Clin Exp Reprod Med Search

CLOSE


Clin Exp Reprod Med > Volume 52(2); 2025 > Article
Navali, Eghbali, Farzadi, Ghasemzadeh, Hamdi, Hakimi, Ghasemnejad-Berenji, and Sadeghpour: Amlodipine in the HRT cycle for frozen embryo transfer to correct uterine artery resistance in women with prior implantation failure: a randomized controlled trial

Abstract

Objective

For successful embryo implantation in women with high pulsatility, uterine vascular resistance and pulsatility must be reduced. We examined the effects of amlodipine on uterine pulsatility index (PI), resistance index (RI), and embryo transfer (ET) outcomes in women with prior implantation failure and at least one elevated uterine PI measurement (especially higher than 3).

Methods

Between February and November 2023, our reproductive facility conducted a single-center randomized clinical trial, enrolling 100 patients with previous implantation failure and at least one uterine PI measurement exceeding 3. Participants were randomly assigned to receive either amlodipine (5 mg) or placebo (n=50 per group). Hormone replacement therapy was the predominant method for endometrial preparation. Transvaginal ultrasonography was used to measure uterine artery resistance and pulsatility on day 1 or 2 of menstruation. Women in the amlodipine group received 5 mg nightly. Following repeat transvaginal ultrasound to assess PI and RI, ET was performed. If a positive pregnancy test was obtained, treatment continued for a total of 7 weeks.

Results

Amlodipine reduced blood flow indices in the uterine artery. Among placebo recipients, 18% tested positive for beta-human chorionic gonadotropin, compared to 26% of medication recipients. However, this difference was statistically insignificant (p=0.472). Gestational sacs were observed in 12% of the placebo group and 22% of the medication group, but this difference was also insignificant (p=0.28).

Conclusion

Amlodipine appears to reduce uterine pulsatility and resistance during ET. Despite the absence of significant differences in pregnancy outcomes, this promising drug merits further study in women with implantation failure.

Introduction

Numerous modifications and advancements have been made to in vitro fertilization (IVF) since its inception, relating to aspects such as ovarian stimulation and catheter selection. Despite these improvements, the implantation rate (IR) and clinical pregnancy rate (CPR) among women undergoing embryo transfer (ET) have consistently failed to meet expected outcomes. This discrepancy is attributed to various factors, including endometrial receptivity and embryo quality. Extensive research has been conducted to understand and improve the effectiveness of ET following IVF [1].
For successful embryo implantation in humans, the endometrium must demonstrate functional competency and receptivity [2]. Challenges arise from both endometrial and embryonic factors, which can obstruct the implantation process that is essential for pregnancy. Implantation failure can result from either a poor-quality embryo or issues with endometrial receptivity [3]. Factors such as endometrial thickness, uterine artery blood flow, and endometrial perfusion all contribute to endometrial receptivity and can potentially reduce the IR in IVF/intracytoplasmic sperm injection (ICSI) cycles [4].
Elevated impedance in uterine blood flow can lead to inadequate endometrial development and a thinning of the uterine lining. Optimal blood flow to the uterus is crucial for the healthy growth and receptivity of the endometrium [4]. Recent research has indicated that a high uterine artery pulsatility index (UtA-PI) is associated with a reduction in this flow, potentially resulting in implantation failure and pregnancy complications [5]. Women undergoing IVF/ICSI cycles with an elevated UtA-PI face relatively low rates of implantation and clinical pregnancy. Pharmacological intervention to address this issue could improve the outcomes of IVF/ICSI cycles [6].
Calcium channel blockers and vasodilators can increase endometrial thickness, promote uterine relaxation, and improve endometrial receptivity, thus potentially increasing the likelihood of a successful assisted pregnancy [7]. A range of vasodilators, including nifedipine, isosorbide mononitrate, pentoxifylline, glyceryl trinitrate, sildenafil, and amlodipine, have been utilized to address issues with endometrial thickening and uterine relaxation in the context of assisted reproductive technology (ART) [7].
Amlodipine is a long-acting oral dihydropyridine calcium channel blocker that is notable for its low incidence of adverse drug reactions associated with vasodilation [8]. It reduces the initial influx of calcium by blocking voltage-dependent L-type calcium channels [8]. As intracellular calcium decreases, the contractility of vascular smooth muscle is reduced, while smooth muscle relaxation and vasodilation are increased. Additionally, amlodipine improves endothelial function in blood vessels of individuals with hypertension, improving blood flow and contributing to its therapeutic efficacy [9]. The purpose of this study is to examine the effects of amlodipine administration prior to ET on uterine PI, resistance index (RI), and ET outcomes.

Methods

1. Study design, setting, and population

The study was conducted as a randomized clinical trial at our fertility center, adhering strictly to the principles of the Declaration of Helsinki and the Consolidated Standards for Reporting Trials (CONSORT) statement. Participants were recruited based on predefined inclusion and exclusion criteria. All prior unsuccessful cycles, including frozen embryo transfer (FET) cycles, were considered, and the women received a uniform hormone treatment protocol. After providing informed consent, participants were formally enrolled in the study. The Research Ethics Committees of Tabriz University of Medical Sciences conducted a thorough review and granted approval for the study, documented under the reference number IR.TBZMED.REC.1401.965.

2. Allocation and intervention

The women were randomly assigned to receive either amlodipine or placebo in a 1:1 ratio, based on a computer-generated randomization list. Allocation numbers were enclosed in sealed envelopes, which a non-participating nurse opened. Women in the amlodipine group were administered 5 mg of oral amlodipine (Zahravi Pharmaceutical Co.), while those in the placebo group received microcrystalline cellulose over the same period. A nurse not involved in the study managed the medication administration for both groups. To maintain blinding, the participants, healthcare providers, and laboratory personnel remained unaware of the group assignments. The codes for the treatment groups were revealed to the investigators only after the study and statistical analysis were completed.

3. Sample size determination

A study by Sohrabvand et al. [10] reported an overall pregnancy rate of 13.6% in the control group compared to 36% in the intervention group. We calculated a total sample size of 159 women, considering a type I error of 0.05, a power of 0.8, and an expected attrition rate of 20% for each group.

4. Clinical trial registration

This clinical trial was registered with the Iranian Registry of Clinical Trials (IRCT), with the assigned registration number IRCT20101227005485N11, on February 6, 2023. The trial protocol has been published at https://en.irct.ir/trial/68330.

5. Eligibility criteria

For inclusion, participants were required to be aged 20 to 45 years (inclusive); have a body mass index (BMI) of 25 to 30 kg/m2; be scheduled for cleavage-stage FET, with vitrification occurring on day 3, during a hormone replacement therapy (HRT) cycle; have at least one uterine PI exceeding 3, of the two measurements taken on the initial day of monitoring; and have a history of at least one failed ET. Furthermore, participants had to be candidates for a simple transfer test, involving the passage of a flexible or rigid catheter through the cervix without the need for anesthesia or Pozzi forceps, and to provide free written informed consent.
The exclusion criteria were as follows: active smokers (defined as at least one cigarette per day at the time of study registration); oocyte recipient patient; those scheduled to undergo pre-implantation genetic screening; patients with stage 3 or 4 endometriosis, or with severe adenomyosis or submucous myoma; individuals with contraindications to amlodipine use (severe angina pectoris, cyclosporine treatment, myocardial infarction less than 1 month prior, severe hepatic insufficiency, hypotension, or hypersensitivity to amlodipine or dihydropyridine derivatives); those concurrently using cytochrome P450 family 3 subfamily A member 4 (CYP3A4) enzyme inhibitors or inducers; those being treated for hypertension during the initial monitoring period; and participants in other human intervention research.

6. Procedures

Demographic data, including age, BMI, and duration of infertility, were extracted from the records of each participant, and a data collection form was completed for every individual. All participants received the standard FET protocol. To assess PI and RI, each woman underwent transvaginal color Doppler examination of the right and left uterine arteries on day 1 or 2 of her menstrual cycle. A single sonographer conducted all transvaginal color Doppler examinations.

7. Drug intervention

Following the cessation of ovarian hormone production, HRT was initiated with daily administration of 6 mg of estradiol. This regimen continued until a pregnancy test was performed. If the result was positive, HRT was maintained for the first 12 weeks of amenorrhea at the same dosage before being gradually tapered. In the event of a negative pregnancy test, HRT was slowly discontinued. The endometrial pattern and vascular perfusion were closely observed between the first and second days of the menstrual cycle. One group was administered 5 mg of oral amlodipine (Zahravi Pharmaceutical Co.), while the other received a placebo consisting of microcrystalline cellulose. The assigned treatment was maintained until pregnancy testing was conducted. If the result was positive, treatment (amlodipine or placebo) was continued for a total of 7 weeks.

8. Monitoring and evaluation

To establish baseline values for each patient, PI and RI were measured prior to the administration of amlodipine. These indices were measured again immediately before FET to assess the impact of amlodipine. Pulsed-wave Doppler ultrasound was performed on the first or second day of the participant’s menstrual cycle. Amlodipine treatment was initiated for the medication group. The evaluation of RI and PI in the uterine artery was conducted at the level of the cervical opening of the cervix into the body of the uterus, with concurrent visualization of the uterus and cervix. Doppler ultrasound was employed to assess endometrial blood flow, focusing on four areas: zone 1, where blood vessels enter the endometrial/myometrial tissue surrounding the endometrium; zone 2, where vessels enter the hyperechoic endometrial margin; zone 3, where vessels enter the internal hypoechoic zone of the endometrium; and zone 4, where vessels enter the endometrial cavity (Figure 1). Doppler sonography was performed using a vaginal probe operating at 7.5 MHz on an Affiniti 70 ultrasound machine (Philips), with the uterus in a sagittal view.

9. Embryo transfer

FET was performed 4 days after the initiation of progesterone therapy. The Kitazato kit was used to thaw the embryos and reassess their quality. The morphological criteria for embryo scoring at both cleavage and blastocyst stages were based on the Istanbul consensus [11] and the Gardner scoring criteria [12]. In accordance with the former, all grade 1 cleavage-stage embryos were defined as being of good quality. An embryo was considered to have survived if at least 50% of its cells were intact; those with a minimum of six cells and less than 20% intracellular fragmentation were also considered good quality. After thawing, the embryos deemed suitable for use were cultured for 2 hours. Subsequently, they were transferred into the uterine cavity under ultrasonographic guidance, with each procedure involving the transfer of 2–3 embryos. The transfers were conducted smoothly using a Cook catheter and ultrasound guidance, without the need for a tenaculum. Amlodipine administration was continued until pregnancy testing produced a positive result. Ultimately, we compared the mean values for blood flow indices in the uterine artery, the spiral artery supply zone, the endometrial thickness, and the clinical and chemical pregnancy rates between groups.

10. Outcomes

In this study, the primary outcomes were two indices of uterine artery blood flow: PI and RI. To measure these parameters, the Doppler wave was activated, and the angle of incidence was adjusted to record two comparable flow waves in the uterine artery. Simple semiquantitative methods were used to analyze the Doppler waveforms, incorporating the measurement of spectral characteristics such as the PI and RI. Radiologists utilized Doppler ultrasound to measure these characteristics for each participant in both study groups. Secondary outcomes included IR, as well as clinical and chemical pregnancy rates. IR was determined by dividing the number of gestational sacs by the number of embryos transferred to the uterus. Fourteen days after transfer, beta-human chorionic gonadotropin (β-hCG) testing was performed to detect chemical pregnancy. Clinical pregnancy was confirmed if ultrasound revealed a gestational sac 4 weeks after the transfer.

11. Statistical analysis

SPSS ver. 16 (SPSS Inc.) was utilized for data analysis. The chi-square test, the independent t-test, and analysis of variance were used to examine differences in variables between the two groups. A p-value of less than 0.05 was considered to indicate statistical significance.

12. Ethical approval

The Ethics Committee has no objections to the implementation of the study project. All methods were conducted in accordance with relevant guidelines and regulations. Informed consent was obtained from all subjects or their legal guardians under the Ethical Approval and Consent to Participate section.

Results

Between February and November 2023, 159 women were screened. Of these, 20 did not meet the selection criteria, and 25 declined to participate. Consequently, 114 participants were randomly assigned to two groups. Seven patients from the amlodipine group and seven from the control group were later excluded due to unwillingness to continue. Ultimately, 100 cases were analyzed, with 50 in the treatment group and 50 in the placebo group. The study flowchart is presented in Figure 2. No significant differences were found between groups regarding demographic variables such as the duration of infertility, BMI, age, endometrial thickness, the number of embryos transferred, and the frequency of fertility treatments (Table 1). This indicates that the randomization of the study groups was successful. The dependent variables exhibited normal distributions. Notably, 18% of placebo recipients tested positive for β-hCG, compared to 26% of medication recipients; nevertheless, this difference was not statistically significant (p=0.472). Additionally, 12% of the placebo group exhibited a gestational sac, while 22% of the medication group did so; this difference was also statistically insignificant (p=0.287). However, a significant distinction was observed between groups regarding the ultrasound pattern of the endometrium (p=0.007). A significant difference was also noted in the distribution of endometrial blood supply zones between the drug and placebo recipients (p<0.001) (Table 2). The UtA-PI and the uterine endometrial RI were assessed in both groups. The UtA-PI was 2.54±0.52 in the drug group and 3.5±0.89 in the placebo group, representing a significant difference (p<0.001). The endometrial RI was 0.83±0.05 in the treatment group and 0.91±0.053 in the placebo group, which was also a statistically significant finding (p<0.001) (Table 3, Figure 3). Pregnancy outcomes are presented in Table 4.

Discussion

The success of ART cycles is highly dependent on the receptivity of the endometrium [13]. The objective of endometrial preparation is to establish optimal conditions for embryo implantation. With advancements in ART, increasing attention has been dedicated to identifying the ideal state of endometrial preparation to maximize the chances of successful pregnancy [14].
The spiral arteries originate from the uterine arteries, and the resistance levels of these vessels are directly related. Consequently, assessing the resistance in the uterine arteries can provide valuable insights into the potential for endometrial implantation. Several studies have indicated that when the PI of the uterine arteries exceeds 3.2, IRs are notably low [15-17]. This suggests that procedures such as ET or intrauterine insemination should be deferred. Blood flow in the uterine arteries is crucial for promoting endometrial growth, supporting the luteal phase, and influencing the duration of pregnancy. Previous research has established a strong relationship between Doppler measurements of the uterine arteries and various complications, including recurrent spontaneous abortion, repeated implantation failure, preeclampsia, and intrauterine fetal growth restriction [18-20]. Timely intervention based on these measurements can help reduce pregnancy complications and improve outcomes for both mother and fetus. On Doppler ultrasound, a high PI is an independent risk factor for implantation failure and pregnancy complications [13]. Thus, it is worthwhile to investigate pharmacological interventions to reduce the UtA-PI prior to ET. These interventions could include various drug classes, such as calcium channel blockers and vasodilators [7,21].
The recommended maximum daily dosage of amlodipine is 10 mg [22]. Amlodipine is characterized by high bioavailability, minimal fluctuations in plasma concentration, and the achievement of a stable plasma concentration within 7 to 8 days following the initiation of a 5 mg daily oral dose. Research indicates that amlodipine exposure during pregnancy does not increase the incidence of embryonic or fetal abnormalities relative to other antihypertensive medications or untreated maternal hypertension [23-25]. A recent meta-analysis also supports the safety and efficacy of amlodipine for managing hypertension during pregnancy [26]. Amlodipine effectively lowers blood pressure and improves vascular function by inducing smooth muscle relaxation and dilating blood vessels. Given its efficacy in reducing blood pressure and supporting vascular health, amlodipine is recommended as an appropriate antihypertensive agent for individuals with moderate to severe hypertension [27].
This study is the first to evaluate the effects of amlodipine on uterine PI, RI, and successful ET in individuals with previous implantation failure. The findings indicate that amlodipine reduced blood flow indices in the uterine artery, although no significant differences were observed in chemical or CPRs between the treatment and control groups. As this is the first study to explore the potential benefits of amlodipine in FET, its findings cannot be compared with previous research. While several studies have investigated the use of additional pharmacological agents to improve ET outcomes, the results have been inconclusive [28]. Two adrenoceptor agonists, terbutaline and ritodrine, are recognized for their capacity to relax the uterus by reducing smooth muscle tension. However, a large randomized trial found no significant differences in IR or CPR between the intervention and control groups [29,30].
Limited research has compared FET and fresh blastocyst transfer regarding UtA-PI in IVF recipients [31-33]. Two studies suggested that patients undergoing frozen blastocyst transfer exhibit superior uterine perfusion and fetal growth relative to those experiencing fresh blastocyst transfer [34,35]. In a study by Choux et al. [33], the PI was significantly higher in fresh ET recipients compared to those who conceived naturally (p=0.001) and significantly lower in the FET group compared to the fresh ET group (p=0.001) [31]. Additionally, two other studies reported lower UtA-PI values in FET as opposed to fresh blastocyst transfer recipients, corroborating these findings [34,36].
Several methods can improve the transfer quality during the final phase of ART. These methods include soft ET, ultrasound guidance, and the use of specific pharmaceutical agents [36]. Various pharmacological strategies are available to improve IVF outcomes, such as the administration of growth hormone-releasing factor or growth hormone, letrozole, transdermal testosterone, oral L-arginine, and nifedipine [29], as well as the application of platelet-rich plasma [37] and granulocyte-macrophage colony-stimulating factor [38]. A wide range of factors associated with different aspects of assisted reproduction can influence the pregnancy rate in IVF-ET cycles [39]. One such factor may be blood flow parameters in the uterus, which can affect implantation success. Inadequate blood flow, as indicated by uterine artery color Doppler measurements, may lead to implantation failure in patients with unsuccessful IVF outcomes. Adequate vascular supply is essential for various IVF-related processes, including endometrial growth and embryo implantation [40].
Studies have demonstrated that the PI and RI of the uterine artery were significantly lower in women with successful IVF-ET cycles compared to those with unsuccessful cycles [41,42]. Our study yielded similar results [41-43]. Ng et al. [28] found that administering a single 20 mg dose of nifedipine 30 minutes before ET did not improve the IR or CPR in infertility treatment. Similarly, our study did not demonstrate a statistically significant difference in these outcomes.
The present results indicate decreases in the PI and RI of the uterine artery after approximately 14 to 20 days of amlodipine administration. This conclusion is supported by consistent pre- and post-intervention measurements. Participants were carefully selected to minimize variability, excluding those with conditions or medications that could influence PI or RI. To increase reliability, the study was performed in a controlled clinical setting to minimize external variables. Before initiating amlodipine administration, baseline values for PI and RI were established for each participant. A control group not receiving amlodipine was included to compare natural variations in PI and RI with those observed among medication recipients. The same protocols for measurement timing and data handling were applied to all participants, ensuring consistency and reliability. Participants were also monitored regarding additional factors that could affect PI and RI, such as diet, activity levels, and other medications, and these were controlled or accounted for in the analysis. Within its pharmacological class, amlodipine is notable for demonstrating prolonged efficacy by inhibiting voltage-dependent L-type calcium channels, thereby interrupting initial calcium influx. With a half-life of 30 to 50 hours, amlodipine enables convenient once-daily dosing, in contrast to shorter-acting alternatives like nifedipine [22]. Due to its long half-life, amlodipine is typically taken once daily, with effects lasting for 24 hours or more. Steady-state plasma levels are achieved after 7 to 8 days of daily dosing [44]. Therefore, given the mechanism of action and the timing of administration, we anticipated that the reduction in PI and RI of the uterine artery should be observable within 14 to 20 days of amlodipine use, a sufficient period to observe the drug’s stable effects. The objectives of the present study were to assess the impact of amlodipine on the PI and RI of the uterine artery and to improve ET outcomes by increasing circulation through blood vessel relaxation. Our findings demonstrate potential as a therapeutic approach in women experiencing implantation failure, meriting additional research. By providing a clear path, this study lays the groundwork for subsequent exploration of amlodipine in ET treatments. Increasing the sample size and potentially examining alternative medication protocols may be beneficial.
In conclusion, this study is the first to examine the use of amlodipine to correct uterine artery resistance in women with prior implantation failure, with the aim of guiding future research. However, our findings suggest that administering amlodipine before FET may not significantly improve clinical pregnancy outcomes for women with a history of FET cycle implantation failure. Further research is needed to explore the clinical potential of amlodipine, and clinical trials with larger sample sizes should be conducted.

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Acknowledgments

We thank all the patients and staff from all the units that participated in the study.

Author contributions

Conceptualization: NN, SS. Methodology: NN, LF, SS. Formal analysis: KH, HGB. Data curation: NN, AG. Funding acquisition: NN. Project administration: SS. Visualization: EE, LF. Software: PH. Validation: NN, SS. Investigation: SS. Writing-original draft: SS. Writing-review & editing: HGB, SS. Approval of final manuscript: NN, EE, LF, AG, KH, PH, HGB, SS.

Figure 1.
Ultrasound two-dimensional of the endometrial zone from 1 to 4.
cerm-2024-07129f1.jpg
Figure 2.
Study Consolidated Standards for Reporting Trials (CONSORT) flowchart.
cerm-2024-07129f2.jpg
Figure 3.
Uterine artery Doppler waveform (A, B) before treatment in amlodipine group and (C, D) after treatment in amlodipine group. RT, right artery; PS, peak systolic velocity; ED, end diastolic velocity; TAMAX, time-averaged maximum velocity; TAMEAN, time-averaged mean velocity; PI, pulsatility index; RI, resistance index; S/D, systolic/diastolic ratio; HR, heart rate; LT, left artery.
cerm-2024-07129f3.jpg
Table 1.
Patient characteristics and demographic data
Characteristic Amlodipine group (n=50) Control group (n=50) p-value
Age (yr) 34.0±3.5 35.4±4.6 0.626
BMI (kg/m2) 25.2±5.3 24.8±2.8 0.824
Infertility 0.240
 Primary infertility, 21 (42) 22 (44)
 Secondary infertility 29 (58) 28 (56)
Duration of subfertility (yr)a) 5.4±3.3 5.8±3.0 0.219
Endometrial thickness (mm) 8.36±0.27 8.6±0.27 0.540
No. of grade A embryos transferred in the cycle 2.96±0.14 2.68±0.12 0.150
No. of previous failed embryo transfer cycles 2.1±0.05 2.4±0.42 0.726

Values are presented as mean±standard deviation or number (%).

BMI, body mass index.

Table 2.
Sonographic endometrial pattern and endometrial blood flow zones in amlodipine and placebo groups
Characteristic Subgroup Amlodipine group (n=50) Control group (n=50) p-value
Sonographic endometrial pattern No layering 28 (56) 22 (44) 0.007
Distinct 3-line appearance 5 (10) 18 (36)
Hazy 3-line appearance 17 (34) 10 (20)
Endometrial blood flow zone Zone I 0 1 (2) <0.001
Zone II 6 (12) 28 (56)
Zone III 43 (86) 21 (42)
Zone IV 1 (2) 0

Values are presented as frequency (%).

Table 3.
Uterine artery PI and RI in amlodipine and placebo groups
Characteristic Amlodipine group (n=50) Control group (n=50) p-value
Uterine artery PI 2.54±0.52 3.5±0.89 0.0001
Uterine artery RI 0.83±0.05 0.91±0.053 0.0001

Values are presented as mean±standard deviation.

PI, pulsatility index; RI, resistance index.

Table 4.
Comparison of pregnancy outcomes
Characteristic Subgroup Amlodipine group (n=50) Control group (n=50) p-value
Chemical pregnancy No 37 (74) 41 (82) 0.472
Yes 13 (26) 9 (18)
Clinical pregnancy No 39 (78) 44 (88) 0.287
Yes 11 (22) 6 (12)

Values are presented as frequency (%).

References

1. Kushnir VA, Smith GD, Adashi EY. The future of IVF: the new normal in human reproduction. Reprod Sci 2022;29:849-56.
crossref pmid pmc pdf
2. Sehring J, Beltsos A, Jeelani R. Human implantation: the complex interplay between endometrial receptivity, inflammation, and the microbiome. Placenta 2022;117:179-86.
crossref pmid
3. Bashiri A, Halper KI, Orvieto R. Recurrent implantation failure-update overview on etiology, diagnosis, treatment and future directions. Reprod Biol Endocrinol 2018;16:121.
crossref pmid pmc pdf
4. Li Z, Wang X, Guan Y, Yu X, Liu J, Zhang Z. Uterine artery blood flow and microvessel density by vaginal color Doppler ultrasonography in embryo implantation failure. Exp Ther Med 2017;14:4797-800.
crossref pmid pmc
5. Ali zarad C, Mohamed MH, Shanab WS. Role of uterine artery Doppler in assessment of unexplained infertility. Egypt J Radiol Nucl Med 2021;52:59.

6. Adibi A, Khadem M, Mardanian F, Hovsepian S. Uterine and arcuate arteries blood flow for predicting of ongoing pregnancy in in vitro fertilization. J Res Med Sci 2015;20:879-84.
crossref pmid pmc
7. Gutarra-Vilchez RB, Bonfill Cosp X, Glujovsky D, Viteri-Garcia A, Runzer-Colmenares FM, Martinez-Zapata MJ. Vasodilators for women undergoing fertility treatment. Cochrane Database Syst Rev 2018;10:CD010001.
crossref pmid pmc
8. Bulsara KG, Patel P, Cassagnol M. Amlodipine. In: StatPearls [Internet]. StatPearls Publishing; 2024 [cited 2024 Aug 13]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK519508

9. Fang Q, Tian M, Wang F, Zhang Z, Du T, Wang W, et al. Amlodipine induces vasodilation via Akt2/Sp1-activated miR-21 in smooth muscle cells. Br J Pharmacol 2019;176:2306-20.
crossref pmid pmc pdf
10. Sohrabvand F, Haghollahi F, Maasomi M, Asgarpoor L, Shariat M, Hamedani M. The effect of administrating indomethacin or hyoscine before embryo transfer on ART outcome (a pilot study). Iran J Reprod Med 2009;7:169-73.

11. ALPHA Scientists in Reproductive Medicine; ESHRE Special Interest Group Embryology. Istanbul consensus workshop on embryo assessment: proceedings of an expert meeting. Reprod Biomed Online 2011;22:632-46.
crossref pmid
12. Gardner DK, Vella P, Lane M, Wagley L, Schlenker T, Schoolcraft WB. Culture and transfer of human blastocysts increases implantation rates and reduces the need for multiple embryo transfers. Fertil Steril 1998;69:84-8.
crossref pmid
13. Silva Martins R, Helio Oliani A, Vaz Oliani D, Martinez de Oliveira J. Subendometrial resistence and pulsatility index assessment of endometrial receptivity in assisted reproductive technology cycles. Reprod Biol Endocrinol 2019;17:62.
crossref pmid pmc
14. Lawrenz B, Coughlan C, Melado L, Fatemi HM. The ART of frozen embryo transfer: back to nature! Gynecol Endocrinol 2020;36:479-83.
crossref pmid
15. Coulam CB, Stern JJ, Soenksen DM, Britten S, Bustillo M. Comparison of pulsatility indices on the day of oocyte retrieval and embryo transfer. Hum Reprod 1995;10:82-4.
crossref pmid
16. Tsai YC, Chang JC, Tai MJ, Kung FT, Yang LC, Chang SY. Relationship of uterine perfusion to outcome of intrauterine insemination. J Ultrasound Med 1996;15:633-6.
crossref pmid pdf
17. Riad ON, Hak AA. Assessment of endometrial receptivity using Doppler ultrasonography in infertile women undergoing intrauterine insemination. Gynecol Endocrinol 2014;30:70-3.
crossref pmid
18. Tian Y, Yang X. A review of roles of uterine artery Doppler in pregnancy complications. Front Med (Lausanne) 2022;9:813343.
crossref pmid pmc
19. Lloyd-Davies C, Collins SL, Burton GJ. Understanding the uterine artery Doppler waveform and its relationship to spiral artery remodelling. Placenta 2021;105:78-84.
crossref pmid
20. Tong R, Zhou Y, He Q, Zhuang Y, Zhou W, Xia F. Analysis of the guidance value of 3D ultrasound in evaluating endometrial receptivity for frozen-thawed embryo transfer in patients with repeated implantation failure. Ann Transl Med 2020;8:944.
crossref pmid pmc
21. Takasaki A, Tamura H, Miwa I, Taketani T, Shimamura K, Sugino N. Endometrial growth and uterine blood flow: a pilot study for improving endometrial thickness in the patients with a thin endometrium. Fertil Steril 2010;93:1851-8.
crossref pmid
22. van Zwieten PA. Amlodipine: an overview of its pharmacodynamic and pharmacokinetic properties. Clin Cardiol 1994;17(9 Suppl 3): III3-6.
pmid
23. Mito A, Murashima A, Wada Y, Miyasato-Isoda M, Kamiya CA, Waguri M, et al. Safety of amlodipine in early pregnancy. J Am Heart Assoc 2019;8:e012093.
crossref pmid pmc
24. Malha L, August P. Safety of antihypertensive medications in pregnancy: living with uncertainty. J Am Heart Assoc 2019;8:e013495.
crossref pmid pmc
25. American College of Obstetricians and Gynecologists’ Committee on Practice Bulletins—Obstetrics. ACOG Practice Bulletin No. 203: chronic hypertension in pregnancy. Obstet Gynecol 2019;133:e26-50.
crossref pmid
26. Yin J, Mei Z, Shi S, Du P, Qin S. Nifedipine or amlodipine?: the choice for hypertension during pregnancy: a systematic review and meta-analysis. Arch Gynecol Obstet 2022;306:1891-900.
crossref pmid pdf
27. Fares H, DiNicolantonio JJ, O’Keefe JH, Lavie CJ. Amlodipine in hypertension: a first-line agent with efficacy for improving blood pressure and patient outcomes. Open Heart 2016;3:e000473.
crossref pmid pmc
28. Ng KK, Rozen G, Stewart T, Agresta F, Polyakov A. Does nifedipine improve outcomes of embryo transfer?: interim analysis of a randomized, double blinded, placebo-controlled trial. Medicine (Baltimore) 2019;98:e14251.
crossref pmid pmc
29. Pinheiro OL, Cavagna M, Baruffi RL, Mauri AL, Petersen C, Franco JG Jr. Administration of beta2-adrenergic agonists during the peri-implantation period does not improve implantation or pregnancy rates in intracytoplasmic sperm injection (ICSI) cycles. J Assist Reprod Genet 2003;20:513-6.
crossref pmid pmc
30. Carbone IF, Cruz JJ, Sarquis R, Akolekar R, Nicolaides KH. Assisted conception and placental perfusion assessed by uterine artery Doppler at 11-13 weeks’ gestation. Hum Reprod 2011;26:1659-64.
crossref pmid
31. Prefumo F, Fratelli N, Soares SC, Thilaganathan B. Uterine artery Doppler velocimetry at 11-14 weeks in singleton pregnancies conceived by assisted reproductive technology. Ultrasound Obstet Gynecol 2007;29:141-5.
crossref pmid
32. Rizzo G, Aiello E, Pietrolucci ME, Arduini D. Are there differences in placental volume and uterine artery Doppler in pregnancies resulting from the transfer of fresh versus frozen-thawed embryos through in vitro fertilization. Reprod Sci 2016;23:1381-6.
crossref pmid pdf
33. Choux C, Ginod P, Barberet J, Rousseau T, Bruno C, Sagot P, et al. Placental volume and other first-trimester outcomes: are there differences between fresh embryo transfer, frozen-thawed embryo transfer and natural conception? Reprod Biomed Online 2019;38:538-48.
crossref pmid
34. Gundogan F, Bianchi DW, Scherjon SA, Roberts DJ. Placental pathology in egg donor pregnancies. Fertil Steril 2010;93:397-404.
crossref pmid
35. van Duijn L, Rousian M, Reijnders IF, Willemsen SP, Baart EB, Laven JS, et al. The influence of frozen-thawed and fresh embryo transfer on utero-placental (vascular) development: the Rotterdam Periconception cohort. Hum Reprod 2021;36:2091-100.
crossref pmid pmc pdf
36. Saravelos SH, Li TC. Embryo transfer techniques. Best Pract Res Clin Obstet Gynaecol 2019;59:77-88.
crossref pmid
37. Coksuer H, Akdemir Y, Ulas Barut M. Improved in vitro fertilization success and pregnancy outcome with autologous platelet-rich plasma treatment in unexplained infertility patients that had repeated implantation failure history. Gynecol Endocrinol 2019;35:815-8.
crossref pmid
38. Armstrong S, MacKenzie J, Woodward B, Pacey A, Farquhar C. GM-CSF (granulocyte macrophage colony-stimulating factor) supplementation in culture media for women undergoing assisted reproduction. Cochrane Database Syst Rev 2020;7:CD013497.
crossref pmid pmc
39. Prasad S, Goyal R, Kumar Y, Nayar P, Hajela S, Kumaran A, et al. The relationship between uterine artery two-dimensional color Doppler measurement and pregnancy outcome: a prospective observational study. J Reprod Infertil 2017;18:251-6.
pmid pmc
40. Steer CV, Tan SL, Dillon D, Mason BA, Campbell S. Vaginal color Doppler assessment of uterine artery impedance correlates with immunohistochemical markers of endometrial receptivity required for the implantation of an embryo. Fertil Steril 1995;63:101-8.
crossref pmid
41. Adakan S, Yoldemir T, Tavmergen E, Goker E, Killi R. Predictivity of uterine artery, arcuate artery, and intraovarian artery Doppler indices measured on the day of human chorionic gonadotropin injection on pregnancy outcomes. Fertil Steril 2005;84:529-32.
crossref pmid
42. Hoozemans DA, Schats R, Lambalk NB, Homburg R, Hompes PG. Serial uterine artery Doppler velocity parameters and human uterine receptivity in IVF/ICSI cycles. Ultrasound Obstet Gynecol 2008;31:432-8.
crossref pmid
43. Ivanovski M, Damcevski N, Radevska B, Doicev G. Assessment of uterine artery and arcuate artery blood flow by transvaginal color Doppler ultrasound on the day of human chorionic gonadotropin administration as predictors of pregnancy in an in vitro fertilization program. Akush Ginekol (Sofiia) 2012;51:55-60.

44. Kato M, Shitara Y, Sato H, Yoshisue K, Hirano M, Ikeda T, et al. The quantitative prediction of CYP-mediated drug interaction by physiologically based pharmacokinetic modeling. Pharm Res 2008;25:1891-901.
crossref pmid pdf


ABOUT
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
AUTHOR INFORMATION
Editorial Office
Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital
82 Gumi-ro 173, Bundang-gu, Seongnam 13620, Korea
Tel: +82-31-787-7254    CP: +82-10-9072-3154    E-mail: blasto@snubh.org                

Copyright © 2025 by Korean Society for Reproductive Medicine.

Developed in M2PI

Close layer
prev next