• Sonuç bulunamadı

A randomized clinical trial of endometrial perfusion with granulocyte colony-stimulating factor in in vitro fertilization cycles: impact on endometrial thickness and clinical pregnancy rates

N/A
N/A
Protected

Academic year: 2022

Share "A randomized clinical trial of endometrial perfusion with granulocyte colony-stimulating factor in in vitro fertilization cycles: impact on endometrial thickness and clinical pregnancy rates"

Copied!
6
0
0

Yükleniyor.... (view fulltext now)

Tam metin

(1)

A randomized clinical trial of endometrial perfusion with

granulocyte colony-stimulating

factor in in vitro fertilization cycles:

impact on endometrial thickness and clinical pregnancy rates

David H. Barad, M.D.,a,bYao Yu, Ph.D.,a,bVitaly A. Kushnir, M.D.,a,bAya Shohat-Tal, Ph.D.,a,b Emanuela Lazzaroni, M.S.,a,bHo-Joon Lee, Ph.D.,a,band Norbert Gleicher, M.D.a,b

aThe Center for Human Reproduction (CHR), andbFoundation for Reproductive Medicine, New York, New York

Objective: To investigate whether granulocyte colony-stimulating factor (G-CSG) affects endometrial thickness, implantation rates, and clinical pregnancy rates in routine, unselected IVF cycles.

Design: Registered, individually randomized, two-group, parallel double-blinded placebo-controlled clinical trial.

Setting: Academically affiliated private clinical and research center.

Patient(s): 141 consecutive, unselected, consenting women with no history of renal disease, sickle cell disease, or malignancy who were undergoing IVF.

Intervention(s): Sealed, numbered, opaque envelopes assigned 73 patients to receive G-CSF (Filgrastim, Amgen, 300 mg/1.0 mL) and 68 to receive placebo (saline).

Main Outcome Measure(s): Endometrial thickness, clinical pregnancy, and embryo implantation rates.

Result(s): The mean age for the whole study group was 39.59  5.56 years (G-CSF: 39.79  5.13 years; placebo: 39.38  6.03 years).

Endometrial thickness statistically significantly increased over the 5-day observation period for the whole group by approximately 1.36 mm. The increase in the G-CSF group was not statistically significantly different from the control group. Statistical models looking at treatment effects on clinical pregnancy and implantation rates demonstrated no effect of G-CSF treatment. There were no adverse events for either treatment group.

Conclusion(s): In normal IVF patients, G-CSF does not affect endometrial thickness, implantation rates, or clinical pregnancy rates.

Because these results were obtained in an older patient population, they may not necessarily apply to younger women.

Clinical Trial Registration Number: NCT01202656. (Fertil SterilÒ2014;101:710–5. Ó2014 by American Society for Reproductive Medicine.)

Key Words: Endometrial thickness, G-CSF, granulocyte colony-stimulating factor, in vitro fertilization, pregnancy rates, randomized controlled trial

Discuss:You can discuss this article with its authors and with other ASRM members athttp://

fertstertforum.com/baraddh-endometrial-perfusion-thickness-ivf-pregnancy-rates/

Use your smartphone to scan this QR code and connect to the discussion forum for this article now.*

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

Received September 23, 2013; revised and accepted December 6, 2013; published online January 11, 2014.

D.H.B. has received research and grant support, travel funds, and speaker honoraria from various pharmaceutical and medical device companies, not related to topics presented in this article; is listed as an inventor on four already-awarded and other still-pending U.S. patents claiming beneficial ef- fects in diminished ovarian reserve (DOR) and embryo ploidy from dehydroepiandrosterone (DHEA) or from other androgen supplementations and has received patent royalties from this company; is listed as coinventor on a number of pending U.S. patents claiming diagnostic relevance for the assessment of triple CGG repeats on the FMR1 gene in determining risk toward DOR and related issues. Y.Y. has nothing to disclose. V.A.K. has nothing to disclose. A.S. has nothing to disclose. E.L. has nothing to disclose. H-J.L. has nothing to disclose. N.G. NG is owner of CHR, where this research was conducted and owns shares in Fertility Nutraceuticals, LLC, a producer of fertility supplements, and has received patent royalties from this company;

has received research and grant support, travel funds, and speaker honoraria from various pharmaceutical and medical device companies, not related to topics presented in this article; is listed as a inventor on four already-awarded and other still-pending U.S. patents claiming beneficial effects in diminished ovarian reserve (DOR) and embryo ploidy from dehydroepiandrosterone (DHEA) or from other androgen supplementations; is listed as coinventor on a number of pending U.S. patents claiming diagnostic relevance for the assessment of triple CGG repeats on the FMR1 gene in deter- mining risk toward DOR and related issues. A full listing of all patent information can be provided on request.

Supported by the Foundation for Reproductive Medicine, a not-for-profit medical research foundation, and by intramural funds from the Center for Human Reproduction.

Reprint requests: David H. Barad, M.D., Center for Human Reproduction, 21 East 69th Street, New York, New York 10021 (E-mail:dbarad@thechr.com).

Fertility and Sterility® Vol. 101, No. 3, March 2014 0015-0282/$36.00

Copyright ©2014 American Society for Reproductive Medicine, Published by Elsevier Inc.

http://dx.doi.org/10.1016/j.fertnstert.2013.12.016

(2)

C

linical pregnancy rates after embryo transfer increase with increasing endometrial thickness(1–6). A large number of studies have defined minimal thickness at approximately 7 mm(7–12). Most in vitro fertilization (IVF) patients reach this minimal thickness with routine protocols. Only a small number of IVF patients fail to achieve normal endometrial thickness with routine treatments, and no established treatments exist for such patients. Sildenafil citrate (Viagra) treatment has been recommended (13), but response is not consistent. There is evidence of increased endometrial thickness among postmenopausal women using antihypertensive beta- blockers such as atenolol (Tenormin)(14, 15), but no formal studies have been reported before embryo transfer to improve endometrial thickness. Even when given vasodilators, many women fail to reach minimal endometrial thickness and demonstrate low pregnancy chances (7–12). Thus, women who fail to achieve minimal endometrial development often do not undergo embryo transfer. A new therapeutic approach to improve endometrial thickness in such patients would be very desirable.

Recent reports from our center have suggested that intra- uterine perfusion with granulocyte colony-stimulating factor (G-CSF) may be effective in women who are otherwise resis- tant to treatment (16, 17). Granulocyte colony-stimulating factor is a cytokine that stimulates neutrophilic granulocyte proliferation and differentiation. It is routinely and safely used in the treatment of neutropenia during cancer chemo- therapy (18, 19). A U.S. patent (US 2009/0226397 A1, September 10, 2009) has claimed a benefit from G-CSF treatment in cases of implantation failure and repeated pregnancy loss, suggesting that G-CSF can affect the endometrium.

Human decidua is now thought to some degree to control trophoblast invasion via secretion of cytokines (20–23). Colony-stimulating factors (CSF) are a family of proteins (GM-CSF, CSF-1, G-CSF, and IL-3) that stimulate cellular proliferation and the induction of terminal differentiation of hemopoietic progenitor cells. CSF-1 has been detected in first-trimester placental tissue and in endometrial glands (24–27). The osteopetrotic mouse, which represents a natural knockout of the CSF-1 gene, demonstrates a low rate of fetal implantation and poor fetal viability(28). Local injury to the endometrium, which may lead to local release of cytokines including CSF, has been claimed to improve rates of embryo implantations, clinical pregnancies, and live births in assisted reproduction(29). CSF-1 may also have a role in establishing early endometriotic lesions, suggesting that CSF may well promote growth of endometrial tissue(30).

In a double-blinded, randomized, controlled trial of 68 women with recurrent miscarriage (four sequential pregnancy losses) 29 (82.9%) out of 35 treated with G-CSF delivered healthy infants, whereas patients receiving placebo delivered only in 16 (48.5%) out of 33 cases (P¼ .0061, odds ratio [OR]

5.1; 95% confidence interval [CI], 1.5–18.4)(31).

Our center for over 10 years has been treating a small number of patients with implantation failure with G-CSF, off label and under appropriate written informed consent.

More recently, we reported successful treatment via intrauter- ine perfusions with G-CSF of patients with thin endometrium who were resistant to other routine treatments(16, 17). In a case series, we described 21 consecutive infertile women with endometria of <7 mm on day of human chorionic gonadotropin (hCG) administration in their first IVF cycles who were treated with 300 mg/1.0 mL of G-CSF (Nupogen, filgrastim; Amgen) administered by intrauterine catheter by slow infusion before noon. Endometrial thickness increased in these patients by 2.9  2.0 mm and did not vary between conception and nonconception cycles. A 19.1% ongoing clinical pregnancy rate was observed, excluding one ectopic pregnancy(16).

Given this endometrial proliferation and subsequent un- expectedly high pregnancy rates in this adversely selected pa- tient population, the question arose as to whether G-CSF perfusion may not also improve implantation and pregnancy rates in women with normal endometrial thickness. Our ran- domized controlled trial was designed to answer these ques- tions. We describe the results of a double-blinded, randomized, controlled trial of intrauterine infusion of G- CSF compared with normal saline (as placebo) on endometrial thickness, implantation, and pregnancy rates among unse- lected IVF patients with normal endometrial thickness who were undergoing IVF.

MATERIALS AND METHODS

This study was approved by the center’s institutional review board before initiation of the trial in 2010. All patients pre- senting for embryos transfer between October 3, 2010, and January 1, 2013, were offered participation in the trial.

Women with renal disease, sickle cell disease, or a history of malignancy were considered ineligible for medical reasons.

Presuming an implantation rate of 10% and anticipating a 10% increase to 20% with treatment, about 200 embryos transferred in each study arm would be needed for 80% power and alpha of 0.05. A total of 419 eligible patients were offered participation: 141 patients consented to participate, and 278 declined. The principal reasons for refusal to participate were ‘‘lack of interest’’ and technical difficulties in presenting in timely fashion for the treatment to the center (a large portion of our center’s patients are long-distance patients who only come to the center for retrieval and transfer). No consenting patient was excluded from participation for med- ical reasons.

Randomization was performed after the patients had con- sented to participate. A computer-generated randomization table was used with separate randomization blocks for pa- tients undergoing IVF and frozen embryo transfer (FET) in a first study cycle. Individual randomization cards were sealed in numbered opaque envelopes that were only accessible to the single staff member who administered the randomization table and prepared the study materials. Treatment assignment was blinded to patients, physicians, and nursing staff.

The flow chart for disposition of all patients is demon- strated inFigure 1. Of 141 participating patients, 129 under- went IVF, and 12 underwent FET in their first study cycle.

Randomization resulted in 73 patients (67 IVF, 6 FET)

VOL. 101 NO. 3 / MARCH 2014

(3)

assigned to G-CSF and 68 patients (62 IVF, 6 FET) to placebo in a first study cycle. All patients entering a second treatment cycle were offered participation in a crossover trial. Thirty- five patients agreed to participate in such a crossover cycle:

16 (14 IVF, 2 FET) placebo to G-CSF, and 19 (18 IVF, 1 FET) G-CSF to placebo. The patients who did not crossover either had conceived in their first study cycle or chose not to have another assisted reproduction cycle.

The G-CSF used was Nupogen (300 mg/1.0 mL, Filgras- tim; Amgen). One mL of G-CSF or placebo (normal saline) was administered on the morning of hCG administration before noon by slow transcervical intrauterine infusion, similar in technique to an intrauterine insemination. Endo- metrial thickness was assessed by routine vaginal ultra- sound before infusion and again 5 days later at the time of embryo transfer.

Pregnancy outcomes were assessed based on positive pregnancy test (chemical pregnancy) and clinical pregnancy (gestational sac and fetal heart on ultrasound examination).

Implantation rates were determined by the number of gesta- tional sacs at least 28 days after embryo transfer based on the total number of embryos transferred per group.

Embryo quality was assessed using standard day-3 and day-5 embryo grading. Day-3 embryos with eight cells and

<15% fragmentation were considered good. In day-5 em-

bryos, expanded blastocysts with at least grade B trophecto- derm and inner cell mass were considered good.

P< .05 was considered statistically significant covariates.

All analyses are conducted using SAS 9.2.

RESULTS

Baseline characteristics and outcomes are listed inTable 1for both the first and second (crossover) cycles of treatment. The mean age for the 141 patients undergoing their first study cy- cle was 39.59  5.56 years (G-CSF 39.79  5.13, and placebo 39.38  6.03; not statistically significant [NS]). The mean baseline follicle-stimulating hormone (FSH) level was 8.47

3.94 (G-CSF 8.64  3.92, and placebo 8.27  4.01). Mean baseline antim€ullerian hormone (AMH) level was 2.03  2.26 (G-CSF 2.07  2.17, and placebo 1.99  2.38). None of the baseline characteristics were statistically significantly different between the two groups.

The overall endometrial thickness statistically signifi- cantly increased over the 5-day observation period for the whole group by approximately 1.36 mm (95% CI, 0.98, 1.74; P< .0001; paired t-test) in the first study cycle; however, it did not statistically significantly increase over the second study cycle 0.27 mm (95% CI, 0.44, 0.98; P¼ .45).

We fit a general linear model (GLM) looking at the change in endometrial thickness from the day of infusion until the FIGURE 1

Consort flow diagram for the granulocyte colony-stimulating factor (G-CSF) randomized control trial (ClinicalTrials.govNCT01202656).

Barad. Endometrial perfusion with G-CSF in IVF. Fertil Steril 2014.

(4)

day of transfer to examine the effect of treatment adjusted for age (Fig. 2). The increase, while nominally greater in the G-CSF group, was not statistically significantly different.

Subdividing the initial endometrial thickness into four levels (endometrium %8.23 mm, 8.24–10.35, 10.36–12.47, and R12.48 mm) also revealed no differences between treatment

and placebo. Only six patients had a pretreatment endome- trial thickness of less than 7 mm.

Forty-eight patients had a positive hCG concentration af- ter their embryo transfer (24 in each group). Thirty-seven pa- tients established a clinical pregnancy in the first cycle of treatment: G-CSF 18 (24.7%) of 73, and placebo 19 (27.9%) of 68. Among these 37 clinical pregnancies, there were four spontaneous pregnancy losses (1 G-CSF; 3 placebo).

Fifty-eight patients who did not become pregnant did not choose to have another IVF cycle and were dropouts from the second cycle (30 G-CSF; 28 placebo). Thirty-five patients completed a crossover treatment cycle (16 G-CSF; 19 pla- cebo). For the 35 patients, the mean of washout time was 95 days. Clinical pregnancy was established in 15 patients: G- CSF 8 (50%) of 16, and placebo 7 (36.8%) of 19. Four of these established pregnancies miscarried, 2 in each group. There was no statistically significant difference in the overall preg- nancy rate between the first and second study cycles (P¼ .14, chi-square).

A logistic regression model (Table 2) looking at treatment effects on ongoing pregnancy rates in the first treatment cycle adjusted for patient age and endometrial thickness at day of first infusion demonstrated no effect of G-CSF on clinical pregnancy rates (OR 1.03; 95% CI, 0.46, 2.264; P¼ .95). As ex- pected, patient age was a marginally statistically significant negative predictor for ongoing pregnancy with an odds ratio of 0.936 (95% CI, 0.87, 1.01; P¼ .07). No adverse events occurred in either treatment group.

TABLE 1

Characteristics (baseline and outcome) of study patients.

Variable G-CSF Placebo

First cycle (N ¼ 141) n ¼ 73 n ¼ 68

Endometrium (day of hCG trigger) 10.23 ( 2.01) 10.37 ( 2.22)

Endometrium (day of embryo transfer) 11.69 ( 2.50) 11.63 ( 2.14)

Change in endometrial thickness 1.45 ( 2.22) 1.25 ( 2.28)

Age (y) 39.79 ( 5.13) 39.38 ( 6.03)

Baseline FSH 8.64 ( 3.92) 8.27 ( 4.01)

Baseline AMH 2.07 ( 2.17) 1.99 ( 2.38)

Oocytes retrieval (patient) 8.88 ( 7.22) 8.56 ( 5.83)

Pregnancy (hCGþ) cycle 1 18/73 (24.65%) 19/68 (27.94%)

SAB 1/18 (5.6%) 3/19 (15.8%)

Implantation rate 22/176 (12.5%) 28/168 (16.7%)

Clinical pregnancy (per hCGþ) 17/18 (94.4%) 16/19 (84.2%)

Good quality embryos 72/158 (45.57%) 55/140 (39.29%)

Second cycle (N ¼ 35)a n ¼ 19 n ¼ 16

Endometrium (day of hCG trigger) 9.96 ( 2.27) 11.10 ( 1.93)

Endometrium (day of embryo transfer) 10.32 ( 2.15) 11.40 ( 1.17)

Change in endometrial thickness 0.36 ( 2.13) 0.16 ( 2.01)

Age (y) 40.09 ( 5.20) 39.68 ( 3.81)

Baseline FSH 9.26 ( 3.73) 8.87 ( 4.14)

Baseline AMH 1.16 ( 0.93) 1.50 ( 2.69)

Oocytes retrieval (patient) 7.08 ( 6.38) 10.18 ( 6.46)

Pregnancy (hCGþ) cycle 2 7/19 (36.84%) 10/16 (62.5%)

SAB 2/7 (28.57%) 2/10 (20%)

Implantation rate 7/51 (13.7%) 11/48 (22.9%)

Clinical pregnancy (per hCGþ) 5/7 (70.43%) 6/10 (60%)

Good quality embryos 14/45 (31.11%) 21/ 48 (43.75%)

Overall implantation rate 33/224 (14.73%) 35/219 (15.98%)

Note: AMH ¼ antim€ullerian hormone; FSH ¼ follicle-stimulating hormone; G-CSF ¼ granulocyte colony-stimulating factor; hCG ¼ human chorionic gonadotropin; SAB ¼ spontaneous abortion.

aThe second cycle was a cross-over cycle. The 16 patients originally allocated to placebo received GCSF in the second cycle and the 19 received placebo.

Barad. Endometrial perfusion with G-CSF in IVF. Fertil Steril 2014.

FIGURE 2

Change in endometrial thickness in mm from day of hCG until day of transfer: C G-CSF; þ placebo. Mean change solid line (G-CSF);

dashed line (placebo).

Barad. Endometrial perfusion with G-CSF in IVF. Fertil Steril 2014.

VOL. 101 NO. 3 / MARCH 2014

(5)

A two-period crossover study analysis was conducted us- ing the data collected from 141 patients using GLM and linear mixed effect models (LME). Responses were the change of endometrial thickness and clinical pregnancy. Analysis of the crossover trial, while adjusting for patient’s age, demon- strated no effect of G-CSF on either the change in endometrial thickness or on clinical pregnancy. We, however, did observe a marginally greater increase in endometrial thickness in first study cycles compared with the second cycles in the G-CSF group (estimated difference 1.21; P¼ .038).

In total, 443 embryos were transferred in this study lead- ing to an implantation rate of 33 (14.73%) of 224 with G-CSF and 35 (15.98%) of 219 with placebo. The implantation rate was not statistically different (P¼ .72). The implantation rate for embryos in fresh IVF cycles of 61 (15.25%) of 400 was also not statistically significantly greater than that for cryopreserved embryos 7 (16.28%) of 43 (P¼ .88, chi-square).

We compared the embryo quality between the two treat- ment groups for 141 patients in first cycles only. There were nominally more good quality embryos in the G-CSF group [72/158 (45.57%)] compared with placebo [55/140 (39.29%)], but the difference was not statistically significant (df 174, t ¼ 1.03; P¼ .31). The odds ratio for higher embryo quality among the G-CSF treated group was 1.44 (95% CI, 0.71, 2.92; P¼ .31).

DISCUSSION

We previously reported successful endometrial expansion to at least minimal thickness of 7 mm after uterine perfusion with G-CSF in a small group of women with thin endome- trium resistant to standard treatments, who all expanded their endometrium and were able to proceed to embryo transfer and conceive(17). In a larger follow-up study, 21 patients with endometria <7 mm on the day of hCG administration were treated with G-CSF, resulting in significant endometrial expansion and a 19.1% ongoing pregnancy rate(16). Based on these observations, we launched this clinical trial to see whether G-CSF infusion could also improve IVF outcomes in random women undergoing IVF who had normal endome- trial thickness, hypothesizing that G-CSF might in addition to its proliferative effects on endometrium also beneficially affect implantation. Such a hypothesis is also supported by

published reports suggesting improved pregnancy survival after G-CSF treatments in habitual aborters and in women with presumed implantation failure(31, 32).

In this randomized clinical trial we were unable to detect a significant treatment effect of intrauterine perfusions with G-CSF on endometrial proliferation, implantation, or preg- nancy rates. This study was designed to detect a 20% increase in implantation rate, which clearly did not occur. The number of participants in this trial was not sufficient to detect a smaller increase; though any increase in implantation rate attributable to treatment might be important, it was not prac- tical to recruit the numbers of patients needed to detect smaller effects.

The results of this study, therefore, suggest that intrauter- ine perfusion of the endometrial cavity with G-CSF in women with normal endometrial proliferation does not offer any clin- ical benefit. Such a conclusion, however, still has to be viewed with caution: our center’s patients are to an unusual degree adversely selected. This is partially demonstrated by the rela- tive advanced age of the study population of approximately 40.0 years (39.58  5.40 years), and relatively high FSH and relatively low AMH levels. In addition, over 80% of women who present to our center for a first IVF cycle have previously failed IVF treatments elsewhere, often multiple times at multiple centers. Thus, our investigated patient pop- ulation does not represent a typical average patient popula- tion going through IVF cycles, even though the observed pregnancy rate of 32.6% in first cycles is quite good, consid- ering their adverse selection. Also notable are the observed implantation rates in the study and placebo groups, both around 15%.

We cannot preclude the possibility that G-CSF effects in a less adversely selected and especially younger patient popula- tion may be more favorable. Our noted nominal advantages in favor of G-CSF-treated patients could point toward such a possibility. More likely, however, G-CSF loses its effective- ness in the presence of a normally proliferating endometrium, or at least does so when infused into the endometrial cavity.

When treating habitual aborters and presumed implantation failure, others have administered G-CSF systemically by sub- cutaneous injection (31, 32). One, therefore, could further speculate that the systemic effects of G-CSF may differ from local effects, though such an explanation also appears unlikely, especially as G-CSF has demonstrated its effectiveness via local application in women with thin endometria resistant to standard treatments(16, 17).

In summary, this clinical trial failed to demonstrate a beneficial effect from intrauterine perfusion with G-CSF on IVF outcomes. This conclusion, however, may not be appli- cable to all patients, especially women younger than those investigated in our study.

REFERENCES

1. Richter KS, Bugge KR, Bromer JG, Levy MJ. Relationship between endome- trial thickness and embryo implantation, based on 1,294 cycles of in vitro fertilization with transfer of two blastocyst-stage embryos. Fertil Steril 2007;87:53–9.

2. Kovacs P, Matyas S, Boda K, Kaali SG. The effect of endometrial thickness on IVF/ICSI outcome. Hum Reprod 2003;18:2337–41.

TABLE 2

The logistic regression model on ongoing pregnancy based on 141 patients in their first cycle.

Parameter DF Estimate Standard error Wald c2 Pr > c2

Intercept 1 1.4053 1.4225 0.9760 0.3232

G-CSF 1 0.0128 0.2020 0.0040 0.9495

Age 1 0.0663 0.0365 3.2981 0.0694

Odds ratio estimates

Effect Point estimate 95% Wald confidence limits

G-CSF vs. placebo 1.026 0.465 2.264

Age 0.936 0.871 1.005

Note: G-CSF ¼ granulocyte colony-stimulating factor.

Barad. Endometrial perfusion with G-CSF in IVF. Fertil Steril 2014.

(6)

3. Noyes N, Liu HC, Sultan K, Schattman G, Rosenwaks Z. Endometrial thick- ness appears to be a significant factor in embryo implantation in in-vitro fertilization. Hum Reprod 1995;10:919–22.

4. Gonen Y, Casper RF, Jacobson W, Blankier J. Endometrial thickness and growth during ovarian stimulation: a possible predictor of implantation in in vitro fertilization. Fertil Steril 1989;52:446–50.

5. Traub ML, Van Arsdale A, Pal L, Jindal S, Santoro N. Endometrial thickness, Caucasian ethnicity, and age predict clinical pregnancy following fresh blas- tocyst embryo transfer: a retrospective cohort. Reprod Biol Endocrinol 2009;

7:33.

6. Check JH, Lurie D, Dietterich C, Callan C, Baker A. Adverse effect of a homo- geneous hyperechogenic endometrial sonographic pattern, despite adequate endometrial thickness on pregnancy rates following in-vitro fertil- ization. Hum Reprod 1993;8:1293–6.

7. Schild RL, Knobloch C, Dorn C, Fimmers R, van der Ven H, Hansmann M.

Endometrial receptivity in an in vitro fertilization program as assessed by spi- ral artery blood flow, endometrial thickness, endometrial volume, and uter- ine artery blood flow. Fertil Steril 2001;75:361–6.

8. El-Toukhy T, Coomarasamy A, Khairy M, Sunkara K, Seed P, Khalaf Y, et al.

The relationship between endometrial thickness and outcome of medicated frozen embryo replacement cycles. Fertil Steril 2008;89:832–9.

9. Bozdag G, Esinler I, Yarali H. The impact of endometrial thickness and texture on intracytoplasmic sperm injection outcome. J Reprod Med 2009;

54:303–11.

10. Remohi J, Ardiles G, Garcia-Velasco JA, Gaitan P, Simon C, Pellicer A. Endo- metrial thickness and serum oestradiol concentrations as predictors of outcome in oocyte donation. Hum Reprod 1997;12:2271–6.

11. Abdalla HI, Brooks AA, Johnson MR, Kirkland A, Thomas A, Studd JW. Endo- metrial thickness: a predictor of implantation in ovum recipients? Hum Re- prod 1994;9:363–5.

12. Okohue JE, Onuh SO, Ebeigbe P, Shaibu I, Wada I, Ikimalo JI, et al. The effect of endometrial thickness on in vitro fertilization (IVF)-embryo transfer/intracyto- plasmic sperm injection (ICSI) outcome. Afr J Reprod Health 2009;13:113–21. 13. Sher G, Fisch JD. Vaginal sildenafil (Viagra): a preliminary report of a novel method to improve uterine artery blood flow and endometrial development in patients undergoing IVF. Hum Reprod 2000;15:806–9.

14. Okman-Kilic T, Kucuk M. The effects of antihypertensive agents on endome- trial thickness in asymptomatic, hypertensive, postmenopausal women.

Menopause 2003;10:362–5.

15. Martinez-Rubio MP, Alcazar JL. Ultrasonographic and pathological endome- trial findings in asymptomatic postmenopausal women taking antihyperten- sive drugs. Maturitas 2003;46:27–32.

16. Gleicher N, Kim A, Michaeli T, Lee HJ, Shohat-Tal A, Lazzaroni E, et al. A pilot cohort study of granulocyte colony-stimulating factor in the treatment of un-

responsive thin endometrium resistant to standard therapies. Hum Reprod 2013;28:172–7.

17. Gleicher N, Vidali A, Barad DH. Successful treatment of unresponsive thin endometrium. Fertil Steril 2011;95:2123.e13–7.

18. Dale DC, Cottle TE, Fier CJ, Bolyard AA, Bonilla MA, Boxer LA, et al. Severe chronic neutropenia: treatment and follow-up of patients in the Severe Chronic Neutropenia International Registry. Am J Hematol 2003;72:82–93. 19. Gomez Raposo C, Pinto Marin A, Gonzalez Baron M. Colony-stimulating factors: clinical evidence for treatment and prophylaxis of chemotherapy- induced febrile neutropenia. Clin Transl Oncol 2006;8:729–34.

20. Loke Y, King A, Burrows T. Decidua in human implantation. Hum Reprod 1995;10:14.

21. Mathialagan N, Roberts RM. A role for cytokines in early pregnancy. Indian J Physiol Pharmacol 1994;38:153–62.

22. Giudice LC. Genes associated with embryonic attachment and implantation and the role of progesterone. J Reprod Med 1999;44:165–71.

23. Kauma SW. Cytokines in implantation. J Reprod Fertil Suppl 2000;55:31–42. 24. Daiter E, Pampfer S, Yeung YG, Barad D, Stanley ER, Pollard JW. Expression of colony-stimulating factor-1 in the human uterus and placenta. J Clin En- docrinol Metab 1992;74:850–8.

25. Pampfer S, Daiter E, Barad D, Pollard JW. Expression of the colony- stimulating factor-1 receptor (c-fms proto-oncogene product) in the human uterus and placenta. Biol Reprod 1992;46:48–57.

26. Saito S, Fukunaga R, Ichijo M, Nagata S. Expression of granulocyte colony- stimulating factor and its receptor at the fetomaternal interface in murine and human pregnancy. Growth Factors 1994;10:135–43.

27. Kauma SW, Aukerman SL, Eierman D, Turner T. Colony-stimulating factor-1 and c-fms expression in human endometrial tissues and placenta during the menstrual cycle and early pregnancy. J Clin Endocrinol Metab 1991;73:746–51. 28. Robertson SA, Roberts CT, Farr KL, Dunn AR, Seamark RF. Fertility impair- ment in granulocyte-macrophage colony-stimulating factor-deficient mice.

Biol Reprod 1999;60:251–61.

29. Zhou L, Li R, Wang R, Huang HX, Zhong K. Local injury to the endometrium in controlled ovarian hyperstimulation cycles improves implantation rates.

Fertil Steril 2008;89:1166–76.

30. Jensen JR, Witz CA, Schenken RS, Tekmal RR. A potential role for colony- stimulating factor 1 in the genesis of the early endometriotic lesion. Fertil Steril 2010;93:251–6.

31. Scarpellini F, Sbracia M. Use of granulocyte colony-stimulating factor for the treatment of unexplained recurrent miscarriage: a randomised controlled trial. Hum Reprod 2009;24:2703–8.

32. Santjohanser C, Knieper C, Franz C, Hirv K, Meri O, Schleyer M, et al. Gran- ulocyte-colony stimulating factor as treatment option in patients with recur- rent miscarriage. Arch Immunol Ther Exp (Warsz) 2013;61:159–64.

VOL. 101 NO. 3 / MARCH 2014

Referanslar

Benzer Belgeler

Benzer şekilde in- ternete bağlı çalışması gereken bazı ürünler de bir süre sonra merkezi sunucudaki değişikliklere adapte olamadı- ğı için tamamen kullanılamaz

the Military Museum, remained in the original building, but interest in this waned, and the collection lay virtually fo rg o tten here u n til 1909, when defence

Güneşin deri yaş- lanması ve deri kanseri gelişimi üzerine etkisine yönelik bilgi düzeyi grup 2’de grup 1’e kıyasla ista- tistiksel olarak anlamlı oranda yüksek saptandı

臺北醫學大學今日北醫-TMU Today: 首屆2010澳洲芳療大師Ron Guba講座#more 首屆2010澳洲芳療大師Ron

IVF-ICSI cycles of 57 patients who had previously experienced difficult ET; easy ET was performed with a soft catheter in 70% of the participants with the remaining

Objective: To define a novel experimental model with maternal intravenous (i.v.) granulocyte-colony stimulating factor (G-CSF) followed by a single- and high-dose of 20

The outcome measures were: obstetric outcome in terms of duration of labor, mode of delivery, and rate of cesarean section with indication; maternal outcomes in terms of postpartum

Keywords: Black hole Computation; Hypercomputation; Ultimate Computer; AdS/CFT correspondence; Holographic principle; Information theory; Rotating black holes;