Skip to main content
Erschienen in: Journal of Ovarian Research 1/2023

Open Access 01.12.2023 | Research

Which endometrial preparation protocol provides better pregnancy and perinatal outcomes for endometriosis patients in frozen-thawed embryo transfer cycles? A retrospective study on 1413 patients

verfasst von: Yaxin Guo, Zishui Fang, Lin Yu, Xin Sun, Fei Li, Lei Jin

Erschienen in: Journal of Ovarian Research | Ausgabe 1/2023

Abstract

Objective

To determine the optimal endometrial preparation protocol for a frozen embryo transfer in patients with endometriosis.

Design

Retrospective cohort study.

Setting

Tertiary care academic medical center.

Patient(s)

One thousand four hundred thirteen patients with endometriosis who underwent oocyte aspiration from 2015 to 2020 and frozen embryo transfer from 2016 to 2020 and received natural cycle, hormone replacement treatment with or without GnRHa pretreatment endometrial preparation.

Intervention(s)

None.

Main outcome measure(s)

Clinical pregnancy rate, live birth rate, miscarriage rate, multiple pregnancy rate, biochemical pregnancy rate and ectopic pregnancy rate. Singleton live births were assessed for perinatal outcomes and obstetric complications.

Result(s)

There were no differences in clinical pregnancy outcomes or prenatal outcomes among the three commonly used endometrial preparation protocols for frozen embryo transfer cycles in patients with endometriosis. Results remained after screening variables using univariate logistic regression into multivariate logistic regression. No advantages or disadvantages were found among the three endometrial preparation protocols in patients with endometriosis.

Conclusion(s)

Natural cycle, hormone replacement cycle, or hormone replacement treatment with GnRHa pretreatment showed no superiority or inferiority in pregnancy and perinatal outcomes in patients with endometriosis.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13048-023-01095-4.
Fei Li and Lei Jin should be considered similar in author order.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Endometriosis, a common health problem affecting 10% of women of reproductive age, is defined as the presence of endometrium-like tissue outside the uterus [1]. Numerous studies have demonstrated cellular and molecular differences in the eutopic endometrium of patients with and without endometriosis, which may lead to altered endometrial receptivity [2, 3]. Many women whose fertility are impaired by endometriosis require treatment with assisted reproductive technology (ART) to achieve pregnancy outcomes. However, the optimal endometrial preparation protocol in frozen-thawed embryo transfer (FET) cycles for patients with endometriosis is rarely discussed.
Despite the increase in FET, the most optimal priming regimen of the endometrium in the ART general population remains controversial [4], not to mention that there are few studies on endometriosis [5]. In a Cochrane review on the effect of hormonal treatment prior to ART, the authors conclude that administration of gonadotropin-releasing hormone agonists (GnRHa) for a period of 3–6 months in women with endometriosis increases the live birth rate and clinical pregnancy rate [6]. However, the updated version of this review showed uncertain results as to whether long-term GnRHa therapy impacts on the live birth rate or indeed the complication rate [7]. Moreover, one study suggested that long-term pituitary downregulation before frozen embryo transfer could improve pregnancy outcomes in women with adenomyosis [8], which often coexists with endometriosis. Whereas, Muzi Li et al. disagreed [9]. These equivocal results raise the question of whether the endometrial preparation regimen with GnRHa downregulation might also be beneficial for patients with endometriosis. To the best of our knowledge, there are still no clinical studies on this issue.
The present study was conducted to assess the effect of different endometrial preparation regimens on pregnancy and perinatal outcomes in women with endometriosis after FET cycles. In this study, we performed a retrospective cohort study to compare the pregnancy outcomes and perinatal outcomes of singletons conceived after FET with natural cycle (NC), hormone replacement treatment (HRT) with or without GnRHa pretreatment in women with endometriosis.

Materials and methods

Patients

This was a retrospective cohort study of patients with endometriosis who underwent oocyte aspiration at the Reproductive Medical Center of Tongji hospital from 2015 to 2020 and underwent FET cycles in our center from 2016 to 2020. Inclusion criteria included a normal uterine cavity as assessed by ultrasonography, hysterosalpingography, or hysteroscopy, and high-quality frozen embryos were transferred. Patients with endometriosis were diagnosed by laparoscopy or laparotomy.
Non-autologous, PGD, and canceled cycles were excluded. In our reproductive medicine center, three endometrial preparation protocols were mainly applied for women with endometriosis namely NC, HRT, and hormone replacement treatment with GnRHa pretreatment (GnRHa + HRT). FET cycles with other endometrial preparation protocols were also excluded (Supplemental Fig. 1). This study was approved by the Institutional Review Board of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (reference: TJ-IRB20211291).

Endometrial preparation before embryo transfer

Three commonly used endometrial preparation protocols have been described elsewhere [1012]. From 2016 to 2020, 74 patients with endometriosis underwent natural cycle FET (NC group). Following spontaneous menstruation, endometrial thickness, follicular development, and ovulation were evaluated by transvaginal ultrasound examination (USE) and the serum progesterone levels were measured starting on days 10–12 of the menstrual cycle. FET was planned for 3 days after ovulation, indicated by serum progesterone > 5 ng/mL. Intramuscular administration of progesterone for luteal support was started from 1 day after ovulation. For hormone replacement treatment cycles (HRT group), oral administration of estradiol (Progynova; Bayer Schering Pharma AG, Germany) was initiated with 2 mg/day from cycle days 1 to 4, 4 mg/day from days 5 to 8, and 6 mg/day from days 9 to 12. Similarly, the endometrial thickness and ovulation were assessed from day 13, and the estradiol dose was adjusted based on the endometrial thickness. 40 mg intramuscular administration of progesterone was administered and maintained for the following 3 days when the endometrial thickness reached at least 8 mm. Embryo transfer was conducted on day 4, after 3 days of progesterone administration. For HRT with GnRHa pretreatment cycles (GnRHa + HRT group), GnRHa including triptorelin and leuprorelin was injected at a dose of 3.75 mg on day 2 of menstruation. After a follow-visit 28 days later, patients started their HRT cycles as previously described.
Embryo culture, vitrification and warming were implemented as previously published [11, 13].

Outcomes

In this study, both pregnancy outcomes and prenatal outcomes were considered. Live birth was defined as the delivery of at least one viable infant. Clinical pregnancy was classified as those cycles resulting in the identification of a gestational sac with fetal heart activity on USE. A positive pregnancy test that did not result in a clinical pregnancy was referred to as a biochemical pregnancy loss. Miscarriage was defined as spontaneous loss after sonographic visualization of an intrauterine gestational sac. The primary outcome was the live birth rate (LBR) and clinical pregnancy rate (CPR).
In order to reduce bias due to vanishing twin pregnancies, only live births from singleton pregnancies were selected for analysis of perinatal outcomes. Low birth weight (LBW) was defined as birth weight of fewer than 2500 g while macrosomia was defined as birth weight of more than 4000 g. Small for gestational age (SGA) was defined as birthweight <10th percentile while large for gestational age (LGA) was defined as birthweight >90th percentile of reference standard birthweight for gestational age, which was based on Chinese populations and adjusted for sex and gestational age [14]. Preterm birth (PTB) was defined as live birth before 37 weeks gestation. Information regarding all abnormal perinatal outcomes was obtained by a follow-up telephone interview and entered into the electronic database.

Statistical analysis

SAS 9.4 and R 4.1.2 were utilized for data analysis in this study. All continuous variables were assessed for normality of distribution by the Shapiro-Wilk test. The median and interquartile range was used for continuous variables since none of them were normally distributed, and categorical data were expressed as frequency and percentage. Patient characteristics and outcomes were summarized descriptively. Continuous variables were assessed by the Kruskal-Wallis test, and categorical variables were analyzed by using the chi-squared test or Fisher’s exact test, as appropriate. Missing values are imputed with median due to missing at random. To evaluate the association between the different endometrial preparation protocols and pregnancy outcomes and perinatal outcomes, confounding variables should be identified and adjusted. Univariate logistic regressions between each outcome and all covariates were firstly applied to identify covariates that effect the outcomes. Then, multivariate logistic regressions were conducted where statistically significant covariates with significance levels less than 0.2, clinically significant variables and the exposure of interest were considered. In the multivariate logistic regressions for pregnancy outcome, we always included the exposure of interest endometrial preparation protocols, and three clinically significant variables including maternal age at FET, BMI and infertility diagnosis. For prenatal outcomes, maternal age at FET, BMI, and endometrial preparation protocols were fixed in the model. The variable selection was done using backward method. Unadjusted odds ratios (ORs) and adjusted odds ratios (AORs) were reported with their 95% confidence intervals (95% CIs) to demonstrate the level of overall association. Bonferroni correction was used for multiple comparisons. The sensitivity analysis was conducted excluding patients with any missing data to test the accuracy of the analysis. All statistical tests were two-sided, and the level of statistical significance was set at P < 0.05.

Results

Baseline and cycle characteristics

A total of 1413 patients who fulfilled the inclusion and exclusion criteria were included in this analysis, which were grouped according to the endometrial preparation protocols. There were 389, 950, and 74 patients in GnRHa + HRT group, HRT group and NC group, respectively. The baseline characteristics of the patient cohort are detailed in Table 1, which revealed statistically significant differences (p < 0.05) for AMH, AFC, infertility duration, and infertility etiology.
Table 1
General characteristics and clinical outcomes of patients with different endometrial preparation protocols
Variable
GnRHa + HRT
(N = 389)
HRT
(N = 950)
NC
(N = 74)
P value
Maternal age at oocyte retrieval, y
31.0 (29.0, 35.0)
31.0 (29.0, 35.0)
32.0 (30.0, 35.0)
0.2013
Body mass index, kg/m2
20.8 (19.4, 22.5)
20.8 (19.5, 22.8)
21.1 (19.6, 22.6)
0.7423
Baseline FSH, mIU/mL
7.8 (6.6, 9.6)
7.8 (6.5, 9.0)
7.9 (6.4, 9.8)
0.6653
Antral follicle count (AFC)
8.0 (5.0, 11.0)
9.0 (6.0, 14.0)
7.0 (4.0, 13.0)
<.0013*
AMH level, ng/ml
2.4 (1.4, 4.5)
2.8 (1.7, 5.3)
2.1 (1.2, 4.0)
<.0013*
Duration of infertility, years
2.0 (1.5, 4.0)
3.0 (2.0, 4.0)
3.0 (2.0, 5.0)
0.0063*
Infertility diagnosis
   
0.9691
Primary infertility, n (%)
257 (66.1%)
629 (66.2%)
50 (67.6%)
 
Secondary infertility, n (%)
132 (33.9%)
321 (33.8%)
24 (32.4%)
 
Infertility etiology, n (%)
 Male factor
64 (16.5%)
167 (17.6%)
19 (25.7%)
0.1611
 Female factors
    
  Tubal factor
189 (48.6%)
472 (49.7%)
35 (47.3%)
0.8811
  Ovulatory
3 (0.8%)
42 (4.4%)
1 (1.4%)
0.0012*
  Diminished ovarian reserve
115 (29.6%)
218 (22.9%)
19 (25.7%)
0.0391*
  Uterine malformation
105 (27.0%)
187 (19.7%)
21 (28.4%)
0.0061*
 Unexplained/Other
0 (0.0%)
6 (0.6%)
1 (1.4%)
0.1152
Ovarian stimulation protocols, n (%)
   
<.0011*
 Long GnRH-a
43 (11.1%)
165 (17.4%)
12 (16.2%)
 
 GnRH-a ultra-long
98 (25.2%)
342 (36.0%)
25 (33.8%)
 
 GnRH antagonist
146 (37.5%)
198 (20.8%)
16 (21.6%)
 
 Other protocols
102 (26.2%)
245 (25.8%)
21 (28.4%)
 
Duration of stimulation, days
10.0 (9.0, 11.0)
10.0 (9.0, 11.0)
10.0 (9.0, 11.0)
0.0183*
Gonadotropin dose, IU
2625.0 (2100.0, 3150.0)
2475.0 (1912.5, 3000.0)
2625.0 (2137.5, 3150.0)
0.0113*
No. of oocytes retrieved
9.0 (5.0, 14.0)
10.0 (6.0, 16.0)
8.0 (5.0, 14.0)
0.0033*
No. of MII oocytes
7.0 (4.0, 12.0)
9.0 (5.0, 14.0)
7.0 (4.0, 12.0)
0.0013*
Oocyte maturation rate
0.9 (0.8, 1.0)
0.9 (0.8, 1.0)
0.9 (0.8, 1.0)
0.4783
Fertilization, n (%)
   
0.0701
 IVF
105 (27.0%)
199 (20.9%)
23 (31.1%)
 
 ICSI
266 (68.4%)
698 (73.5%)
48 (64.9%)
 
 Rescue ICSI
18 (4.6%)
53 (5.6%)
3 (4.1%)
 
The number of 2PN
5.0 (3.0, 9.0)
6.0 (4.0, 10.0)
5.0 (3.0, 10.0)
0.0123*
Normal fertilization rate
0.7 (0.5, 0.9)
0.7 (0.5, 0.8)
0.7 (0.5, 0.9)
0.1873
Blastocyst formation rate
0.8 (0.5, 1.0)
0.7 (0.5, 0.9)
0.7 (0.5, 1.0)
0.0253*
Maternal age at FET, y
32.0 (29.0, 35.0)
32.0 (29.0, 35.0)
33.0 (31.0, 35.0)
0.0633
Interval between FET and IVF/ICSI, days
125.0 (85.0, 248.0)
91.0 (60.0, 172.0)
112.0 (63.0, 263.0)
<.0013*
No. of embryos thawed
   
0.2262
 1
242 (62.2%)
537 (56.5%)
44 (59.5%)
 
 2
143 (36.8%)
393 (41.4%)
28 (37.8%)
 
 > = 3
4 (1.0%)
20 (2.1%)
2 (2.7%)
 
No. of surviving embryos
   
0.1942
 1
243 (62.5%)
540 (56.8%)
44 (59.5%)
 
 2
143 (36.8%)
394 (41.5%)
28 (37.8%)
 
 3
3 (0.8%)
16 (1.7%)
2 (2.7%)
 
total no. of surviving embryos/no. of embryos thawed
1.0 (1.0, 1.0)
1.0 (1.0, 1.0)
1.0 (1.0, 1.0)
0.5283
No. of embryos transferred, n (%)
   
0.3241
 1
249 (64.0%)
568 (59.8%)
47 (63.5%)
 
 2
140 (36.0%)
382 (40.2%)
27 (36.5%)
 
Type of embryo transferred, n (%)
   
0.0291*
 Cleavage embryo
142 (36.5%)
296 (31.2%)
32 (43.2%)
 
 Blastocyst
247 (63.5%)
654 (68.8%)
42 (56.8%)
 
Endometrial thickness, mm
9.7 (8.8, 11.0)
9.2 (8.4, 10.1)
9.7 (8.8, 10.5)
<.0013*
Luteal phase support, n (%)
   
<.0011*
 Intramuscular injection and oral administration
11 (2.8%)
179 (18.8%)
10 (13.5%)
 
 Vaginal gel administration and oral administration
179 (46.0%)
387 (40.7%)
26 (35.1%)
 
 Vaginal suppository administration and oral administration
197 (50.6%)
381 (40.1%)
24 (32.4%)
 
 Others
2 (0.5%)
3 (0.3%)
14 (18.9%)
 
Live Birth, n (%)
156 (40.1%)
384 (40.4%)
30 (40.5%)
0.9941
Clinical Pregnancy, n (%)
200 (51.4%)
475 (50.0%)
34 (45.9%)
0.6771
Miscarriage, n (%)
44 (11.3%)
91 (9.6%)
4 (5.4%)
0.2812
Multiple pregnancy, n (%)
37 (9.5%)
108 (11.4%)
4 (5.4%)
0.2212
Biochemical Pregnancy, n (%)
29 (7.5%)
61 (6.4%)
2 (2.7%)
0.3212
Ectopic pregnancy, n (%)
2 (0.5%)
7 (0.7%)
0 (0.0%)
>.9992
1Chi-Square p-value; 2Fisher Exact p-value; 3Kruskal-Wallis p-value; *P < .05
As for cycle characteristics, ovarian stimulation protocols, gonadotropin dose and duration, the number of retrieved oocytes, MII oocytes and 2PN, blastocyst formation rate, the interval between FET and IVF/ICSI, type of embryo transferred, endometrial thickness and luteal phase support were significantly different in the study groups (p < 0.05) (Table 1).

Clinical pregnancy outcomes

The clinical pregnancy outcomes of the total study population are shown in Table 1. There were no statistically significant differences in any pregnancy outcomes including live birth rate (LBR), clinical pregnancy rate (CPR), miscarriage rate (MR), multiple pregnancy rate (MPR), biochemical pregnancy rate (BPR), and ectopic pregnancy rate (EPR). After univariate analysis (Table 2), predictors with significance levels less than 0.2, endometrial preparation protocols as well as maternal age at FET, BMI and infertility diagnosis were selected for the following multivariate logistic regression. After adjusting for possible confounding factors, no association was found between endometrial preparation protocols and clinical pregnancy outcomes. Sensitivity analysis by excluding patients with any missing data showed similar significance (data not shown).
Table 2
Crude and adjusted odds ratios of clinical outcomes
Variable
HRT vs. NC
GnRHa + HRT vs. NC
GnRHa + HRT vs. HRT
Crude OR (95% CI)
Adjusted OR (95% CI)
Crude OR (95% CI)
Adjusted OR (95% CI)
Crude OR (95% CI)
Adjusted OR (95% CI)
Live Birth a
1.00 (0.61-1.61)
0.83 (0.49-1.38)
0.98 (0.59-1.63)
0.86 (0.50-1.48)
0.99 (0.78-1.26)
1.04 (0.80-1.35)
Clinical Pregnancy b
1.18 (0.73-1.89)
0.99 (0.60-1.64)
1.24 (0.76-2.05)
1.13 (0.67-1.93)
1.06 (0.84-1.34)
1.15 (0.89-1.48)
Miscarriage c
1.85 (0.66-5.20)
1.80 (0.64-5.07)
2.23 (0.78-6.41)
2.22 (0.77-6.39)
1.20 (0.82-1.76)
1.23 (0.84-1.80)
Multiple pregnancy d
2.24 (0.80-6.27)
1.91 (0.56-6.53)
1.84 (0.64-5.32)
2.24 (0.63-7.92)
0.82 (0.55-1.21)
1.17 (0.70-1.96)
Biochemical Pregnancy e
2.47 (0.59-10.30)
2.31 (0.55-9.66)
2.90 (0.68-12.41)
2.80 (0.65-12.02)
1.17 (0.74-1.86)
1.21 (0.76-1.92)
Odds ratios (ORs) and 95% confidence intervals (CIs) are based on the univariate analysis while adjusted odds ratios (AORs) and 95% CIs are based on the multiple logistic regression model
a Adjusted for maternal age at FET, BMI, infertility diagnosis, diminished ovarian reserve, No. of surviving embryos, type of embryos transferred and endometrial thickness
b Adjusted for maternal age at FET, BMI, infertility diagnosis, diminished ovarian reserve,endometrial thickness, No and type of embryos transferred
c Adjusted for maternal age at FET, BMI, infertility diagnosis, type of embryos transferred
d Adjusted for maternal age at FET, BMI, infertility diagnosis, type of embryos transferred
e Adjusted for maternal age at FET, BMI, infertility diagnosis, type of embryos transferred

Singleton perinatal outcomes

A total of 465 singleton live births conceived through GnRHa + HRT cycles (n = 131), HRT cycles (n = 304), and NC (n = 30) were evaluated for obstetric complications and adverse birth outcomes. Patient demographics, treatment factors, and singleton perinatal outcomes are shown in Table 3. The median birth weights for GnRHa + HRT, HRT, and NC ETs were 3.3 kg, 3.4 kg, and 3.2 kg, respectively. The median gestational ages were 38.9 weeks, 39.0 weeks, and 38.6 weeks, respectively. There were no significant differences between the three groups in gestational age, birth weight, delivery mode, gender, abnormal perinatal outcomes, and obstetric complications. After univariate regression analysis and multivariate regression analysis, the results were still retained, with no statistical difference found (Table 4). Sensitivity analysis was conducted by excluding patients with any missing data and showed similar findings (data not shown).
Table 3
General characteristics and perinatal outcomes of patients with singleton live births
Variable
GnRHa + HRT
(N = 131)
HRT
(N = 304)
NC
(N = 30)
P value
Maternal age at oocyte retrieval, y
30.0 (28.0, 33.0)
30.0 (28.0, 33.0)
32.0 (29.0, 33.0)
0.3053
Body mass index, kg/m2
20.7 (19.2, 22.1)
20.8 (19.5, 22.6)
21.6 (19.7, 22.9)
0.3403
Baseline FSH, mIU/mL
7.7 (6.5, 8.9)
7.5 (6.3, 8.8)
7.7 (6.2, 8.5)
0.7573
Antral follicle count (AFC)
8.0 (5.0, 12.0)
11.0 (7.0, 15.5)
9.5 (6.0, 18.0)
<.0013*
AMH level, ng/ml
2.7 (1.4, 4.9)
3.3 (2.1, 5.6)
3.0 (1.6, 4.8)
0.0143*
Duration of infertility, years
2.0 (1.5, 3.0)
3.0 (1.5, 4.0)
3.0 (2.0, 3.0)
0.2843
Infertility diagnosis
   
0.7471
Primary infertility, n (%)
91 (69.5%)
219 (72.0%)
20 (66.7%)
 
Secondary infertility, n (%)
40 (30.5%)
85 (28.0%)
10 (33.3%)
 
Infertility etiology, n (%)
 Male factor
21 (16.0%)
50 (16.4%)
5 (16.7%)
>.9992
 Female factors
    
  Tubal factor
67 (51.1%)
155 (51.0%)
15 (50.0%)
0.9941
  Ovulatory
1 (0.8%)
12 (3.9%)
1 (3.3%)
0.1642
  Diminished ovarian reserve
29 (22.1%)
43 (14.1%)
3 (10.0%)
0.0822
  Uterine malformation
30 (22.9%)
44 (14.5%)
9 (30.0%)
0.0221*
 Unexplained/Other
0 (0.0%)
3 (1.0%)
0 (0.0%)
0.6382
Ovarian stimulation protocols, n (%)
   
0.0051*
 Long GnRH-a
21 (16.0%)
66 (21.7%)
8 (26.7%)
 
 GnRH-a ultra-long
34 (26.0%)
124 (40.8%)
13 (43.3%)
 
 GnRH antagonist
47 (35.9%)
65 (21.4%)
5 (16.7%)
 
 Other protocols
29 (22.1%)
49 (16.1%)
4 (13.3%)
 
Duration of stimulation, days
10.0 (9.0, 11.0)
10.0 (9.0, 11.0)
10.0 (10.0, 12.0)
0.0203*
Gonadotropin dose, IU
2625.0 (2025.0, 3165.0)
2400.0 (1875.0, 3000.0)
2550.0 (2100.0, 3075.0)
0.1613
No. of oocytes retrieved
9.0 (5.0, 14.0)
12.0 (8.0, 17.0)
11.0 (7.0, 16.0)
<.0013*
No. of MII oocytes
8.0 (4.0, 12.0)
11.0 (7.0, 15.0)
10.0 (6.0, 14.0)
<.0013*
Oocyte maturation rate
0.9 (0.8, 1.0)
0.9 (0.8, 1.0)
0.9 (0.9, 1.0)
0.7453
Fertilization, n (%)
   
0.3432
 IVF
35 (26.7%)
62 (20.4%)
5 (16.7%)
 
 ICSI
88 (67.2%)
225 (74.0%)
25 (83.3%)
 
 Rescue ICSI
8 (6.1%)
17 (5.6%)
0 (0.0%)
 
The number of 2PN
6.0 (3.0, 8.0)
7.0 (4.5, 10.5)
8.0 (4.0, 11.0)
0.0043*
Normal fertilization rate
0.7 (0.6, 0.9)
0.7 (0.6, 0.8)
0.7 (0.6, 0.9)
0.1463
Blastocyst formation rate
0.8 (0.6, 1.0)
0.8 (0.5, 0.9)
0.7 (0.5, 0.8)
0.1173
Maternal age at FET, y
31.0 (28.0, 34.0)
31.0 (29.0, 33.0)
32.0 (30.0, 33.0)
0.3273
Interval between FET and IVF/ICSI, days
131.0 (85.0, 229.0)
90.5 (60.0, 166.0)
80.5 (59.0, 220.0)
<.0013*
No. of embryos thawed
   
0.4802
 1
19 (63.3%)
187 (61.5%)
89 (67.9%)
 
 2
10 (33.3%)
113 (37.2%)
41 (32.1%)
 
 > = 3
1 (3.3%)
4 (1.3%)
1 (0.8%)
 
No. of surviving embryos
   
0.3142
 1
19 (63.3%)
188 (61.8%)
89 (67.9%)
 
 2
10 (33.3%)
113 (37.2%)
42 (32.1%)
 
 > = 3
1 (3.3%)
3 (1.0%)
0 (0.0%)
 
total no. of surviving embryos/no. of embryos thawed
1.0 (1.0, 1.0)
1.0 (1.0, 1.0)
1.0 (1.0, 1.0)
0.8953
No. of embryos transferred, n (%)
   
0.8161
 1
89 (67.9%)
198 (65.1%)
19 (63.3%)
 
 2
42 (32.1%)
106 (34.9%)
11 (36.7%)
 
Type of embryo transferred, n (%)
   
0.0511
 Cleavage embryo
35 (26.7%)
52 (17.1%)
8 (26.7%)
 
 Blastocyst
96 (73.3%)
252 (82.9%)
22 (73.3%)
 
Endometrial thickness, mm
10.1 (8.9, 11.3)
9.3 (8.6, 10.3)
9.3 (8.7, 10.4)
<.0013*
Luteal phase support, n (%)
   
<.0011*
 Intramuscular injection and oral administration
1 (0.8%)
60 (19.7%)
5 (16.7%)
 
 Vaginal gel administration and oral administration
64 (48.9%)
128 (42.1%)
9 (30.0%)
 
 Vaginal suppository administration and oral administration
66 (50.4%)
115 (37.8%)
9 (30.0%)
 
 Others
0 (0.0%)
1 (0.3%)
7 (23.3%)
 
Gestational age, wk
38.9 (38.0, 39.4)
39.0 (38.0, 39.6)
38.6 (37.0, 39.0)
0.0823
Birth weight, kg
3.3 (3.0, 3.6)
3.4 (3.1, 3.7)
3.2 (2.7, 3.6)
0.2373
Delivery mode, n (%)
   
0.8481
 Cesarean delivery
114 (87.0%)
260 (85.5%)
25 (83.3%)
 
 Natural labor
17 (13.0%)
44 (14.5%)
5 (16.7%)
 
Gender, n (%)
   
0.1601
 Male
81 (61.8%)
159 (52.3%)
15 (50.0%)
 
 Female
50 (38.2%)
145 (47.7%)
15 (50.0%)
 
Low birth weight < 2500 g
9 (6.9%)
19 (6.3%)
2 (6.7%)
0.9522
Macrosomia > 4000 g
5 (3.8%)
18 (5.9%)
1 (3.3%)
0.7262
Small for gestational age, n (%)
10 (7.6%)
17 (5.6%)
4 (13.3%)
0.1942
Large for gestational age, n (%)
21 (16.0%)
51 (16.8%)
5 (16.7%)
0.9791
Preterm birth < 37 wk., n (%)
16 (12.2%)
37 (12.2%)
4 (13.3%)
0.9452
Gestational diabetes mellitus, n (%)
4 (3.1%)
18 (5.9%)
3 (10.0%)
0.1852
Hypertensive disorders of pregnancy, n (%)
125 (95.4%)
291 (95.7%)
29 (96.7%)
>.9992
Placenta previa, n (%)
8 (6.1%)
19 (6.3%)
2 (6.7%)
>.9992
Fetal malformation, n (%)
2 (1.5%)
11 (3.6%)
1 (3.3%)
0.5102
1Chi-Square p-value; 2Fisher Exact p-value; 3Kruskal-Wallis p-value; *P < .05
Table 4
Singleton perinatal outcomes of patients in different endometrial preparation protocols
Variable
HRT vs. NC
GnRHa + HRT vs. NC
GnRHa + HRT vs. HRT
Crude OR (95% CI)
Adjusted OR (95% CI)
Crude OR (95% CI)
Adjusted OR (95% CI)
Crude OR (95% CI)
Adjusted OR (95% CI)
Delivery mode
1.18 (0.43-3.25)
1.30 (0.47-3.61)
1.34 (0.45-3.98)
1.50 (0.50-4.49)
1.13 (0.62-2.07)
1.15 (0.63-2.11)
Low birth weight < 2500 ga
0.93 (0.21-4.22)
2.12 (0.38-11.95)
1.03 (0.21-5.05)
1.45 (0.24-8.63)
1.11 (0.49-2.51)
0.68 (0.28-1.68)
Macrosomia > 4000 gb
1.83 (0.24-14.17)
1.74 (0.22-13.77)
1.15 (0.13-10.23)
1.14 (0.13-10.33)
0.63 (0.23-1.74)
0.66 (0.24-1.82)
Small for gestational agec, n (%)
0.39 (0.12-1.23)
0.42 (0.13-1.37)
0.54 (0.16-1.85)
0.48 (0.14-1.69)
1.39 (0.62-3.12)
1.14 (0.50-2.62)
Large for gestational aged, n (%)
1.01 (0.37-2.77)
0.97 (0.35-2.69)
0.95 (0.33-2.78)
1.10 (0.37-3.26)
0.94 (0.54-1.64)
1.13 (0.64-2.01)
Preterm birth < 37 wke, n (%)
0.90 (0.30-2.73)
0.68 (0.22-2.16)
0.90 (0.28-2.93)
0.68 (0.20-2.31)
1.00 (0.54-1.88)
0.99 (0.51-1.92)
Gestational diabetes mellitusf, n (%)
0.57 (0.16-2.05)
0.67 (0.18-2.51)
0.28 (0.06-1.34)
0.27 (0.05-1.31)
0.50 (0.17-1.51)
0.40 (0.13-1.23)
Hypertensive disorders of pregnancyg, n (%)
1.30 (0.16-10.25)
1.47 (0.17-12.78)
1.39 (0.16-12.00)
1.65 (0.17-15.87)
1.07 (0.40-2.89)
1.12 (0.39-3.22)
Fetal malformationh, n (%)
1.09 (0.14-8.74)
0.96 (0.12-7.88)
0.45 (0.04-5.13)
0.41 (0.04-4.78)
0.41 (0.09-1.89)
0.43 (0.09-1.97)
Odds ratios (ORs) and 95% confidence intervals (CIs) are based on the univariate analysis while adjusted odds ratios (AORs) and 95% CIs are based on the multiple logistic regression model
a Adjusted for maternal age at FET, BMI, AMH, duration of infertility, No. of oocytes retrieved, No. of MII oocytes, endometrial thickness,No. of embryos transferred
b Adjusted for maternal age at FET, BMI, interval between FET and IVF/ICSI
c Adjusted for maternal age at FET, BMI, No. of MII oocytes
d Adjusted for maternal age at FET, BMI, AFC and unexplained/other ovarian stimulation protocols
e Adjusted for maternal age at FET, BMI, ovarian stimulation protocols, normal fertilization rate, and type of embryo transferred
f Adjusted for maternal age at FET, BMI, No. of MII oocytes
g Adjusted for maternal age at FET, BMI,diminished ovarian reserve, normal fertilization rate, and endometrial thickness
h Adjusted for maternal age at FET, BMI and FSH

Discussion

To the best of our knowledge, this is the first study comparing the impact of different endometrial preparation regimens among patients with endometriosis. According to our results, HRT with GnRHa pretreatment did not show any extra benefit in women with endometriosis compared with HRT and NC, neither in clinical pregnancy outcomes nor perinatal outcomes.
Extensive clinical and molecular data support the existence of biological differences in the eutopic endometrium of women with endometriosis [1518].The transcriptomic analysis confirmed these earlier observations [19]. Previous studies have reported endometrial gene expression changes associated with defective endometrial receptivity, which reflected a shift away from normal progesterone action and toward excessive estrogen activity [2, 20]. Continuous hypophyseal exposure to GnRHa can deprive the main growth stimulus, therefore long-term therapy with GnRHa has been reported to be effective in treating symptomatic endometriosis [21]. The treatment desensitizes the pituitary gland and contributes to the hypogonadotropic-hypogonadal state, which leads to prolonged amenorrhoea and a low estradiol level. Not only does this improve endometriosis symptoms, but it also reverses the negative effects of endometriosis on ART including poor folliculogenesis leading to decreased oocyte quality, hostile peritoneal environment, and et al. [7]. Various clinical studies have claimed that poor oocyte quality results in impaired implantation rates [2225].
However, whether this treatment improves fecundity is equivocal. Several studies have investigated the effect of long-term treatment with GnRHa before IVF cycles on women with endometriosis-related infertility [2629]. In two Cochrane reviews, the authors reach opposite conclusions [6, 7]. Sallam et al. [6] provided evidence of the association of long-term pituitary down-regulation with GnRHa prior to standard IVF/ICSI with higher LBR and CPR in women with endometriosis, making it the first-choice treatment for such patients [30]. On contrary, Georgiou et al. concluded that there was no such benefit and that this treatment was not associated with complication rate and the number of oocytes retrieved, and the number of embryos [7]. In fact, many clinicians are also skeptical of its effectiveness. Given the possible benefit of GnRHa administration for endometriosis and adenomyoma, there are studies focused on different protocols for endometrial preparation during FET cycles. Studies exploring whether GnRHa+ HRT regimen is superior to HRT regimen in patients with adenomyoma have come to different conclusions [8, 9].
In the present study, we analyzed the three most commonly used protocols for endometrial preparation prior to FET in patients with endometriosis. In view of the possible effects of ART on neonatal health [31, 32], prenatal outcomes were also taken into account in addition to pregnancy outcomes. The results showedthat no protocol was superior to the others. Pretreatment of GnRHa did not increase live birth rate and clinical pregnancy rate, but neither did miscarriage rate nor perinatal complications. One possible explanation may be that the GnRHa treatment in our study was only 1 month, whereas previous studies that found benefits tended to treat patients for 3-6 months. Based on the assumption that GnRHa pretreatment would be beneficial to ART in women with endometriosis, 1 month of duration may not be sufficient. What we cannot ignore, however, is that its effect is yet to be explored, so this result is expected. As for the comparison of natural and hormone replacement cycles, varieties of studies have given insights into different patient populations [3337]. The present study firstly concentrated on endometriosis patients and found no superiority of either protocol.
The study is strengthened by the large cohort size and generally complete baseline and cycle data. Anyway, we cannot exclude a selection bias due to the retrospective design of the study. Although we tried to eliminate confounding factors, it is indisputable that there are inevitably some confounding factors that have not been taken into account. Further randomized control trials are required to determine the impact of the different endometrial preparation protocols in endometriosis patients. Moreover, the importance of investigating the neonatal outcomes and long-term follow-up of children born from frozen embryos [32, 38] and the psychological effects cannot be ignored [3941]. Notably, there are other overlooked conditions that require the exploration of FET strategies other than endometriosis, such as the diagnosis of cancer [4245] and thyroid autoimmunity [4648]. Fertility preservation is a crucial issue to be addressed in all cancer patients of reproductive age, and the safety of different ART strategies for them warrants careful investigation. Furthermore, the role of inositols supplementation in ART has also been reported recently [4953]. And the influence of cryptic sperm defects on pregnancy outcomes should not be ignored [54].
In conclusion, our findings indicated that endometrial preparation regimen selection of natural cycle, hormone replacement cycle, or hormone replacement treatment with GnRHa pretreatment had no beneficial or detrimental effects on pregnancy and perinatal outcomes in patients with endometriosis.

Acknowledgments

The authors thank the staff of the Reproductive Medicine Center and the obstetric ward in Tongji hospital for their cooperation and support.

Declarations

Approval for this study was obtained from the Institutional Review Board of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
All co-authors have seen and approved the final version of the paper and have agreed to its submission for publication.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Zondervan KT, Becker CM, Missmer SA. Endometriosis. N Engl J Med. 2020;382(13):1244–56.CrossRef Zondervan KT, Becker CM, Missmer SA. Endometriosis. N Engl J Med. 2020;382(13):1244–56.CrossRef
2.
Zurück zum Zitat Lessey BA, Kim JJ. Endometrial receptivity in the eutopic endometrium of women with endometriosis: it is affected, and let me show you why. Fertil Steril. 2017;108(1):19–27.CrossRef Lessey BA, Kim JJ. Endometrial receptivity in the eutopic endometrium of women with endometriosis: it is affected, and let me show you why. Fertil Steril. 2017;108(1):19–27.CrossRef
3.
Zurück zum Zitat Vallve-Juanico J, Houshdaran S, Giudice LC. The endometrial immune environment of women with endometriosis. Hum Reprod Update. 2019;25(5):564–91.CrossRef Vallve-Juanico J, Houshdaran S, Giudice LC. The endometrial immune environment of women with endometriosis. Hum Reprod Update. 2019;25(5):564–91.CrossRef
4.
Zurück zum Zitat Glujovsky D, Pesce R, Sueldo C, Quinteiro Retamar AM, Hart RJ, Ciapponi A. Endometrial preparation for women undergoing embryo transfer with frozen embryos or embryos derived from donor oocytes. Cochrane Database Syst Rev. 2020;10:CD006359. Glujovsky D, Pesce R, Sueldo C, Quinteiro Retamar AM, Hart RJ, Ciapponi A. Endometrial preparation for women undergoing embryo transfer with frozen embryos or embryos derived from donor oocytes. Cochrane Database Syst Rev. 2020;10:CD006359.
5.
Zurück zum Zitat Mumusoglu S, Polat M, Ozbek IY, Bozdag G, Papanikolaou EG, Esteves SC, et al. Preparation of the endometrium for frozen embryo transfer: a systematic review. Front Endocrinol (Lausanne). 2021;12:688237. Mumusoglu S, Polat M, Ozbek IY, Bozdag G, Papanikolaou EG, Esteves SC, et al. Preparation of the endometrium for frozen embryo transfer: a systematic review. Front Endocrinol (Lausanne). 2021;12:688237.
6.
Zurück zum Zitat Sallam HN, Garcia-Velasco JA, Dias S, Arici A. Long-term pituitary down-regulation before in vitro fertilization (IVF) for women with endometriosis. Cochrane Db Syst Rev. 2006;2006(1):CD004635. Sallam HN, Garcia-Velasco JA, Dias S, Arici A. Long-term pituitary down-regulation before in vitro fertilization (IVF) for women with endometriosis. Cochrane Db Syst Rev. 2006;2006(1):CD004635.
7.
Zurück zum Zitat Georgiou EX, Melo P, Baker PE, Sallam HN, Arici A, Garcia-Velasco JA, et al. Long-term GnRH agonist therapy before in vitro fertilisation (IVF) for improving fertility outcomes in women with endometriosis. Cochrane Db Syst Rev. 2019;2019(11):CD013240. Georgiou EX, Melo P, Baker PE, Sallam HN, Arici A, Garcia-Velasco JA, et al. Long-term GnRH agonist therapy before in vitro fertilisation (IVF) for improving fertility outcomes in women with endometriosis. Cochrane Db Syst Rev. 2019;2019(11):CD013240.
8.
Zurück zum Zitat Niu ZH, Chen Q, Sun YJ, Feng Y. Long-term pituitary downregulation before frozen embryo transfer could improve pregnancy outcomes in women with adenomyosis. Gynecol Endocrinol. 2013;29(12):1026–30.CrossRef Niu ZH, Chen Q, Sun YJ, Feng Y. Long-term pituitary downregulation before frozen embryo transfer could improve pregnancy outcomes in women with adenomyosis. Gynecol Endocrinol. 2013;29(12):1026–30.CrossRef
9.
Zurück zum Zitat Li MZ, Xu LH, Zhao H, Du YB, Yan L. Effects of artificial cycles with and without gonadotropin-releasing hormone agonist pretreatment on frozen embryo transfer outcomes in patients with adenomyosis. Sci Rep-Uk. 2021;11(1):19326.CrossRef Li MZ, Xu LH, Zhao H, Du YB, Yan L. Effects of artificial cycles with and without gonadotropin-releasing hormone agonist pretreatment on frozen embryo transfer outcomes in patients with adenomyosis. Sci Rep-Uk. 2021;11(1):19326.CrossRef
10.
Zurück zum Zitat Huang B, Hu D, Qian K, Ai JH, Li YF, Jin L, et al. Is frozen embryo transfer cycle associated with a significantly lower incidence of ectopic pregnancy? An analysis of more than 30,000 cycles. Fertil Steril. 2014;102(5):1345–9.CrossRef Huang B, Hu D, Qian K, Ai JH, Li YF, Jin L, et al. Is frozen embryo transfer cycle associated with a significantly lower incidence of ectopic pregnancy? An analysis of more than 30,000 cycles. Fertil Steril. 2014;102(5):1345–9.CrossRef
11.
Zurück zum Zitat Hu SQ, Xu B, Long R, Jin L. Pregnancy and perinatal outcomes in pregnancies resulting from time interval between a freeze-all cycle and a subsequent frozen-thawed single blastocyst transfer. Bmc Pregnancy Childb. 2020;20(1):161.CrossRef Hu SQ, Xu B, Long R, Jin L. Pregnancy and perinatal outcomes in pregnancies resulting from time interval between a freeze-all cycle and a subsequent frozen-thawed single blastocyst transfer. Bmc Pregnancy Childb. 2020;20(1):161.CrossRef
12.
Zurück zum Zitat Yang W, Zhang T, Li Z, Ren X, Huang B, Zhu G, et al. Combined analysis of endometrial thickness and pattern in predicting clinical outcomes of frozen embryo transfer cycles with morphological good-quality blastocyst: a retrospective cohort study. Medicine (Baltimore). 2018;97(2):e9577.CrossRef Yang W, Zhang T, Li Z, Ren X, Huang B, Zhu G, et al. Combined analysis of endometrial thickness and pattern in predicting clinical outcomes of frozen embryo transfer cycles with morphological good-quality blastocyst: a retrospective cohort study. Medicine (Baltimore). 2018;97(2):e9577.CrossRef
13.
Zurück zum Zitat Zhu LX, Xi QS, Zhang HW, Li YF, Ai JH, Jin L. Blastocyst culture and cryopreservation to optimize clinical outcomes of warming cycles. Reprod BioMed Online. 2013;27(2):154–60.CrossRef Zhu LX, Xi QS, Zhang HW, Li YF, Ai JH, Jin L. Blastocyst culture and cryopreservation to optimize clinical outcomes of warming cycles. Reprod BioMed Online. 2013;27(2):154–60.CrossRef
14.
Zurück zum Zitat Dai L, Deng C, Li Y, Zhu J, Mu Y, Deng Y, et al. Birth weight reference percentiles for Chinese. PLoS One. 2014;9(8):e104779.CrossRef Dai L, Deng C, Li Y, Zhu J, Mu Y, Deng Y, et al. Birth weight reference percentiles for Chinese. PLoS One. 2014;9(8):e104779.CrossRef
15.
Zurück zum Zitat Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase expression in endometriosis. J Clin Endocrinol Metab. 1996;81(1):174–9. Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase expression in endometriosis. J Clin Endocrinol Metab. 1996;81(1):174–9.
16.
Zurück zum Zitat Noble LS, Takayama K, Zeitoun KM, Putman JM, Johns DA, Hinshelwood MM, et al. Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J Clin Endocrinol Metab. 1997;82(2):600–6. Noble LS, Takayama K, Zeitoun KM, Putman JM, Johns DA, Hinshelwood MM, et al. Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J Clin Endocrinol Metab. 1997;82(2):600–6.
17.
Zurück zum Zitat Tseng JF, Ryan IP, Milam TD, Murai JT, Schriock ED, Landers DV, et al. Interleukin-6 secretion in vitro is up-regulated in ectopic and eutopic endometrial stromal cells from women with endometriosis. J Clin Endocrinol Metab. 1996;81(3):1118–22. Tseng JF, Ryan IP, Milam TD, Murai JT, Schriock ED, Landers DV, et al. Interleukin-6 secretion in vitro is up-regulated in ectopic and eutopic endometrial stromal cells from women with endometriosis. J Clin Endocrinol Metab. 1996;81(3):1118–22.
18.
Zurück zum Zitat Ota H, Igarashi S, Sasaki M, Tanaka T. Distribution of cyclooxygenase-2 in eutopic and ectopic endometrium in endometriosis and adenomyosis. Hum Reprod. 2001;16(3):561–6.CrossRef Ota H, Igarashi S, Sasaki M, Tanaka T. Distribution of cyclooxygenase-2 in eutopic and ectopic endometrium in endometriosis and adenomyosis. Hum Reprod. 2001;16(3):561–6.CrossRef
19.
Zurück zum Zitat Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, et al. Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003;144(7):2870–81.CrossRef Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, et al. Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003;144(7):2870–81.CrossRef
20.
Zurück zum Zitat Aghajanova L, Velarde MC, Giudice LC. Altered gene expression profiling in endometrium: evidence for progesterone resistance. Semin Reprod Med. 2010;28(1):51–8.CrossRef Aghajanova L, Velarde MC, Giudice LC. Altered gene expression profiling in endometrium: evidence for progesterone resistance. Semin Reprod Med. 2010;28(1):51–8.CrossRef
21.
Zurück zum Zitat Surrey ES. Gonadotropin-releasing hormone agonist and add-back therapy: what do the data show? Curr Opin Obstet Gynecol. 2010;22(4):283–8.CrossRef Surrey ES. Gonadotropin-releasing hormone agonist and add-back therapy: what do the data show? Curr Opin Obstet Gynecol. 2010;22(4):283–8.CrossRef
22.
Zurück zum Zitat Pellicer A, Navarro J, Bosch E, Garrido N, Garcia-Velasco JA, Remohi J, et al. Endometrial quality in infertile women with endometriosis. Ann N Y Acad Sci. 2001;943:122–30.CrossRef Pellicer A, Navarro J, Bosch E, Garrido N, Garcia-Velasco JA, Remohi J, et al. Endometrial quality in infertile women with endometriosis. Ann N Y Acad Sci. 2001;943:122–30.CrossRef
23.
Zurück zum Zitat Garrido N, Navarro J, Remohi J, Simon C, Pellicer A. Follicular hormonal environment and embryo quality in women with endometriosis. Hum Reprod Update. 2000;6(1):67–74.CrossRef Garrido N, Navarro J, Remohi J, Simon C, Pellicer A. Follicular hormonal environment and embryo quality in women with endometriosis. Hum Reprod Update. 2000;6(1):67–74.CrossRef
24.
Zurück zum Zitat Pal L, Shifren JL, Isaacson KB, Chang YC, Leykin L, Toth TL. Impact of varying stages of endometriosis on the outcome of in vitro fertilization-embryo transfer. J Assist Reprod Genet. 1998;15(1):27–31.CrossRef Pal L, Shifren JL, Isaacson KB, Chang YC, Leykin L, Toth TL. Impact of varying stages of endometriosis on the outcome of in vitro fertilization-embryo transfer. J Assist Reprod Genet. 1998;15(1):27–31.CrossRef
25.
Zurück zum Zitat Bergendal A, Naffah S, Nagy C, Bergqvist A, Sjoblom P, Hillensjo T. Outcome of IVF in patients with endometriosis in comparison with tubal-factor infertility. J Assist Reprod Genet. 1998;15(9):530–4.CrossRef Bergendal A, Naffah S, Nagy C, Bergqvist A, Sjoblom P, Hillensjo T. Outcome of IVF in patients with endometriosis in comparison with tubal-factor infertility. J Assist Reprod Genet. 1998;15(9):530–4.CrossRef
26.
Zurück zum Zitat Ma C, Qiao J, Liu P, Chen G. Ovarian suppression treatment prior to in-vitro fertilization and embryo transfer in Chinese women with stage III or IV endometriosis. Int J Gynaecol Obstet. 2008;100(2):167–70.CrossRef Ma C, Qiao J, Liu P, Chen G. Ovarian suppression treatment prior to in-vitro fertilization and embryo transfer in Chinese women with stage III or IV endometriosis. Int J Gynaecol Obstet. 2008;100(2):167–70.CrossRef
27.
Zurück zum Zitat Marcus SF, Edwards RG. High rates of pregnancy after long-term down-regulation of women with severe endometriosis. Am J Obstet Gynecol. 1994;171(3):812–7.CrossRef Marcus SF, Edwards RG. High rates of pregnancy after long-term down-regulation of women with severe endometriosis. Am J Obstet Gynecol. 1994;171(3):812–7.CrossRef
28.
Zurück zum Zitat van der Houwen LE, Mijatovic V, Leemhuis E, Schats R, Heymans MW, Lambalk CB, et al. Efficacy and safety of IVF/ICSI in patients with severe endometriosis after long-term pituitary down-regulation. Reprod BioMed Online. 2014;28(1):39–46.CrossRef van der Houwen LE, Mijatovic V, Leemhuis E, Schats R, Heymans MW, Lambalk CB, et al. Efficacy and safety of IVF/ICSI in patients with severe endometriosis after long-term pituitary down-regulation. Reprod BioMed Online. 2014;28(1):39–46.CrossRef
29.
Zurück zum Zitat Nakamura K, Oosawa M, Kondou I, Inagaki S, Shibata H, Narita O, et al. Menotropin stimulation after prolonged gonadotropin releasing hormone agonist pretreatment for in vitro fertilization in patients with endometriosis. J Assist Reprod Genet. 1992;9(2):113–7.CrossRef Nakamura K, Oosawa M, Kondou I, Inagaki S, Shibata H, Narita O, et al. Menotropin stimulation after prolonged gonadotropin releasing hormone agonist pretreatment for in vitro fertilization in patients with endometriosis. J Assist Reprod Genet. 1992;9(2):113–7.CrossRef
30.
Zurück zum Zitat Dunselman G, Vermeulen N, Nelen W. The 2013 ESHRE guideline on the management of women with endometriosis. Hum Reprod. 2013;28:86.CrossRef Dunselman G, Vermeulen N, Nelen W. The 2013 ESHRE guideline on the management of women with endometriosis. Hum Reprod. 2013;28:86.CrossRef
31.
Zurück zum Zitat Berntsen S, Söderström-Anttila V, Wennerholm U-B, Laivuori H, Loft A, Oldereid NB, et al. The health of children conceived by ART: 'the chicken or the egg?'. Hum Reprod Update. 2019;25(2):137–58.CrossRef Berntsen S, Söderström-Anttila V, Wennerholm U-B, Laivuori H, Loft A, Oldereid NB, et al. The health of children conceived by ART: 'the chicken or the egg?'. Hum Reprod Update. 2019;25(2):137–58.CrossRef
32.
Zurück zum Zitat Gullo G, Scaglione M, Cucinella G, Chiantera V, Perino A, Greco ME, et al. Neonatal outcomes and Long-term follow-up of children born from frozen embryo, a narrative review of latest research findings. Medicina (Kaunas). 2022;58(9):1218.CrossRef Gullo G, Scaglione M, Cucinella G, Chiantera V, Perino A, Greco ME, et al. Neonatal outcomes and Long-term follow-up of children born from frozen embryo, a narrative review of latest research findings. Medicina (Kaunas). 2022;58(9):1218.CrossRef
33.
Zurück zum Zitat Xu J, Li S-Z, Yin M-N, Liang P-L, Li P, Sun L. Endometrial preparation for frozen-thawed embryo transfer with or without pretreatment with GnRH agonist: a randomized controlled trial at two centers. Front Endocrinol (Lausanne). 2021;12:722253.CrossRef Xu J, Li S-Z, Yin M-N, Liang P-L, Li P, Sun L. Endometrial preparation for frozen-thawed embryo transfer with or without pretreatment with GnRH agonist: a randomized controlled trial at two centers. Front Endocrinol (Lausanne). 2021;12:722253.CrossRef
34.
Zurück zum Zitat Hu KL, Zhang D, Li R. Endometrium preparation and perinatal outcomes in women undergoing single-blastocyst transfer in frozen cycles. Fertil Steril. 2021;115(6):1487–94.CrossRef Hu KL, Zhang D, Li R. Endometrium preparation and perinatal outcomes in women undergoing single-blastocyst transfer in frozen cycles. Fertil Steril. 2021;115(6):1487–94.CrossRef
35.
Zurück zum Zitat Mackens S, Santos-Ribeiro S, van de Vijver A, Racca A, Van Landuyt L, Tournaye H, et al. Frozen embryo transfer: a review on the optimal endometrial preparation and timing. Hum Reprod. 2017;32(11):2234–42.CrossRef Mackens S, Santos-Ribeiro S, van de Vijver A, Racca A, Van Landuyt L, Tournaye H, et al. Frozen embryo transfer: a review on the optimal endometrial preparation and timing. Hum Reprod. 2017;32(11):2234–42.CrossRef
36.
Zurück zum Zitat Tomas C, Alsbjerg B, Martikainen H, Humaidan P. Pregnancy loss after frozen-embryo transfer--a comparison of three protocols. Fertil Steril. 2012;98(5):1165–9.CrossRef Tomas C, Alsbjerg B, Martikainen H, Humaidan P. Pregnancy loss after frozen-embryo transfer--a comparison of three protocols. Fertil Steril. 2012;98(5):1165–9.CrossRef
37.
Zurück zum Zitat Asserhoj LL, Spangmose AL, Aaris Henningsen AK, Clausen TD, Ziebe S, Jensen RB, et al. Adverse obstetric and perinatal outcomes in 1,136 singleton pregnancies conceived after programmed frozen embryo transfer (FET) compared with natural cycle FET. Fertil Steril. 2021;115(4):947–56.CrossRef Asserhoj LL, Spangmose AL, Aaris Henningsen AK, Clausen TD, Ziebe S, Jensen RB, et al. Adverse obstetric and perinatal outcomes in 1,136 singleton pregnancies conceived after programmed frozen embryo transfer (FET) compared with natural cycle FET. Fertil Steril. 2021;115(4):947–56.CrossRef
38.
Zurück zum Zitat Gullo G, Scaglione M, Cucinella G, Perino A, Chiantera V, D’Anna R, et al. Impact of assisted reproduction techniques on the neuro-psycho-motor outcome of newborns: a critical appraisal. J Obstet Gynaecol. 2022;42(7):2583–7.CrossRef Gullo G, Scaglione M, Cucinella G, Perino A, Chiantera V, D’Anna R, et al. Impact of assisted reproduction techniques on the neuro-psycho-motor outcome of newborns: a critical appraisal. J Obstet Gynaecol. 2022;42(7):2583–7.CrossRef
39.
Zurück zum Zitat Burgio S, Polizzi C, Buzzaccarini G, Laganà AS, Gullo G, Perricone G, et al. Psychological variables in medically assisted reproduction: a systematic review. Prz Menopauzalny. 2022;21(1):47–63. Burgio S, Polizzi C, Buzzaccarini G, Laganà AS, Gullo G, Perricone G, et al. Psychological variables in medically assisted reproduction: a systematic review. Prz Menopauzalny. 2022;21(1):47–63.
40.
Zurück zum Zitat Cousineau TM, Domar AD. Psychological impact of infertility. Best Pract Res Clin Obstet Gynaecol. 2007;21(2):293–308.CrossRef Cousineau TM, Domar AD. Psychological impact of infertility. Best Pract Res Clin Obstet Gynaecol. 2007;21(2):293–308.CrossRef
41.
Zurück zum Zitat Frederiksen Y, Farver-Vestergaard I, Skovgård NG, Ingerslev HJ, Zachariae R. Efficacy of psychosocial interventions for psychological and pregnancy outcomes in infertile women and men: a systematic review and meta-analysis. BMJ Open. 2015;5(1):e006592.CrossRef Frederiksen Y, Farver-Vestergaard I, Skovgård NG, Ingerslev HJ, Zachariae R. Efficacy of psychosocial interventions for psychological and pregnancy outcomes in infertile women and men: a systematic review and meta-analysis. BMJ Open. 2015;5(1):e006592.CrossRef
42.
Zurück zum Zitat Zaami S, Melcarne R, Patrone R, Gullo G, Negro F, Napoletano G, et al. Oncofertility and reproductive counseling in patients with breast Cancer: a retrospective study. J Clin Med. 2022;11(5):1311.CrossRef Zaami S, Melcarne R, Patrone R, Gullo G, Negro F, Napoletano G, et al. Oncofertility and reproductive counseling in patients with breast Cancer: a retrospective study. J Clin Med. 2022;11(5):1311.CrossRef
43.
Zurück zum Zitat Tanos P, Dimitriou S, Gullo G, Tanos V. Biomolecular and genetic prognostic factors that can facilitate fertility-sparing treatment (FST) decision making in early stage endometrial Cancer (ES-EC): a systematic review. Int J Mol Sci. 2022;23(5):2653.CrossRef Tanos P, Dimitriou S, Gullo G, Tanos V. Biomolecular and genetic prognostic factors that can facilitate fertility-sparing treatment (FST) decision making in early stage endometrial Cancer (ES-EC): a systematic review. Int J Mol Sci. 2022;23(5):2653.CrossRef
44.
Zurück zum Zitat Zaami S, Stark M, Signore F, Gullo G, Marinelli E. Fertility preservation in female cancer sufferers: (only) a moral obligation? Eur J Contracept Reprod Health Care. 2022;27(4):335–40.CrossRef Zaami S, Stark M, Signore F, Gullo G, Marinelli E. Fertility preservation in female cancer sufferers: (only) a moral obligation? Eur J Contracept Reprod Health Care. 2022;27(4):335–40.CrossRef
45.
Zurück zum Zitat Mutlu L, Manavella DD, Gullo G, McNamara B, Santin AD, Patrizio P. Endometrial cancer in Reproductive Age: Fertility-Sparing Approach and Reproductive Outcomes. Cancers (Basel). 2022;14(21):5187.CrossRef Mutlu L, Manavella DD, Gullo G, McNamara B, Santin AD, Patrizio P. Endometrial cancer in Reproductive Age: Fertility-Sparing Approach and Reproductive Outcomes. Cancers (Basel). 2022;14(21):5187.CrossRef
46.
Zurück zum Zitat Medenica S, Abazovic D, Ljubić A, Vukovic J, Begovic A, Cucinella G, et al. The role of cell and gene therapies in the treatment of infertility in patients with thyroid autoimmunity. Int J Endocrinol. 2022;2022:4842316.CrossRef Medenica S, Abazovic D, Ljubić A, Vukovic J, Begovic A, Cucinella G, et al. The role of cell and gene therapies in the treatment of infertility in patients with thyroid autoimmunity. Int J Endocrinol. 2022;2022:4842316.CrossRef
47.
Zurück zum Zitat Weetman AP. Thyroid disease in pregnancy in 2011: Thyroid function--effects on mother and baby unraveled. Nat Rev Endocrinol. 2011;8(2):69–70.CrossRef Weetman AP. Thyroid disease in pregnancy in 2011: Thyroid function--effects on mother and baby unraveled. Nat Rev Endocrinol. 2011;8(2):69–70.CrossRef
48.
Zurück zum Zitat Weetman AP. Immunity, thyroid function and pregnancy: molecular mechanisms. Nat Rev Endocrinol. 2010;6(6):311–8.CrossRef Weetman AP. Immunity, thyroid function and pregnancy: molecular mechanisms. Nat Rev Endocrinol. 2010;6(6):311–8.CrossRef
49.
Zurück zum Zitat D'Anna R, Santamaria A, Giorgianni G, Vaiarelli A, Gullo G, Di Bari F, et al. Myo-inositol and melatonin in the menopausal transition. Gynecol Endocrinol. 2017;33(4):279–82.CrossRef D'Anna R, Santamaria A, Giorgianni G, Vaiarelli A, Gullo G, Di Bari F, et al. Myo-inositol and melatonin in the menopausal transition. Gynecol Endocrinol. 2017;33(4):279–82.CrossRef
50.
Zurück zum Zitat Bezerra Espinola MS, Laganà AS, Bilotta G, Gullo G, Aragona C, Unfer V. D-chiro-inositol induces ovulation in non-polycystic ovary syndrome (PCOS), non-insulin-resistant young women, likely by modulating aromatase expression: a report of 2 cases. Am J Case Rep. 2021;22:e932722.CrossRef Bezerra Espinola MS, Laganà AS, Bilotta G, Gullo G, Aragona C, Unfer V. D-chiro-inositol induces ovulation in non-polycystic ovary syndrome (PCOS), non-insulin-resistant young women, likely by modulating aromatase expression: a report of 2 cases. Am J Case Rep. 2021;22:e932722.CrossRef
51.
Zurück zum Zitat Prapas Y, Petousis S, Panagiotidis Y, Gullo G, Kasapi L, Papadeothodorou A, et al. Injection of embryo culture supernatant to the endometrial cavity does not affect outcomes in IVF/ICSI or oocyte donation cycles: a randomized clinical trial. Eur J Obstet Gynecol Reprod Biol. 2012;162(2):169–73.CrossRef Prapas Y, Petousis S, Panagiotidis Y, Gullo G, Kasapi L, Papadeothodorou A, et al. Injection of embryo culture supernatant to the endometrial cavity does not affect outcomes in IVF/ICSI or oocyte donation cycles: a randomized clinical trial. Eur J Obstet Gynecol Reprod Biol. 2012;162(2):169–73.CrossRef
52.
Zurück zum Zitat Gullo G, Carlomagno G, Unfer V, D'Anna R. Myo-inositol: from induction of ovulation to menopausal disorder management. Minerva Ginecol. 2015;67(5):485–6. Gullo G, Carlomagno G, Unfer V, D'Anna R. Myo-inositol: from induction of ovulation to menopausal disorder management. Minerva Ginecol. 2015;67(5):485–6.
53.
Zurück zum Zitat D'Anna R, Corrado F, Loddo S, Gullo G, Giunta L, Di Benedetto A. Myoinositol plus α-lactalbumin supplementation, insulin resistance and birth outcomes in women with gestational diabetes mellitus: a randomized, controlled study. Sci Rep-Uk. 2021;11(1):8866.CrossRef D'Anna R, Corrado F, Loddo S, Gullo G, Giunta L, Di Benedetto A. Myoinositol plus α-lactalbumin supplementation, insulin resistance and birth outcomes in women with gestational diabetes mellitus: a randomized, controlled study. Sci Rep-Uk. 2021;11(1):8866.CrossRef
54.
Zurück zum Zitat Goudakou M, Kalogeraki A, Matalliotakis I, Panagiotidis Y, Gullo G, Prapas Y. Cryptic sperm defects may be the cause for total fertilization failure in oocyte donor cycles. Reprod BioMed Online. 2012;24(2):148–52.CrossRef Goudakou M, Kalogeraki A, Matalliotakis I, Panagiotidis Y, Gullo G, Prapas Y. Cryptic sperm defects may be the cause for total fertilization failure in oocyte donor cycles. Reprod BioMed Online. 2012;24(2):148–52.CrossRef
Metadaten
Titel
Which endometrial preparation protocol provides better pregnancy and perinatal outcomes for endometriosis patients in frozen-thawed embryo transfer cycles? A retrospective study on 1413 patients
verfasst von
Yaxin Guo
Zishui Fang
Lin Yu
Xin Sun
Fei Li
Lei Jin
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Journal of Ovarian Research / Ausgabe 1/2023
Elektronische ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-023-01095-4

Weitere Artikel der Ausgabe 1/2023

Journal of Ovarian Research 1/2023 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.