments: a critical review.
ulation cycle appears to be a reasonable first line treatment Hum Reprod Update 1996;2(4):323–35.
option, whereas intrauterine G-CSF infusion before ovulation (16) Sher G, Herbert C, Maassarani G, Jacobs MH. Assessment of
trigger could be a second line treatment option, provided the late proliferative phase endometrium by ultrasonography in
large randomized studies evaluating outcomes including when patients undergoing in-vitro fertilization and embryo transfer
is the best time point in a cycle (and at what dose) to admin- (IVF/ET). Hum Reprod 1991;6:232–7.
ister G-CSF are performed. Stem cell therapy appears to be (17) Check JH, Nowroozi K, Choe J, Dietterich C. Influence of
endometrial thickness and echo patterns on pregnancy rates
promising in refractory cases; however, significantly more
during in vitro fertilization. Fertil Steril 1991;6:1173–5.
research on safety, effectiveness, and cost, is needed before (18) Dickey RP, Olar TT, Curole DN, Taylor SN, Rye PH.
this modality becomes adopted in the treatment of this frus- Endometrial pattern and thickness associated with pregnancy
trating condition. outcome after assisted reproduction technologies. Hum Reprod
1991;7:418–21.
Conflict of interest (19) Fleischer AC, herbert CM, Hill GA, Kepple DM, Worrell JA.
Transvaginal sonography of the endometrium during induced
cycles. J Ultrasound Med 1991;10:93–5.
The authors declared that there is no conflict of interest. (20) Check JH, Lurie D, Dietterich C, Callan C, Baker A. Adverse
effect of a homogeneous hyperechogenic endometrial sono-
References graphic pattern, despite adequate endometrial thickness on
pregnancy rates following in-vitro fertilization. Hum Reprod
(1) Simón C, Moreno C, Remohı́ J, Pellicer A. Molecular interac- 1993;8:1293–6.
tions between embryo and uterus in the adhesion phase of (21) Oliviera JBA, Baruffi RLR, Mauri AL, Petersen CG, Campos
human implantation. Hum Reprod 1998;13(Suppl 3):219–32. MS, Franco JG. Endometrial ultrasonography as a predictor of
(2) Simón C, Martı́n JC, Galan A, Valbuena D, Pellicer A. pregnancy in an in-vitro fertilization programme. Hum Reprod
Embryonic regulation in implantation. Semin Reprod Endocri- 1993;8:1312–5.
nol 1999;17:267–74. (22) Coulam CB, Bustillo M, Soenksen DM, Britten S. Ultrasono-
(3) Simón C, Martin JC, Pellicer A. Paracrine regulators of graphic predictors of implantation after assisted reproduction.
implantation. Baillieres Best Pract Res Clin Obstet Gynecol Fertil Steril 1994;62:1004–10.
2000;14:815–26. (23) Sharara FI, Lim J, McClamrock HD. Endometrial pattern on
(4) Paria BC, Lim H, Das SK, Reese J, Dey SK. Molecular signaling the day of oocyte retrieval is more predictive of implantation
in uterine receptivity for implantation. Semin Cell Dev Biol success than the pattern or thickness on the day of hCG
2000;11:67–76. administration. J Assist Reprod Genet 1999;16:523–8.
(5) Paiva P, Hannan NJ, Hincks C, Meehan KL, Pruysers E, (24) Detti L, Yelian FD, Kruger ML, Diamond MP, Puscheck EE.
Dimitriadis E, et al. Human chorionic gonadotrophin regulates Endometrial thickness dynamics and morphologic characteris-
FGF2 and other cytokines produced by human endometrial tics during pituitary downregulation with antagonists in assisted
epithelial cells, providing a mechanism for enhancing endome- reproductive technology cycles. J Ultrasound Med
trial receptivity. Hum Reprod 2011;26:1153–62. 2008;27:1591–6.
Modern management of thin lining 9

(25) Gingold Ja, Lee Ja, Rodriguez-Purata J, Whitehouse MC, (44) Richter KS, Bugge KR, Bromer JG, Levy MJ. Relationship
Sandler B, Grunfeld L, et al. Endometrial pattern, but not between endometrial thickness and embryo implantation, based
endometrial thickness, affects implantation rates in euploid on 1294 cycles of in vitro fertilization with transfer of two
embryo transfers. Fertil Steril 2015;104(3):620–8. blastocyst-stage embryos. Fertil Steril 2007;87(1):53–9.
(26) Shapiro H, Cowell C, Casper R. The use of vaginal ultrasound (45) El-Toukhy T, Coomarasamy A, Khairy M, Sunkara K, Seed P,
for monitoring endometrial preparation in a donor oocyte Khalaf Y, et al. The relationship between endometrial thickness
program. Fertil Steril 1993;59(5):1055–8. and outcome of medicated frozen embryo replacement cycles.
(27) Sundström P. Establishment of a successful pregnancy following Fertil Steril 2008;89:832–9.
in-vitro fertilization with an endometrial thickness of no more (46) Dechaud H, Bessueille E, Bousquet P-J, Reyftmann L,
than 4 mm. Hum Reprod 1998;13(6):1550–2. Hamamah S, Hedon B. Optimal timing of ultrasonographic
(28) Check JH, Dietterich C, Check ML, Katz Y. Successful delivery and Doppler evaluation of uterine receptivity to implantation.
despite conception with a maximal endometrial thickness of 4 Reprod Biomed Online 2008;16(3):368–75.
mm. Clin Exp Obstet Gynecol 2003;30(2–3):93–4. (47) Bozdag G, Esinler I, Yarali H. The impact of endometrial
(29) Dix E, Check JH. Successful pregnancies following embryo thickness and texture on intracytoplasmic sperm injection
transfer despite very thin late proliferative endometrium. Clin outcome. J Reprod Med 2009;54(5):303–11.
Exp Obstet Gynecol 2010;37:15–6. (48) Traub ML, Van Arsdale A, Pal L, Jindal S, Santoro N.
(30) Yaman C, Ebner T, Sommergruber M, Pölz W, Tews G. Role of Endometrial thickness, Caucasian ethnicity, and age predict
three-dimensional ultrasonographic measurement of endome- clinical pregnancy following fresh blastocyst embryo transfer: a
trium volume as a predictor of pregnancy outcome in an IVF-ET retrospective cohort. Reprod Biol Endocrinol 2009;7:33.
program: a preliminary study. Fertil Steril 2000;74(4):797–801. (49) Okohue JE, Onuh SO, Ebeigbe P, Shaibu I, Wada I, Ikimalo JI,
(31) Rashidi BH, Sadeghi M, Jafarabadi M, Nejad ES Tehrani. et al. The effect of endometrial thickness on in vitro fertilization
Relationships between pregnancy rates following in vitro fertil- (IVF)-embryo transfer/intracytoplasmic sperm injection (ICSI)
ization or intracytoplasmic sperm injection and endometrial outcome. Afr J Reprod Health 2009;13(1):113.
thickness and pattern. Eur J Obs Gynecol Reprod Biol 2005;120 (50) Kuc P, Kuczynska A, Topczewska M, Tadejko P, Kuezynski W.
(2):179–84. The dynamics of endometrial growth and the triple layer
(32) Al-Ghamdi A, Coskun S, Al-Hassan S, Al-Rejjal R, Awartani appearance in three different controlled ovarian hyperstimula-
K. The correlation between endometrial thickness and outcome tion protocols and their influence on IVF outcomes. Gynecol
of in vitro fertilization and embryo transfer (IVF-ET) outcome. Endocrinol 2011;27(11):867–73.
Reprod Biol Endocrinol 2008;6:37. (51) Singh N, Bahadur A, Mittal S, Malhotra N, Bhatt A. Predictive
(33) Basir GS, So WWK, Ng EHY, Ho PC. Evaluation of cycle-to- value of endometrial thickness, pattern and sub-endometrial
cycle variation of endometrial responsiveness using transvaginal blood flows on the day of hCG by 2D doppler in in-vitro
sonography in women undergoing assisted reproduction. Ultras fertilization cycles: a prospective clinical study from a tertiary
Obstet Gynecol 2002;19(5):484–9. care unit. J Hum Reprod Sci 2011;4:29–33.
(34) McWilliams GD, Frattarelli JL. Changes in measured endome- (52) Zhao J, Zhang Q, Li Y. The effect of endometrial thickness and
trial thickness predict in vitro fertilization success. Fertil Steril pattern measured by ultrasonography on pregnancy outcomes
2007;88(1):74–81. during IVF-ET cycles. Reprod Biol Endocrinol 2012;10(1):100.
(35) Gonen Y, Casper RF, Jacobson W, Blankier J. Endometrial (53) Schild RL, Indefrei D, Eschweiler S, Van der Ven H, Fimmers
thickness and growth during ovarian stimulation: a possible R, Hansmann M. Three-dimensional endometrial volume cal-
predictor of implantation in in vitro fertilization. Fertil Steril culation and pregnancy rate in an in-vitro fertilization pro-
1989;52:446–50. gramme. Hum Reprod 1999;14:1255–8.
(36) Alam V, Bernardini L, Gonzales J, Asch RH, Balmaceda JP. A (54) De Geyter C, Schmitter M, De Geyter M, Nieschlag E,
prospective study of echographic endometrial characteristics and Holzgreve W, Schneider HP. Prospective evaluation of the
pregnancy rates during hormonal replacement cycles. J Assist ultrasound appearance of the endometrium in a cohort of 1.186
Reprod Genet 1993;10:215–9. infertile women. Fertil Steril 2000;1:106–13.
(37) Abdalla HI, Brooks AA, Johnson MR, Kirkland A, Thomas A, (55) Kupesic S, Bekavac I, Bjelos D, Kurjak A. Assessment of
et al. Endometrial thickness: a predictor of implantation in ovum endometrial receptivity by transvaginal color Doppler and three-
recipients? Hum Reprod 1994;9:363–5. dimensional power Doppler ultrasonography in patients under-
(38) Rinaldi L, Lisi F, Floccari A, Lisi R, Pepe G, Fishel S. going in vitro fertilization procedures. J Ultrasound Med
Endometrial thickness as a predictor of pregnancy after in-vitro 2001;20:125–34.
fertilization but not after intracytoplasmic sperm injection. Hum (56) Dietterich C, Check JH, Choe JK, Nazari A, Lurie D. Increased
Reprod 1996;11(7):1538–41. endometrial thickness on the day of human chorionic gonado-
(39) Isaacs Jr JD, Wells CS, Williams DB, Odem RR, Gast MJ, tropin injection does not adversely affect pregnancy or implan-
Strickler RC. Endometrial thickness is a valid monitoring tation rates following in vitro fertilization-embryo transfer.
parameter in cycles of ovulation induction with metropins alone. Fertil Steril 2002;77(4):781–6.
Fertil Steril 1996;65:262–6. (57) Aboulghar MM. Three dimensional endometrial volume versus
(40) Kovacs P. The effect of endometrial thickness on IVF/ICSI endometrial thickness measurement in prediction of IVF/ICSI
outcome. Hum Reprod 2003;18(11):2337–41. outcome. Middle East Fertil Soc J 2005;10(1):63–7.
(41) Zenke U, Chetkowski RJ. Transfer and uterine factors are the (58) Kinay T, Tasci Y, Dilbaz S, Cinar O, Demir B, Haberal A. The
major recipient- related determinants of success with donor eggs. relationship between endometrial thickness and pregnancy rates
Fertil Steril 2004;82:850–6. in GnRH antagonist down-regulated ICSI cycles. Gynecol
(42) Zhang X, Chen CH, Confino E, Barnes R, Milad M, Kazer RR. Endocrinol 2010;26(11):833–7.
Increased endometrial thickness is associated with improved (59) Weissman A, Gotlieb L, Casper RF. The detrimental effect of
outcome for selected patients undergoing in vitro fertilization- increased endometrial thickness on implantation and pregnancy
embryo transfer. Fertil Steril 2005;2:336–40. rates and outcome in in vitro fertilization program. Fertil Steril
(43) Amir W, Micha B, Ariel H, Liat L-G, Jehoshua D, Adrian S. 1999;71:147–9.
Predicting factors for endometrial thickness during treatment (60) Yoeli R, Ashkenazi J, Orvieto R, Shelef M, Kaplan B, Bar-Hava
with assisted reproductive technology. Fertil Steril 2007;87 I. Significance of increased endometrial thickness in assisted
(4):799–804.
10 Y. Mouhayar, F.I. Sharara

reproduction technology treatments. J Assist Reprod Genet in women with thin endometrium despite adequate follicular
2004;8:285–9. response with clomiphene citrate. Ain Shams Med J
(61) Al-Inany H. Intrauterine adhesions. An update. Acta Obs 2011;62:725–36.
Gynecol Scand 2001;80(11):986–93. (81) Chen X, Chen SL. Successful pregnancy in recurrent thin
(62) Gonen Y, Casper RF. Sonographic determination of a possible endometrium with new uses for an old drug. J IVF Reprod Med
adverse effect of clomiphene citrate on endometrial growth. Genet 2013;1:2.
Hum Reprod 1990;5(6):670–4. (82) Chiao T, Lee A. Role of pentoxifylline and vitamin E in
(63) Casper RF. It’s time to pay attention to the endometrium. Fertil attenuation of radiation-induced fibrosis. Ann Pharmacother
Steril 2011;96(3):519–21. 2005;39:16–22.
(64) Miwa I, Tamura H, Takasaki A, Yamagata Y, Shimamura K, (83) Lédée-Bataille N, Olivennes F, Lefaix J-L, Chaouat G, Frydman
Sugino N. Pathophysiologic features of ‘‘thin” endometrium. R, Delanian S. Combined treatment by pentoxifylline and
Fertil Steril 2009;91(4):998–1004. tocopherol for recipient women with a thin endometrium
(65) Senturk LM, Erel CT. Thin endometrium in assisted reproduc- enrolled in an oocyte donation programme. Hum Reprod
tive technology. Curr Opin Obstet Gynecol 2008;3:221–8. 2002;17(5):1249–53.
(66) Lebovitz O, Orvieto R. Treating patients with ‘‘thin” endome- (84) Letur-Konirsch H, Delanian S. Successful pregnancies after
trium- an ongoing challenge. Gynecol Endocrinol 2014;30 combined pentoxifylline-tocopherol treatment in women with
(6):409–14. premature ovarian failure who are resistant to hormone
(67) Kasius A, Smit JG, Torrance HL, Eijkemans MJC, Mol BW, replacement therapy. Fertil Steril 2003;79(2):439–41.
Opmeer BC, et al. Endometrial thickness and pregnancy rates (85) Acharya S, Yasmin E, Balen AH. The use of a combination of
after IVF: a systematic review and meta-analysis. Hum Reprod pentoxifylline and tocopherol in women with a thin endome-
Update 2014;20(4):530–41. trium undergoing assisted conception therapies – a report of 20
(68) Chen MJ, Yang JH, Peng FH, Chen SU, Ho HN, Yang YS. cases. Hum Fertil 2009;12(4):198–203.
Extended estrogen administration for women with thin endome- (86) Takasaki A, Tamura H, Miwa I, Taketani T, Shimamura K,
trium in frozen-thawed in-vitro fertilization programs. J Assist Sugino N. Endometrial growth and uterine blood flow: a pilot
Reprod Genet 2006;23(7–8):337–42. study for improving endometrial thickness in the patients with a
(69) Shen MS, Wang CW, Chen CH, Tzeng CR. New horizon on thin endometrium. Fertil Steril 2010;93(6):1851–8.
successful management for a woman with repeated implantation (87) Cicek N, Eryilmaz OG, Sarikaya E, Gulerman C, Genc Y.
failure due to unresponsive thin endometrium: use of extended Vitamin E effect on controlled ovarian stimulation of unex-
estrogen supplementation. J Obstet Gynaecol Res 2013;39 plained infertile women. J Assist Reprod Genet 2012;29
(5):1092–4. (4):325–8.
(70) Elizur SE, Son WY, Yap R, Gidoni Y, Levin D, Demirtas E, (88) Wada I, Hsu CC, Williams G, Macnamee MC, Brinsden PR.
et al. Comparison of low-dose human menopausal gonadotropin The benefits of low-dose aspirin therapy in women with impaired
and micronized 17 b-estradiol supplementation in in vitro uterine perfusion during assisted conception. Hum Reprod
maturation cycles with thin endometrial lining. Fertil Steril 1994;9(10):1954–7.
2009;92(3):907–12. (89) Check JH, Diatterich C, Lurie D, Nazari A, Chuong J. A
(71) Qublan H, Amarin Z, Al-Quda M, Diab F, Nawasreh M, matched study to determine whether low-dose aspirin without
Malkawi S, et al. Luteal phase support with GnRH-a improves heparin improves pregnancy rates following frozen embryo
implantation and pregnancy rates in IVF cycles with thin transfer and/or affects endometrial sonographic parameters. J
endometrium of 67 mm on day of egg retrieval. Human Fert Assist Reprod Genet 1998;15(10):579–82.
2008;11(1):43–7. (90) Weckstein LN, Jacobson A, Galen D, Hampton K, Hammel J.
(72) Papanikolaou EG, Kyrou D, Zervakakou G, Paggou E, Low-dose aspirin for oocyte donation recipients with a thin
Humaidan P. Follicular HCG endometrium priming for IVF endometrium: prospective, randomized study. Fertil Steril
patients experiencing resisting thin endometrium. A proof of 1997;68(5):927–30.
concept study. J Assist Reprod Genet 2013;30:1341–5. (91) Hsieh Y-Y, Tsai H-D, Chang C-C, Lo H-Y, Chen C-L. Low-
(73) Adashi EY. Clomiphene citrate: mechanism(s) and site(s) of dose aspirin for infertile women with thin endometrium receiving
action – a hypothesis revisited. Fertil Steril 1984;42:331–44. intrauterine insemination: a prospective, randomized study. J
(74) Drake TS, Tredway DR, Buchanan GC. Continued clinical Assist Reprod Genet 2000;17(3).
experience with an increasing dosage regimen of clomiphene (92) Paulus WE, Zhang M, Strehler E, El-Danasouri I, Sterzik K.
citrate administration. Fertil Steril 1978;30:274–7. Influence of acupuncture on the pregnancy rate in patients who
(75) Gysler M, March CM, Mishell DR, Baily EJ. A decade’s undergo assisted reproduction therapy. Fertil Steril 2002;77
experience with an individualized clomiphene treatment regimen (4):721–4.
including its effect on the postcoital test. Fertil Steril (93) Westergaard LG, Mao Q, Krogslund M, Sandrini S, Lenz S,
1982;37:161–7. Grinsted J. Acupuncture on the day of embryo transfer
(76) Wu CH, Winkel CA. The effect of therapy initiation day on significantly improves the reproductive outcome in infertile
clomiphene citrate therapy. Fertil Steril 1989;52:564–8. women: a prospective, randomized trial. Fertil Steril 2006;85
(77) Deligdisch L. Hormonal pathology of the endometrium. Mod (5):1341–6.
Pathol 2000;13:285–94. (94) Stener-Victorin E, Waldenstrom U, Andersson SA, Wikland M.
(78) Wang CW, Horng SG, Chen CK, Wang HS, Huang HY, Lee Reduction of blood flow impedance in the uterine arteries of
CL, et al. Ovulation induction with tamoxifen and alternate-day infertile women with electro-acupuncture. Hum Reprod 1996;11
gonadotropin in patients with thin endometrium. Reprod (6):1314–7.
Biomed Online 2008;17(1):20–6. (95) Ho M, Huang LC, Chang YY, Chen HY, Chang WC, Yang TC,
(79) Reynolds K, Khoury J, Sosnowski J, Thie J, Hofmann G. et al. Electroacupuncture reduces uterine artery blood flow
Comparison of the effect of tamoxifen on endometrial thickness impedance in infertile women. Taiwan J Obstet Gynecol 2009;48
in women with thin endometrium (<7mm) undergoing ovula- (2):148–51.
tion induction with clomiphene citrate. Fertil Steril 2009;93 (96) Shuai Z, Lian Fand, Li P, Yang W. Effect of transcutaneous
(6):2091–3. electrical acupuncture points stimulation on endometrial recep-
(80) Morad AWA, Abdelhamid AA, Al-Shazly IM. Tamoxifen and tivity in women undergoing frozen-thawed embryo transfer: a
sildenafil improve endometrial thickness and uterine blood flow
Modern management of thin lining 11

single-blind prospective randomized controlled trial. Acupunct (113) Padykula HA, Coles LG, McCracken JA, King Jr NW, Loncope
Med 2015;33:33–9. C, et al. A zonal pattern of cell proliferation and differentiation
(97) Bodombossou-Djobo MMa, Zheng C, Chen S, Yang D. in the rhesus endometrium during the estrogen surge. Biol
Neuromuscular electrical stimulation and biofeedback therapy Reprod 1984;31:1103–18.
may improve endometrial growth for patients with thin (114) Padykula HA, Coles LG, Okulicz WC, Rapaport SI,
endometrium during frozen-thawed embryo transfer: a prelim- McCracken JA, et al. The basalis of the primate endometrium:
inary report. Reprod Biol Endocrinol 2011;9:122. a bifunctional germinal compartment. Biol Reprod
(98) Sher G, Fisch JD. Vaginal sildenafil (Viagra): a preliminary 1989;40:681–90.
report of a novel method to improve uterine artery blood flow (115) Gargett CE. Uterine stem cells: what is the evidence? Hum
and endometrial development in patients undergoing IVF. Hum Reprod Update 2007;13(1):87–101.
Reprod 2000;15(4):806–9. (116) Gargett CE, Nguyen HPT, Ye L. Endometrial regeneration and
(99) Sher G, Fisch JD. Effect of vaginal sildenafil on the outcome of endometrial stem/progenitor cells. Rev Endocr Metab Disord
in vitro fertilization (IVF) after multiple IVF failures attributed to 2012;13(4):235–51.
poor endometrial development. Fertil Steril 2002;78(5):1073–6. (117) Cervello I, Mas A, Gil Sanchis C, Peris L, Saunders PT, et al.
(100) Check JH, Graziano V, Lee G, Nazari A, Choe JK, Dietterich C. Reconstruction of endometrium from human endometrial side
Neither sildenafil nor vaginal estradiol improves endometrial population cell lines. PLoS ONE 2011;6:e21221.
thickness in women with thin endometria after taking oral (118) Taylor HS. Endometrial cells derived from donor stem cells in
estradiol in graduating dosages. Clin Exp Obstet Gynecol bone marrow transplant recipients. JAMA 2004;292(1):81–5.
2004;31:99–102. (119) Bratincsák A, Brownstein MJ, Cassiani-Ingoni R, Pastorino S,
(101) Zinger M, Liu JH, Thomas MA. Successful use of vaginal Szalayova I, Tóth ZE, et al. CD45-positive blood cells give rise
sildenafil citrate in two infertility patients with Asherman’s to uterine epithelial cells in mice. Stem Cells 2007;25
syndrome. J Women’s Heal 2006;15(4):442–4. (11):2820–6.
(102) Takasaki A, Tamura H, Miwa I, Taketani T, Shimamura K, (120) Du H, Taylor HS. Contribution of bone marrow-derived stem
Sugino N. Endometrial growth and uterine blood flow: a pilot cells to endometrium and endometriosis. Stem cells 2007;25
study for improving endometrial thickness in patients with a thin (8):2082–6.
endometrium. Fertil Steril 2010;93(6):1851–8. (121) Cervello I, Gil Sanchis C, Mas A, Faus A, Sanz J, Moscardó,
(103) Eid ME. Sildenafil improves implantation rate in women with a et al. Bone marrow-derived cells from male donors do not
thin endometrium secondary to improvement of uterine blood contribute to the endometrial side population of the recipient.
flow; ‘‘pilot study”. Fertil Steril 2015;104:e342. PLoS ONE 2012;7(1):e30260.
(104) Jensen JR, Witz CA, Schenken RS, Tekmal RR. A potential role (122) Morelli SS, Rameshwar P, Goldsmith LT. Experimental evi-
for colony-stimulating factor 1 in the genesis of the early dence for bone marrow as a source of nonhematopoietic
endometriotic lesion. Fertil Steril 2010;93(1):251–6. endometrial stromal and epithelial compartment cells in a
(105) Gleicher N, Vidali A, Barad DH. Successful treatment of murine model. Biol Reprod 2013;89(1):7.
unresponsive thin endometrium. Fertil Steril 2011;95(6):2123. (123) Mints M, Jansson M, Sadeghi B, Westgren M, Uzunel M,
e13–e17. Hassan M, et al. Endometrial endothelial cells are derived from
(106) Gleicher N, Kim A, Michaeli T, Lee H-J, Shohat-Tal A, donor stem cells in a bone marrow transplant. Hum Reprod
Lazzaroni E, et al. A pilot cohort study of granulocyte colony- 2008;23:139–43.
stimulating factor in the treatment of unresponsive thin (124) Ikoma T, Kyo S, Maida Y, Ozaki S, Takakura M, Nakao S,
endometrium resistant to standard therapies. Hum Reprod et al. Bone marrow-derived cells from male donors can compose
2013;28(1):172–7. endometrial glands in female transplant recipients. Am J Obstet
(107) Lucena E, Moreno-Ortiz H. Granulocyte colony-stimulating Gynecol 2009;201:608.e1–8.
factor (G-CSF): a mediator in endometrial receptivity for a (125) Du H, Naqvi H, Taylor HS. Ischemia/reperfusion injury
patient with polycystic ovary (PCO) undergoing in vitro matu- promotes and pranulocyte-colony stimulating factor inhibits
ration (IVM). BMJ Case Rep 2013. http://dx.doi.org/10.1136/ migration of bone marrow-derived stem cells to endometrium.
bcr-2012-008115. Stem Cells Dev 2012;21(18):3324–31.
(108) Check JH, Cohen R, Choe JK. Failure to improve a thin (126) Alawadhi F, Du H, Cakmak H, Taylor H. Bone marrow-derived
endometrium in the late proliferative phase with uterine infusion stem cell (BMDSC) transplantation improves fertility in a
of granulocyte-colony stimulating factor. Clin Exp Obstet murine model of Asherman’s syndrome. PLoS ONE 2014;9(5):
Gynecol 2014;41(4):473–5. e96662.
(109) Li Y, Pan P, Chen X, Li L, Li Y, Yang D. Granulocyte colony- (127) Kilic S, Yuksel B, Pinarli F, Albayrak A, Boztok B, Delibasi T.
stimulating factor administration for infertile women with thin Effect of stem cell application on Asherman syndrome, an
endometrium in frozen embryo transfer program. Reprod Sci experimental rat model. J Assist Reprod Genet 2013;31:975–82.
2014;21(3):381–5. (128) Zhao J, Zhang Q, Wang Y, Li Y. Rat bone marrow mesenchy-
(110) Kunicki M, Łukaszuk K, Woclawek-Potocka I, Liss J, Kul- mal stem cells improve regeneration of thin endometrium in rat.
wikowska P, Szczyptańska J. Evaluation of granulocyte colony- Fertil Steril 2014;101(2):587–94.
stimulating factor effects on treatment-resistant thin endome- (129) Zhao J, Zhang Q, Wang Y, Li Y. Uterine infusion with bone
trium in women undergoing in vitro fertilization. Biomed Res Int marrow mesenchymal stem cells improves endometrium thick-
2014;2014:913235. ness in a rat model of thin endometrium. Reprod Sci 2015;22
(111) Barad DH, Yu Y, Kushnir Va, Shohat-Tal A, Lazzaroni E, Lee (2):181–8.
HJ, et al. A randomized clinical trial of endometrial perfusion (130) Gil-Sanchis C, Cervelló I, Khurana S, Faus A, Verfaillie C,
with granulocyte colony-stimulating factor in in vitro fertiliza- Simón C. Contribution of different bone marrow-derived cell
tion cycles: Impact on endometrial thickness and clinical types in endometrial regeneration using an irradiated murine
pregnancy rates. Fertil Steril 2014;101(3):710–5. model. Fertil Steril 2015;103(6):1596–605.
(112) Xu B, Zhang Q, Hao J, Xu D, Li Y. Two protocols to treat thin (131) Nagori CB, Panchal SY, Patel H. Endometrial regeneration
endometrium with granulocyte colony-stimulating factor during using autologous adult stem cells followed by conception by
frozen embryo transfer cycles. Reprod Biomed Online 2015;30 in vitro fertilization in a patient of severe Asherman’s syndrome.
(4):349–58. J Hum Reprod Sci 2011;4(1):43–8.
12 Y. Mouhayar, F.I. Sharara

(132) Singh N, Mohanty S, Seth T, Shankar M, Bhaskaran S, (134) Santamaria X, Cabanillas S, Cervello I, Arbona C, Raga F,
Dharmendra S. Autologous stem cell transplantation in refrac- Ferro J, et al. Autologous cell therapy with CD133+ bone
tory Asherman’s syndrome: a novel cell based therapy. J Hum marrow-derived stem cells from refractory Asherman’s syn-
Reprod Sci 2014;7(2):93–8. drome and endometrial atrophy: a pilot cohort study. Hum
(133) Cervello I, Gil-Sanchis C, Santamaria X, Cabanillas S, Diaz A, Reprod 2016;31(5):1087–96.
Faus A, et al. Human CD133+ bone marrow-derived stem cells (135) Mahajan N. Endometrial receptivity array: clinical application. J
promote endometrial proliferation in a murine model of Ash- Hum Reprod Sci 2015;8(3):121–9.
erman syndrome. Fertil Steril 2015;104:1552–60.

S-ar putea să vă placă și