1
|
Harvey AJ, Willson BE, Surrey ES, Gardner DK. Ovarian stimulation protocols: impact on oocyte and endometrial quality and function. Fertil Steril 2024:S0015-0282(24)01973-3. [PMID: 39197516 DOI: 10.1016/j.fertnstert.2024.08.340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Ovarian stimulation (OS) truly is an art. There exists a myriad of protocols used to achieve the same goal: stimulating the ovaries to produce more than one mature oocyte to improve the chance of a live birth. However, considerable debate remains as to whether OS impacts oocyte and endometrial quality to affect in vitro fertilization outcomes. Although "more is better" has long been considered the best approach for oocyte retrieval, this review challenges that notion by examining the influence of stimulation on oocyte quality. Likewise, improved outcomes after frozen blastocyst transfer suggest that OS perturbs endometrial preparation and/or receptivity, although correlating changes with implantation success remains a challenge. Therefore, the focus of this review is to summarize our current understanding of perturbations in human oocyte quality and endometrial function induced by exogenous hormone administration. We highlight the need for further research to identify more appropriate markers of oocyte developmental competence as well as those that define the roles of the endometrium in the success of assisted reproductive technology.
Collapse
Affiliation(s)
- Alexandra J Harvey
- Melbourne IVF, East Melbourne, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Bryn E Willson
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai, Los Angeles, California
| | - Eric S Surrey
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado
| | - David K Gardner
- Melbourne IVF, East Melbourne, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
2
|
Bui BN, Kukushkina V, Meltsov A, Olsen C, van Hoogenhuijze N, Altmäe S, Mol F, Teklenburg G, de Bruin J, Besselink D, Stevens Brentjens L, Obukhova D, Zamani Esteki M, van Golde R, Romano A, Laisk T, Steba G, Mackens S, Salumets A, Broekmans F. The endometrial transcriptome of infertile women with and without implantation failure. Acta Obstet Gynecol Scand 2024; 103:1348-1365. [PMID: 38520066 PMCID: PMC11168281 DOI: 10.1111/aogs.14822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 01/26/2024] [Accepted: 02/16/2024] [Indexed: 03/25/2024]
Abstract
INTRODUCTION Implantation failure after transferring morphologically "good-quality" embryos in in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) may be explained by impaired endometrial receptivity. Analyzing the endometrial transcriptome analysis may reveal the underlying processes and could help in guiding prognosis and using targeted interventions for infertility. This exploratory study investigated whether the endometrial transcriptome profile was associated with short-term or long-term implantation outcomes (ie success or failure). MATERIAL AND METHODS Mid-luteal phase endometrial biopsies of 107 infertile women with one full failed IVF/ICSI cycle, obtained within an endometrial scratching trial, were subjected to RNA-sequencing and differentially expressed genes analysis with covariate adjustment (age, body mass index, luteinizing hormone [LH]-day). Endometrial transcriptomes were compared between implantation failure and success groups in the short term (after the second fresh IVF/ICSI cycle) and long term (including all fresh and frozen cycles within 12 months). The short-term analysis included 85/107 women (33 ongoing pregnancy vs 52 no pregnancy), excluding 22/107 women. The long-term analysis included 46/107 women (23 'fertile' group, ie infertile women with a live birth after ≤3 embryos transferred vs 23 recurrent implantation failure group, ie no live birth after ≥3 good quality embryos transferred), excluding 61/107 women not fitting these categories. As both analyses drew from the same pool of 107 samples, there was some sample overlap. Additionally, cell type enrichment scores and endometrial receptivity were analyzed, and an endometrial development pseudo-timeline was constructed to estimate transcriptomic deviations from the optimum receptivity day (LH + 7), denoted as ΔWOI (window of implantation). RESULTS There were no significantly differentially expressed genes between implantation failure and success groups in either the short-term or long-term analyses. Principal component analysis initially showed two clusters in the long-term analysis, unrelated to clinical phenotype and no longer distinct following covariate adjustment. Cell type enrichment scores did not differ significantly between groups in both analyses. However, endometrial receptivity analysis demonstrated a potentially significant displacement of the WOI in the non-pregnant group compared with the ongoing pregnant group in the short-term analysis. CONCLUSIONS No distinct endometrial transcriptome profile was associated with either implantation failure or success in infertile women. However, there may be differences in the extent to which the WOI is displaced.
Collapse
Affiliation(s)
- Bich Ngoc Bui
- Department of Gynecology and Reproductive MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Alvin Meltsov
- Competence Center on Health TechnologiesTartuEstonia
- Department of Obstetrics and Gynecology, GROW, School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Catharina Olsen
- Center for Medical Genetics, Research Group Reproduction and GeneticsVrije Universiteit BrusselBrusselsBelgium
- Brussels Interuniversity Genomics High Throughput Core (BRIGHTcore)VUB‐ULBBrusselsBelgium
- Interuniversity Institute of Bioinformatics in Brussels (IB)BrusselsBelgium
| | - Nienke van Hoogenhuijze
- Department of Gynecology and Reproductive MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of SciencesUniversity of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria, ibs.GRANADAGranadaSpain
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC)Karolinska Institute and Karolinska University HospitalStockholmSweden
| | - Femke Mol
- Center for Reproductive Medicine, Reproduction and Development, Amsterdam University Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Jan‐Peter de Bruin
- Department of Obstetrics and GynecologyJeroen Bosch Hospital‘s‐HertogenboschThe Netherlands
| | - Dagmar Besselink
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Linda Stevens Brentjens
- Department of Obstetrics and Gynecology, GROW, School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Darina Obukhova
- Department of Clinical GeneticsMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Genetics and Cell Biology, GROW School for Oncology and ReproductionMaastricht UniversityMaastrichtThe Netherlands
| | - Masoud Zamani Esteki
- Department of Clinical GeneticsMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Genetics and Cell Biology, GROW School for Oncology and ReproductionMaastricht UniversityMaastrichtThe Netherlands
| | - Ron van Golde
- Department of Obstetrics and Gynecology, GROW, School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Andrea Romano
- Department of Obstetrics and Gynecology, GROW, School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Triin Laisk
- Estonian Genome Center, Institute of GenomicsUniversity of TartuTartuEstonia
| | - Gaby Steba
- Department of Gynecology and Reproductive MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Shari Mackens
- Brussels IVFUniversitair Ziekenhuis Brussel, Vrije Universiteit BrusselBrusselsBelgium
| | - Andres Salumets
- Competence Center on Health TechnologiesTartuEstonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC)Karolinska Institute and Karolinska University HospitalStockholmSweden
- Department of Obstetrics and Gynecology, Institute of Clinical MedicineUniversity of TartuTartuEstonia
| | - Frank Broekmans
- Department of Gynecology and Reproductive MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
- Center for Infertility Care, Dijklander HospitalPurmerendThe Netherlands
| |
Collapse
|
3
|
Chemerinski A, Shen M, Valero-Pacheco N, Zhao Q, Murphy T, George L, Lemenze A, Sherman L, Heller D, Chen X, Wu T, McGovern PG, Morelli SS, Arora R, Beaulieu AM, Douglas NC. The impact of ovarian stimulation on the human endometrial microenvironment. Hum Reprod 2024; 39:1023-1041. [PMID: 38511208 PMCID: PMC11063567 DOI: 10.1093/humrep/deae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
STUDY QUESTION How does ovarian stimulation (OS), which is used to mature multiple oocytes for ART procedures, impact the principal cellular compartments and transcriptome of the human endometrium in the periovulatory and mid-secretory phases? SUMMARY ANSWER During the mid-secretory window of implantation, OS alters the abundance of endometrial immune cells, whereas during the periovulatory period, OS substantially changes the endometrial transcriptome and impacts both endometrial glandular and immune cells. WHAT IS KNOWN ALREADY Pregnancies conceived in an OS cycle are at risk of complications reflective of abnormal placentation and placental function. OS can alter endometrial gene expression and immune cell populations. How OS impacts the glandular, stromal, immune, and vascular compartments of the endometrium, in the periovulatory period as compared to the window of implantation, is unknown. STUDY DESIGN, SIZE, DURATION This prospective cohort study carried out between 2020 and 2022 included 25 subjects undergoing OS and 25 subjects in natural menstrual cycles. Endometrial biopsies were performed in the proliferative, periovulatory, and mid-secretory phases. PARTICIPANTS/MATERIALS, SETTING, METHODS Blood samples were processed to determine serum estradiol and progesterone levels. Both the endometrial transcriptome and the principal cellular compartments of the endometrium, including glands, stroma, immune, and vasculature, were evaluated by examining endometrial dating, differential gene expression, protein expression, cell populations, and the three-dimensional structure in endometrial tissue. Mann-Whitney U tests, unpaired t-tests or one-way ANOVA and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE In the periovulatory period, OS induced high levels of differential gene expression, glandular-stromal dyssynchrony, and an increase in both glandular epithelial volume and the frequency of endometrial monocytes/macrophages. In the window of implantation during the mid-secretory phase, OS induced changes in endometrial immune cells, with a greater frequency of B cells and a lower frequency of CD4 effector T cells. LARGE SCALE DATA The data underlying this article have been uploaded to the Genome Expression Omnibus/National Center for Biotechnology Information with accession number GSE220044. LIMITATIONS, REASONS FOR CAUTION A limited number of subjects were included in this study, although the subjects within each group, natural cycle or OS, were homogenous in their clinical characteristics. The number of subjects utilized was sufficient to identify significant differences; however, with a larger number of subjects and additional power, we may detect additional differences. Another limitation of the study is that proliferative phase biopsies were collected in natural cycles, but not in OS cycles. Given that the OS cycle subjects did not have known endometrial factor infertility, and the comparisons involved subjects who had a similar and robust response to stimulation, the findings are generalizable to women with a normal response to OS. WIDER IMPLICATIONS OF THE FINDINGS OS substantially altered the periovulatory phase endometrium, with fewer transcriptomic and cell type-specific changes in the mid-secretory phase. Our findings show that after OS, the endometrial microenvironment in the window of implantation possesses many more similarities to that of a natural cycle than does the periovulatory endometrium. Further investigation of the immune compartment and the functional significance of this cellular compartment under OS conditions is warranted. STUDY FUNDING/COMPETING INTERESTS Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases (R01AI148695 to A.M.B. and N.C.D.), Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01HD109152 to R.A.), and the March of Dimes (5-FY20-209 to R.A.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or March of Dimes. All authors declare no conflict of interest.
Collapse
Affiliation(s)
- Anat Chemerinski
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - May Shen
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
| | - Nuriban Valero-Pacheco
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Trystn Murphy
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Lea George
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Alex Lemenze
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Molecular and Genomics Informatics Core, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Lauren Sherman
- Department of Medicine-Hematology/Oncology, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Debra Heller
- Department of Pathology, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Xiaowei Chen
- Department of Pathology, Columbia University Medical Center, New York, NY, USA
| | - Tracy Wu
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Peter G McGovern
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Sara S Morelli
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Ripla Arora
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
| | - Aimee M Beaulieu
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Nataki C Douglas
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| |
Collapse
|
4
|
Choo SP, Lee I, Lee JH, Lee D, Park H, Park JH, Cho S, Choi YS. Transcriptomic patterns in early-secretory and mid-secretory endometrium in a natural menstrual cycle immediately before in vitro fertilization and embryo transfer. Obstet Gynecol Sci 2023; 66:417-429. [PMID: 37460099 PMCID: PMC10514596 DOI: 10.5468/ogs.22315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 05/06/2023] [Accepted: 06/04/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the endometrial transcriptomic patterns in the early secretory phase (ESP) and mid-secretory phase (MSP) of the natural menstrual cycle before in vitro fertilization and embryo transfer (IVF-ET). METHODS Thirty patients whose endometrial tissues were obtained from the ESP or MSP of a natural menstrual cycle immediately before IVF-ET were included. Endometrial dating was histologically confirmed as ESP (cycle days 16-18) or MSP (cycle days 19-21), according to the noyes criteria. The patients were divided into two groups depending on the IVF-ET outcome: pregnant (n=14; 7 in ESP and 7 in MSP) or non-pregnant (n=16; 8 in ESP and 8 in MSP). Differentially expressed genes (DEGs) in the MSP, compared to the ESP, were identified using NanoString nCounter (NanoString Technologies, Seattle, WA, USA) data for both the pregnant and non-pregnant groups. RESULTS Thirteen DEGs in the pregnant group and 11 DEGs in the non-pregnant group were identified in the MSP compared to those in the ESP. In both groups, adrenoceptor alpha 2A, interleukin 1 receptor-associated kinase 2, a disintegrin and metalloproteinase with thrombospondin repeats 15 (ADAMTS15), serpin family E member 1, integrin subunit beta 3, transmembrane protein 252 (TMEM252), huntingtin associated protein 1, C2 calcium-dependent domain containing 4A, and integrin subunit alpha 2 were upregulated in the MSP, compared to the ESP. TMEM37, galactosidase beta 1 like 2, Rho family GTPase 3, and cytochrome P450 family 24 subfamily A member 1 were upregulated in the MSP only in the pregnant group. ADAMTS8 was downregulated and monoamine oxidase A was upregulated in the MSP only in the non-pregnant group. CONCLUSION Transcriptomic patterns in the endometrium immediately before IVF-ET appear to differ according to the IVF-ET outcome. These novel DEGs, which have not been previously studied, may have functional significance during the window of implantation and serve as potential biomarkers of endometrial receptivity.
Collapse
Affiliation(s)
- Sung Pil Choo
- Department of Obstetrics and Gynecology, Inha University Hospital, College of Medicine, Inha University, Incheon,
Korea
| | - Inha Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul,
Korea
| | - Jae-Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul,
Korea
| | - Dowon Lee
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
| | - Hyemin Park
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
| | - Joo Hyun Park
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul,
Korea
- Department of Obstetrics and Gynecology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin,
Korea
| | - SiHyun Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul,
Korea
| | - Young Sik Choi
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul,
Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
| |
Collapse
|
5
|
Meltsov A, Saare M, Teder H, Paluoja P, Arffman RK, Piltonen T, Laudanski P, Wielgoś M, Gianaroli L, Koel M, Peters M, Salumets A, Krjutškov K, Palta P. Targeted gene expression profiling for accurate endometrial receptivity testing. Sci Rep 2023; 13:13959. [PMID: 37633957 PMCID: PMC10460380 DOI: 10.1038/s41598-023-40991-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/20/2023] [Indexed: 08/28/2023] Open
Abstract
Expressional profiling of the endometrium enables the personalised timing of the window of implantation (WOI). This study presents and evaluates a novel analytical pipeline based on a TAC-seq (Targeted Allele Counting by sequencing) method for endometrial dating. The expressional profiles were clustered, and differential expression analysis was performed on the model development group, using 63 endometrial biopsies spanning over proliferative (PE, n = 18), early-secretory (ESE, n = 18), mid-secretory (MSE, n = 17) and late-secretory (LSE, n = 10) endometrial phases of the natural cycle. A quantitative predictor model was trained on the development group and validated on sequenced samples from healthy women, consisting of 52 paired samples taken from ESE and MSE phases and five LSE phase samples from 31 individuals. Finally, the developed test was applied to 44 MSE phase samples from a study group of patients diagnosed with recurrent implantation failure (RIF). In validation samples (n = 57), we detected displaced WOI in 1.8% of the samples from fertile women. In the RIF study group, we detected a significantly higher proportion of the samples with shifted WOI than in the validation set of samples from fertile women, 15.9% and 1.8% (p = 0.012), respectively. The developed model was evaluated with an average cross-validation accuracy of 98.8% and an accuracy of 98.2% in the validation group. The developed beREADY screening model enables sensitive and dynamic detection of selected transcriptome biomarkers, providing a quantitative and accurate prediction of endometrial receptivity status.
Collapse
Affiliation(s)
- Alvin Meltsov
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Merli Saare
- Competence Centre On Health Technologies, 50411, Tartu, Estonia.
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.
| | - Hindrek Teder
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Priit Paluoja
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, FI-90014, Oulu, Finland
| | - Terhi Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, FI-90014, Oulu, Finland
| | - Piotr Laudanski
- Oviklinika Infertility Center, 01-377, Warsaw, Poland
- Women's Health Research Institute, Calisia University, 62-800, Kalisz, Poland
- Department of Obstetrics, Gynecology and Gynaecological Oncology, Medical University of Warsaw, 02-091, Warsaw, Poland
| | | | - Luca Gianaroli
- SISMeR, Reproductive Medicine Institute, 40138, Bologna, Italy
| | - Mariann Koel
- Institute of Genomics, University of Tartu, 51010, Tartu, Estonia
| | - Maire Peters
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
| | - Andres Salumets
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, SE-141 52, Stockholm, Sweden
| | - Kaarel Krjutškov
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
| | - Priit Palta
- Competence Centre On Health Technologies, 50411, Tartu, Estonia
- Institute of Genomics, University of Tartu, 51010, Tartu, Estonia
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
6
|
Meng Y, Tao L, Xia T, Zhu J, Lin X, Zhou W, Liu Y, Ou J, Xing W. Elevated estradiol levels on hCG trigger day adversely effects on the clinical pregnancy rates of blastocyst embryo transfer but not cleavage-stage embryo transfer in fresh cycles: a retrospective cohort study. PeerJ 2023; 11:e15709. [PMID: 37483963 PMCID: PMC10361074 DOI: 10.7717/peerj.15709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Background Elevated estradiol (E2) levels are an inevitable outcome of the controlled ovulation hyperstimulation. However, the effect of this change on pregnancy is still uncertain. Our study aimed to analyze the impact of increased serum E2 at the day of human chorionic gonadotropin (hCG) administration on the clinical outcomes of women with fresh embryo transfer (ET) cycles. Methods This study included 3,009 fresh ET cycles from October 2015 to September 2021. Based on the stage of embryos transferred, these cycles were categorized into the cleavage group and blastocyst group. Both groups were then divided into four sets according to E2 levels when hCG was administered: set 1 (E2 ≤ 2,000 pg/ml), set 2 (E2 = 2,001-3,000 pg/ml), set 3 (E2 = 3,001-4,000 pg/ml), and set 4 (E2 > 4,000 pg/ml). The primary outcome was the clinical pregnancy rate (CPR). Binary logistics regression analysis was established to explore the association between CPR and E2 levels. Specifically, the threshold effect of serum E2 on CPR was revealed using the two-piecewise linear regression analyses. Results The multivariate regression model in the cleavage group showed that patients' CPR in set 4 was 1.59 times higher than those in reference set 1, but the statistical difference was insignificant (P = 0.294). As for the blastocyst group, patients in set 4 had a lower CPR with adjusted ORs of 0.43 (P = 0.039) compared to patients in set 1. The inflection point for the blastocyst group was 39.7 pg/dl according to the results of the two-piecewise linear regression model. When E2 levels were over the point, the CPR decreased by 17% with every 1 pg/dl increases in serum E2 (adjusted OR = 0.83, 95% CI [0.72-0.96], P = 0.012). Conclusions Elevated E2 levels (>39.7 pg/dl) on hCG trigger day were associated with decreased CPR in patients with fresh blastocyst ET. However, it had no similar effect on the CPR of patients with fresh cleavage-stage ET.
Collapse
Affiliation(s)
- Yue Meng
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Linlin Tao
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Tingting Xia
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Jieru Zhu
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Xiaoqi Lin
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Wen Zhou
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Yuxia Liu
- Reproductive Medicine Center, the First People’s Hospital of Kashi Prefecture, Affiliated Kashi Hospital of Sun Yat-Sen University, Kashi, China
| | - Jianping Ou
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Weijie Xing
- Reproductive Medicine Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong province, China
| |
Collapse
|
7
|
Huang J, Lu Y, He Y, Wang Y, Zhu Q, Qi J, Ding Y, Zhao H, Ding Z, Sun Y. The effect of peak serum estradiol level during ovarian stimulation on cumulative live birth and obstetric outcomes in freeze-all cycles. Front Endocrinol (Lausanne) 2023; 14:1130211. [PMID: 37529616 PMCID: PMC10390295 DOI: 10.3389/fendo.2023.1130211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Objective To determine whether the peak serum estradiol (E2) level during ovarian stimulation affects the cumulative live birth rate (CLBR) and obstetric outcomes in freeze-all cycles. Methods This retrospective cohort study involved patients who underwent their first cycle of in vitro fertilization followed by a freeze-all strategy and frozen embryo transfer cycles between January 2014 and June 2019 at a tertiary care center. Patients were categorized into four groups according to quartiles of peak serum E2 levels during ovarian stimulation (Q1-Q4). The primary outcome was CLBR. Secondary outcomes included obstetric and neonatal outcomes of singleton and twin pregnancies. Poisson or logistic regression was applied to control for potential confounders for outcome measures, as appropriate. Generalized estimating equations were used to account for multiple cycles from the same patient for the outcome of CLBR. Results A total of 11237 patients were included in the analysis. Cumulatively, live births occurred in 8410 women (74.8%). The live birth rate (LBR) and CLBR improved as quartiles of peak E2 levels increased (49.7%, 52.1%, 54.9%, and 56.4% for LBR; 65.1%, 74.3%, 78.4%, and 81.6% for CLBR, from the lowest to the highest quartile of estradiol levels, respectively, P<0.001). Such association remained significant for CLBR after accounting for potential confounders in multivariable regression models, whereas the relationship between LBR and peak E2 levels did not reach statistical significance. In addition, no significant differences were noticed in adverse obstetric and neonatal outcomes (gestational diabetes mellitus, pregnancy-induced hypertension, preeclampsia, placental disorders, preterm birth, low birthweight, and small for gestational age) amongst E2 quartiles for either singleton or twin live births, both before and after adjustment. Conclusion In freeze-all cycles, higher peak serum E2 levels during ovarian stimulation were associated with increased CLBR, without increasing the risks of adverse obstetric and neonatal outcomes.
Collapse
Affiliation(s)
- Jiaan Huang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yao Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yaqiong He
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yuan Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jia Qi
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ying Ding
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hanting Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ziyin Ding
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
8
|
Pathare ADS, Saare M, Meltsov A, Lawarde A, Modhukur V, Kalinina A, Sekavin A, Kukushkina V, Karro H, Salumets A, Peters M. The cervical transcriptome changes during the menstrual cycle but does not predict the window of implantation. FRONTIERS IN REPRODUCTIVE HEALTH 2023; 5:1224919. [PMID: 37519341 PMCID: PMC10375708 DOI: 10.3389/frph.2023.1224919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/04/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction The expression of genes in female reproductive organs is influenced by the cyclic changes in hormone levels during the menstrual cycle. While the molecular changes in the endometrium that facilitate embryo implantation have been extensively studied, there is limited knowledge about the impact of the menstrual cycle on cervical cells. Cervical cells can be easily and routinely collected using a cytobrush during gynecological examination, offering a standardized approach for diagnostic testing. In this study we investigated how the transcriptome of cervical cells changes during the menstrual cycle and assessed the utility of these cells to determine endometrial receptivity. Methods Endocervical cells were collected with cytobrushes from 16 healthy women at different menstrual cycle phases in natural cycles and from four women undergoing hormonal replacement cycles. RNA sequencing was applied to gain insight into the transcriptome of cervical cells. Results Transcriptome analysis identified four differentially expressed genes (DEGs) between early- and mid-secretory samples, suggesting that the transcriptome of cervical cells does not change significantly during the opening of the implantation window. The most differences appeared during the transition to the late secretory phase (2136 DEGs) before the onset of menstruation. Cervical cells collected during hormonal replacement cycles showed 1899 DEGs enriched in immune system processes. Conclusions The results of our study suggested that cervical cells undergo moderate transcriptomic changes throughout the menstrual cycle; however, these changes do not reflect the gene expression pattern of endometrial tissue and offer little or no potential for endometrial receptivity diagnostics.
Collapse
Affiliation(s)
- Amruta D. S. Pathare
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Alvin Meltsov
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Ankita Lawarde
- Competence Centre on Health Technologies, Tartu, Estonia
- Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | | | - Aire Sekavin
- Women’s Clinic, Tartu University Hospital, Tartu, Estonia
| | | | - Helle Karro
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Women’s Clinic, Tartu University Hospital, Tartu, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Institute of Genomics, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Maire Peters
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| |
Collapse
|
9
|
Bajpai K, Acharya N, Prasad R, Wanjari MB. Endometrial Receptivity During the Preimplantation Period: A Narrative Review. Cureus 2023; 15:e37753. [PMID: 37214054 PMCID: PMC10198587 DOI: 10.7759/cureus.37753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
Endometrial receptivity is a complex and critical process fundamental to achieving a successful pregnancy. While researchers have made significant strides in understanding the underlying mechanisms governing endometrial receptivity, effective diagnostic and therapeutic strategies remain scarce. This review article aims to elucidate the various factors that contribute to endometrial receptivity, including the hormonal regulation and molecular mechanisms that govern this process, as well as potential biomarkers for assessing endometrial receptivity. One of the major challenges in identifying reliable biomarkers for endometrial receptivity is the intricate nature of the process itself. Nonetheless, recent advances in transcriptomic and proteomic technologies have identified several candidate biomarkers that could potentially enhance our ability to predict endometrial receptivity. Furthermore, emerging technologies such as single-cell RNA sequencing and mass spectrometry-based proteomics hold great promise for providing novel insights into the molecular mechanisms underlying endometrial receptivity. Despite the lack of reliable biomarkers, various therapeutic strategies have been proposed to improve endometrial receptivity. One promising approach involves the transplantation of mesenchymal stem cells (MSCs), which have been shown to increase endometrial thickness and receptivity in both animal models and clinical trials. Growth factors, cytokines, and exosomes derived from MSCs and other cell types may also have therapeutic potential for addressing endometrial dysfunction.
Collapse
Affiliation(s)
- Kshitij Bajpai
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Neema Acharya
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roshan Prasad
- Medicine and Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Mayur B Wanjari
- Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
10
|
Zhang WY, Gardner RM, Johal JK, Beshar IE, Bavan B, Milki AA, Lathi RB, Aghajanova L. Pregnancy and neonatal outcomes of letrozole versus natural cycle frozen embryo transfer of autologous euploid blastocyst. J Assist Reprod Genet 2023; 40:873-881. [PMID: 36849755 PMCID: PMC10224882 DOI: 10.1007/s10815-023-02759-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/01/2023] Open
Abstract
PURPOSE To investigate the pregnancy and neonatal outcomes of letrozole-stimulated frozen embryo transfer (LTZ-FET) cycles compared with natural FET cycles (NC-FET). METHODS Our retrospective cohort included all LTZ-FET (n = 161) and NC-FET (n = 575) cycles that transferred a single euploid autologous blastocyst from 2016 to 2020 at Stanford Fertility Center. The LTZ-FET protocol entailed 5 mg of daily letrozole for 5 days starting on cycle day 2 or 3. Outcomes were compared using absolute standardized differences (ASD), in which a larger ASD signifies a larger difference. Multivariable regression models adjusted for confounders: maternal age, BMI, nulliparity, embryo grade, race, infertility diagnosis, and endometrial thickness. RESULTS The demographic and clinical characteristics were overall similar. A greater proportion of the letrozole cohort was multiparous, transferred high-graded embryos, and had ovulatory dysfunction. The cohorts had similar pregnancy rates (67.1% LTZ vs 62.1% NC; aOR 1.31, P = 0.21) and live birth rates (60.9% LTZ vs 58.6% NC; aOR 1.17, P = 0.46). LTZ-FET neonates on average were born 5.7 days earlier (P < 0.001) and had higher prevalence of prematurity (18.6% vs. 8.0%NC, ASD = 0.32) and low birth weight (10.4% vs. 5.0%, ASD = 0.20). Both cohorts' median gestational ages (38 weeks and 1 day for LTZ; 39 weeks and 0 day for NC) were full term. CONCLUSION There were similar rates of pregnancy and live birth between LTZ-FET and NC-FET cycles. However, there was a higher prevalence of prematurity and low birth weight among LTZ-FET neonates. Reassuringly, the median gestational age in both cohorts was full term, and while the difference in gestational length of almost 6 days does not appear to be clinically significant, this warrants larger studies.
Collapse
Affiliation(s)
- Wendy Y Zhang
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Rebecca M Gardner
- Department of Medicine, Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, CA, USA
| | - Jasmyn K Johal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Isabel E Beshar
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brindha Bavan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amin A Milki
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruth B Lathi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lusine Aghajanova
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Hu KL, Li S, Hunt S, Yang R, Xu H, Li R. High anti-Müllerian hormone (AMH) is associated with increased risks of ectopic pregnancy in women undergoing fresh embryo transfer cycle, a cohort study. Reprod Biol Endocrinol 2023; 21:18. [PMID: 36737777 PMCID: PMC9896741 DOI: 10.1186/s12958-022-01038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/17/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Ectopic pregnancy is more common amongst assisted reproduction cycles and is a cause of significant maternal morbidity. Few predictive markers exist to help identify and modify risk of ectopic pregnancy in preparing for embryo transfer. The relationship between serum and AMH and ectopic pregnancy rate is unknown. METHODS This was a retrospective cohort study investigating women who underwent fresh embryo transfer cycles from January 2017 to December 2019 in Peking University Third Hospital. The primary outcome was ectopic pregnancy. Restricted cubic splines with four knots for AMH concentration (0-3, 3-6, 6-12, 12-max) were used to map out the non-linear relationship between the predicted ectopic pregnancy rate and the serum AMH concentration. Log binomial regression was used to test the crude risk ratio (cRR) and the adjusted risk ratio (aRR) after adjustment for confounders with 95% confidence intervals (CI) to determine the difference across various groups. RESULTS A total of 13,718 cycles in women undergoing fresh embryo transfer were eligible for analysis. The ectopic pregnancy rate was 1.3% per embryo transfer cycle initiated and 3.3% per pregnancy. Serum AMH concentrations were higher amongst women with ectopic pregnancy than in women with a confirmed intrauterine pregnancy or heterotopic pregnancy or who did not become pregnant (Mean levels: 4.0 ng/ml vs 3.2 ng/ml, 1.7 ng/ml, and 2.8 ng/ml). An AMH concentration of 7 ng/ml represented the best cut-off value to predict ectopic pregnancy. The ectopic pregnancy rate was 3.4% per cycle and 7.5% per pregnancy in women with AMH levels ≥ 7 ng/ml; and 1.2% per cycle and 2.9% per pregnancy in women with AMH levels < 7 ng/ml. Serum AMH concentration ≥ 7 ng/ml was associated with an increased risk of ectopic pregnancy in all fresh embryo transfer cycles started (aRR = 2.35 (1.45, 3.58)) as well in women who became pregnant (aRR = 2.23 (1.49, 3.33). CONCLUSIONS Baseline AMH concentration ≥ 7 ng/ml is associated with an increased risk of ectopic pregnancy in fresh embryo transfer cycles.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Shan Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Sarah Hunt
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3168, Australia
| | - Rui Yang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Huiyu Xu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 HuaYuan North Road, HaiDian District, Beijing, 100191, People's Republic of China.
| |
Collapse
|
12
|
Dysregulation in Multiple Transcriptomic Endometrial Pathways Is Associated with Recurrent Implantation Failure and Recurrent Early Pregnancy Loss. Int J Mol Sci 2022; 23:ijms232416051. [PMID: 36555686 PMCID: PMC9782216 DOI: 10.3390/ijms232416051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Overlapping disease aetiologies associated with multiple altered biological processes have been identified that change the endometrial function leading to recurrent implantation failure (RIF) and recurrent early pregnancy loss (REPL). We aimed to provide a detailed insight into the nature of the biological malfunction and related pathways of differentially expressed genes in RIF and REPL. Endometrial biopsies were obtained from 9 women experiencing RIF, REPL and control groups. Affymetrix microarray analysis was performed to measure the gene expression level of the endometrial biopsies. Unsupervised clustering of endometrial samples shows scattered distribution of gene expression between the RIF, REPL and control groups. 2556 and 1174 genes (p value < 0.05, Fold change > 1.2) were significantly altered in the endometria of RIF and REPL patients’ group, respectively compared to the control group. Downregulation in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the differentially expressed genes (DEGs) in RIF and REPL including ribosome and oxidative phosphorylation pathways. Gene Ontology (GO) analysis revealed ribosomes and mitochondria inner membrane as the most significantly downregulated cellular component (CC) affected in RIF and REPL. Determination of the dysregulated genes and related biological pathways in RIF and REPL will be key in understanding their molecular pathology and of major importance in addressing diagnosis, prognosis, and treatment issues
Collapse
|
13
|
Maziotis E, Kalampokas T, Giannelou P, Grigoriadis S, Rapani A, Anifantakis M, Kotsifaki A, Pantou A, Triantafyllidou O, Tzanakaki D, Neofytou S, Vogiatzi P, Bakas P, Simopoulou M, Vlahos N. Commercially Available Molecular Approaches to Evaluate Endometrial Receptivity: A Systematic Review and Critical Analysis of the Literature. Diagnostics (Basel) 2022; 12:2611. [PMID: 36359455 PMCID: PMC9689742 DOI: 10.3390/diagnostics12112611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/26/2022] [Indexed: 08/17/2023] Open
Abstract
Despite the advances in the field of reproductive medicine, implantation failure represents a challenging condition affecting 10-30% of patients subjected to in vitro fertilization (IVF). Research has focused on the identification of molecules playing crucial roles in endometrial receptivity, with the aim of designing predictive tools for efficient detection of the implantation window. To that end, novel molecular genomic and transcriptomic approaches have been introduced as promising tools to enable personalized approaches with the aim of optimizing embryo transfer dating. However, the clinical value of these approaches remains unclear. The aim of this study is to provide a systematic review and critical analysis of the existing evidence regarding the employment of commercially available novel approaches to evaluate endometrial receptivity. An Embase and PubMed/Medline search was performed on 1 February 2022. From the 475 articles yielded, only 27 were included and analyzed. The considerable heterogeneity of the included articles indicates the uniqueness of the implantation window, showcasing that the optimal time for embryo transfer varies significantly between women. Moreover, this study provides information regarding the technical aspects of these advanced molecular tools, as well as an analysis of novel possible biomarkers for endometrial receptivity, providing a basis for future research in the field.
Collapse
Affiliation(s)
- Evangelos Maziotis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
| | - Theodoros Kalampokas
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Polina Giannelou
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli Str., 15232 Athens, Greece
| | - Sokratis Grigoriadis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Anna Rapani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
| | - Marios Anifantakis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
| | - Amalia Kotsifaki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
| | - Agni Pantou
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli Str., 15232 Athens, Greece
| | - Olga Triantafyllidou
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Despoina Tzanakaki
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Spyridoula Neofytou
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Paraskevi Vogiatzi
- Andromed Health & Reproduction, Fertility Diagnostics Center, 3, Mesogion Str., 15126 Athens, Greece
| | - Panagiotis Bakas
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Mara Simopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., 11527 Athens, Greece
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece
| |
Collapse
|
14
|
Erten O, Taskomur AT, Albayrak M. Current Biomarkers for Endometrial Receptivity. Biomark Med 2022. [DOI: 10.2174/9789815040463122010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Implantation and the continuation of pregnancy occur through a complicated
and sophisticated dialogue, called “cross-talk,” which starts between the embryo and
the endometrium in the early stages of oocyte maturation. This dialogue provides
synchronization of the journey of the embryo to be implanted with the receptive
endometrium. Understanding the activity and function of the hormones and factors
involved in this dialogue will provide an understanding of endometrial receptivity,
which plays a key role in implantation, and the determination of biomarkers specific
for this period. As a result of the development of omics technology, it has become
possible to identify biomarkers specific to endometrial receptivity by performing
genomic, proteomic, and lipidomic analyses of these hormones and factors. The
determination of these biomarkers, their optimization, and making them usable in the
clinic will allow increased success in ART.
Collapse
Affiliation(s)
- Ozlem Erten
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences
University, Kutahya, Turkey
| | - Aysun Tekeli Taskomur
- Department of Obstetrics and Gynecology, Faculty of Medicine, Amasya University, Amasya,
Turkey
| | - Mustafa Albayrak
- Department of Obstetrics and Gynecology, Sisli Florence Nightingale Hospital, Istanbul, Turkey
| |
Collapse
|
15
|
Qu Y, Zhang J, Guo S, Zhang L, Qian J, Zhu X, Duan E, Zhang Y. Three-Dimensional Visualization of Mouse Endometrial Remodeling After Superovulation. Front Cell Dev Biol 2022; 10:933852. [PMID: 35846371 PMCID: PMC9284589 DOI: 10.3389/fcell.2022.933852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Uterine status determines pregnancy success. Although it is well known that superovulation operations can disrupt uterine function, our understanding of the morphological changes in the uterine endometrium at the three-dimensional (3D) level is limited. Here, combining the tissue clearing with 3D deep imaging, we reveal an increase in epithelial density and angiogenesis after ovarian stimulation, which is accompanied by a circulating surge in P4 levels. Using an ovariectomized mouse model, we further detected the separate regulatory effects of P4 and E2 on the uterine endometrium, with P4 promoting endothelial cell growth and E2 inducing epithelial proliferation. Additionally, we observed that the effects of E2 can be partially neutralized by P4, and vice versa. By analyzing the 3D uterine imaging, we discovered an interesting phenomenon in which the growing blood vessels closely surround the remodeling uterine epithelium, indicating a close relationship between angiogenesis and epithelial growth. These findings provide new insight into the uterine epithelial changes and angiogenesis at the 3D level, and explain a potential reason for endometrial changes due to the low implantation rate in patients undergoing clinic super-ovulation.
Collapse
Affiliation(s)
- Yongcun Qu
- Institute of Artificial Intelligence in Sports, Capital University of Physical Education and Sports, Beijing, China
| | - Jia Zhang
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Shanshan Guo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Liwen Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jingjing Qian
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xili Zhu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Enkui Duan
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying Zhang
- College of Life Sciences, Beijing Normal University, Beijing, China
- *Correspondence: Ying Zhang,
| |
Collapse
|
16
|
Chang Y, Shen M, Wang S, Li X, Duan H. Association of embryo transfer type with infertility in endometriosis: a systematic review and meta-analysis. J Assist Reprod Genet 2022; 39:1033-1043. [PMID: 35332423 PMCID: PMC9107540 DOI: 10.1007/s10815-022-02460-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/10/2022] [Indexed: 11/28/2022] Open
Abstract
PURPOSE The study aims to evaluate whether frozen embryo transfer can restore optimal receptivity leading to better assisted reproductive technology outcomes in women with endometriosis. METHODS This systematic review and meta-analysis, conducted from January 10, 2021 to July 1, 2021, searched the Cochrane Library, PubMed, Embase, Web of Science, OVID, and Clinicaltrials.gov databases from inception to January 10, 2021. The search strategy combined search terms as follows: ("endometriosis" OR "deep endometriosis" OR "endometrioma") AND ("frozen-thawed embryo transfer" OR "frozen embryo transfer" OR "freeze-all strategy") AND ("pregnancy outcome" OR "live birth rate" OR "clinical pregnancy rate" OR "miscarriage rate"). No publication time or language limits were set during the searches. In addition, references of the related articles were searched by hand. Patients were included if they had a history of endometriosis and had received fresh or frozen embryo transfer. Only the first transfer cycle was included. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to express outcomes, and data synthesis was conducted using RevMan, version 5.4 software. RESULTS A total of six studies with moderate methodologic quality were retrieved in the meta-analysis. The studies included 3010 women with endometriosis who wanted to conceive; 1777 (59.0%) had frozen embryo transfer, and 1233 (41.0%) had fresh embryo transfer. There was a significantly higher frequency of live births in the frozen embryo group than in the fresh embryo group (OR, 1.53; 95% CI, 1.13-2.08; P = .007). Despite a similar clinical pregnancy rate in the two groups (OR, 1.26; 95% CI, 0.95-1.69; P = .11), the difference in miscarriage rate was significant (OR, 0.70; 95% CI, 0.50-0.97; P = .03). Evidence quality was considered moderate. CONCLUSION Cryopreserved embryo transfer has resulted in preferable reproduction outcomes when compared with fresh embryo transfer in patients with endometriosis, but the evidence is not yet abundant. More strictly designed research is needed to evaluate whether frozen embryo transfer leads to better reproductive outcomes in women with endometriosis compared with those receiving fresh embryo transfer. REGISTRATION NUMBER PROSPERO CRD42021248313.
Collapse
Affiliation(s)
- Yanan Chang
- Department of Minimally Invasive Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 17 Qihelou Street, Dong Cheng District, Beijing, 100006, China
| | - Minghong Shen
- Department of Minimally Invasive Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 17 Qihelou Street, Dong Cheng District, Beijing, 100006, China
- Department of Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, 350001, Fujian Province, China
| | - Sha Wang
- Department of Minimally Invasive Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 17 Qihelou Street, Dong Cheng District, Beijing, 100006, China
| | - Xiao Li
- Department of Minimally Invasive Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 17 Qihelou Street, Dong Cheng District, Beijing, 100006, China
| | - Hua Duan
- Department of Minimally Invasive Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 17 Qihelou Street, Dong Cheng District, Beijing, 100006, China.
| |
Collapse
|
17
|
Diaz-Gimeno P, Sebastian-Leon P, Sanchez-Reyes JM, Spath K, Aleman A, Vidal C, Devesa-Peiro A, Labarta E, Sánchez-Ribas I, Ferrando M, Kohls G, García-Velasco JA, Seli E, Wells D, Pellicer A. Identifying and optimizing human endometrial gene expression signatures for endometrial dating. Hum Reprod 2022; 37:284-296. [PMID: 34875061 DOI: 10.1093/humrep/deab262] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION What are the key considerations for developing an enhanced transcriptomic method for secretory endometrial tissue dating? SUMMARY ANSWER Multiple gene expression signature combinations can serve as biomarkers for endometrial dating, but their predictive performance is variable and depends on the number and identity of the genes included in the prediction model, the dataset characteristics and the technology employed for measuring gene expression. WHAT IS KNOWN ALREADY Among the new generation of transcriptomic endometrial dating (TED) tools developed in the last decade, there exists variation in the technology used for measuring gene expression, the gene makeup and the prediction model design. A detailed study, comparing prediction performance across signatures for understanding signature behaviour and discrepancies in gene content between them, is lacking. STUDY DESIGN, SIZE, DURATION A multicentre prospective study was performed between July 2018 and October 2020 at five different centres from the same group of clinics (Spain). This study recruited 281 patients and finally included in the gene expression analysis 225 Caucasian patients who underwent IVF treatment. After preprocessing and batch effect filtering, gene expression measurements from 217 patients were combined with artificial intelligence algorithms (support vector machine, random forest and k-nearest neighbours) allowing evaluation of different prediction models. In addition, secretory-phase endometrial transcriptomes from gene expression omnibus (GEO) datasets were analysed for 137 women, to study the endometrial dating capacity of genes independently and grouped by signatures. This provided data on the consistency of prediction across different gene expression technologies and datasets. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial biopsies were analysed using a targeted TruSeq (Illumina) custom RNA expression panel called the endometrial dating panel (ED panel). This panel included 301 genes previously considered relevant for endometrial dating as well as new genes selected for their anticipated value in detecting the secretory phase. Final samples (n = 217) were divided into a training set for signature discovery and an independent testing set for evaluation of predictive performance of the new signature. In addition, secretory-phase endometrial transcriptomes from GEO were analysed for 137 women to study endometrial dating capacity of genes independently and grouped by signatures. Predictive performance among these signatures was compared according to signature gene set size. MAIN RESULTS AND THE ROLE OF CHANCE Testing of the ED panel allowed development of a model based on a new signature of 73 genes, which we termed 'TED' and delivers an enhanced tool for the consistent dating of the secretory phase progression, especially during the mid-secretory endometrium (3-8 days after progesterone (P) administration (P + 3-P + 8) in a hormone replacement therapy cycle). This new model showed the best predictive capacity in an independent test set for staging the endometrial tissue in the secretory phase, especially in the expected window of implantation (average of 114.5 ± 7.2 h of progesterone administered; range in our patient population of 82-172 h). Published sets of genes, in current use for endometrial dating and the new TED genes, were evaluated in parallel in whole-transcriptome datasets and in the ED panel dataset. TED signature performance was consistently excellent for all datasets assessed, frequently outperforming previously published sets of genes with a smaller number of genes for dating the endometrium in the secretory phase. Thus, this optimized set exhibited prediction consistency across datasets. LARGE SCALE DATA The data used in this study is partially available at GEO database. GEO identifiers GSE4888, GSE29981, GSE58144, GSE98386. LIMITATIONS, REASONS FOR CAUTION Although dating the endometrial biopsy is crucial for investigating endometrial progression and the receptivity process, further studies are needed to confirm whether or not endometrial dating methods in general are clinically useful and to guide the specific use of TED in the clinical setting. WIDER IMPLICATIONS OF THE FINDINGS Multiple gene signature combinations provide adequate endometrial dating, but their predictive performance depends on the identity of the genes included, the gene expression platform, the algorithms used and dataset characteristics. TED is a next-generation endometrial assessment tool based on gene expression for accurate endometrial progression dating especially during the mid-secretory. STUDY FUNDING/COMPETING INTEREST(S) Research funded by IVI Foundation (1810-FIVI-066-PD). P.D.-G. visiting scientist fellowship at Oxford University (BEFPI/2010/032) and Josefa Maria Sanchez-Reyes' predoctoral fellowship (ACIF/2018/072) were supported by a program from the Generalitat Valenciana funded by the Spanish government. A.D.-P. is supported by the FPU/15/01398 predoctoral fellowship from the Ministry of Science, Innovation and Universities (Spanish Government). D.W. received support from the NIHR Oxford Biomedical Research Centre. The authors do not have any competing interests to declare.
Collapse
Affiliation(s)
- P Diaz-Gimeno
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Level 3, Women's Centre John Radcliffe Hospital, Oxford, UK
| | - P Sebastian-Leon
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - J M Sanchez-Reyes
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - K Spath
- Research Department, JUNO Genetics, Oxford, UK
| | - A Aleman
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - C Vidal
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Reproductive medicine, IVI RMA Valencia, Valencia, Spain
| | - A Devesa-Peiro
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - E Labarta
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Reproductive medicine, IVI RMA Valencia, Valencia, Spain
| | - I Sánchez-Ribas
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Reproductive medicine, IVI RMA Barcelona, Barcelona, Spain
| | - M Ferrando
- Reproductive medicine, IVI RMA Bilbao, Leioa, Bizkaia, Spain
| | - G Kohls
- Reproductive medicine, IVI RMA Madrid, Madrid, Spain
| | - J A García-Velasco
- Reproductive medicine, IVI RMA Madrid, Madrid, Spain
- Department of Obstetrics and Gynecology, Universidad Rey Juan Carlos, Madrid, Spain
| | - E Seli
- Research Department, IVI RMA New Jersey, Basking Ridge, NJ, USA
- Department of Obstetrics, Gynecology & Reproductive Science, Yale School of Medicine, New Haven, CT, USA
| | - D Wells
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Level 3, Women's Centre John Radcliffe Hospital, Oxford, UK
- Research Department, JUNO Genetics, Oxford, UK
| | - A Pellicer
- Genomic & Systems Reproductive Medicine, IVI Foundation/Instituto de investigación sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
- Research Department, JUNO Genetics, Oxford, UK
- Reproductive medicine, IVI RMA Rome, Roma, Italy
| |
Collapse
|
18
|
Ji M, Jin B, Guo X, Wu R, Jiang Y, Zhang L, Shu J. It is not worth postponing frozen embryo transfers after oocyte pickup: A retrospective cohort study based on propensity score matching. Front Endocrinol (Lausanne) 2022; 13:971616. [PMID: 36133317 PMCID: PMC9483166 DOI: 10.3389/fendo.2022.971616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
This study was to explore whether postponing frozen embryo transfers (FET) after oocyte pickup (OPU) improves clinical and neonatal outcomes. From May 2018 to Dec 2020, a total of 1109 patients underwent their first OPU cycles adopting a non-selective freeze-all policy were included in this retrospective cohort study. In the immediate group (n=219), patients underwent FET in the first menstrual cycle after OPU, and patients in the postponed group (n=890) waited for more than 1 menstrual cycle after OPU to perform FET. A propensity score matching (PSM) model was used to evaluate the clinical outcomes and neonatal outcomes between the two groups. There were 209 patients in the immediate group and 499 patients in the postponed one after PSM. Patients waited for a significantly shorter period for FET in the immediate group (30.74 ± 3.85 days) compared with the postponed group (80.39 ± 26.25 days, P<0.01). The clinical pregnancy rate (CPR) and live birth rate (LBR) in the immediate group were 58.4% and 48.3%, respectively, which were comparable to those of the postponed one (58.1%, 49.7%, P > 0.05). No statistical significance was found in the average birth weight (3088.82 ± 565.35 g vs 3038.64 ± 625.78 g, P > 0.05) and height (49.08 ± 1.87 cm vs 49.30 ± 2.52 cm) of neonates between the two groups. The gender ratio, the incidence of macrosomia and low birth weight did not differ significantly between the two groups. In conclusion, postponing FET does not improve clinical and neonatal outcomes. If patients have no contraindications, FETs should be carried out immediately after OPU.
Collapse
Affiliation(s)
- Mengxia Ji
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Bihui Jin
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaoyan Guo
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ruifang Wu
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yunqing Jiang
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ling Zhang
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jing Shu
- Department of Reproductive Endocrinology, Center for Reproductive Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
19
|
Wang N, Zhu W, Gong Y. Association of raised serum progesterone level with ovulation trigger and histology of endometrium in stimulated cycles. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Na Wang
- Shanghai Jiao Tong University, China
| | | | | |
Collapse
|
20
|
Abe M, Yamamoto Y, Noguchi H, Tamura K, Aoki H, Takeda A, Minato S, Kamada S, Tachibana A, Iwasa T. Is a freeze-all strategy necessary for all embryo transfers : Fresh embryo transfer without progesterone elevation results in an equivalent pregnancy rate to cryopreserved embryo transfer. THE JOURNAL OF MEDICAL INVESTIGATION 2022; 69:224-229. [PMID: 36244773 DOI: 10.2152/jmi.69.224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Objectives : It has been suggested that the clinical outcomes of frozen-thawed embryo transfer (ET) are superior to those of fresh embryo transfer. We examined whether a freeze-all strategy is necessary for all embryo transfers, and, if not, to evaluate the conditions in which the pregnancy rates of fresh embryo transfer and frozen-thawed ET did not differ. Methods : Patients who underwent blastocyst transfer at Tokushima University Hospital between 2008 and 2019 were enrolled. The clinical outcomes and clinical characteristics of 1,022 patients that underwent fresh embryo transfer and 1,728 patients that underwent frozen-thawed ET were examined retrospectively. We considered the factors that influenced the pregnancy outcomes of fresh embryo transfer. Results : The frozen-thawed ET group exhibited significantly higher pregnancy, live-birth, and miscarriage rates than the fresh embryo transfer group. In the fresh embryo transfer group, a high progesterone level on the day of the human chorionic gonadotropin (hCG) trigger and lower grade embryos were risk factors for a low pregnancy rate. However, in the cases in which the progesterone level was < 1.0 ng / mL the pregnancy rate was equal to that of frozen-thawed ET. Conclusions : A freeze-all strategy is not necessary for embryo transfers, but should be employed in cases involving pre-ovulatory progesterone elevation. J. Med. Invest. 69 : 224-229, August, 2022.
Collapse
Affiliation(s)
- Masami Abe
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuri Yamamoto
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroki Noguchi
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kou Tamura
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hidenori Aoki
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Asuka Takeda
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Saki Minato
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shuhei Kamada
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ayaka Tachibana
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takeshi Iwasa
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
21
|
Haouzi D, Entezami F, Tuaillon E, Gala A, Ferrières-Hoa A, Brouillet S, Thierry AR, Hamamah S. SARS-CoV-2 and Implantation Window: Gene Expression Mapping of Human Endometrium and Preimplantation Embryo. Life (Basel) 2021; 11:life11121378. [PMID: 34947909 PMCID: PMC8706202 DOI: 10.3390/life11121378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022] Open
Abstract
Understanding whether SARS-CoV-2 could infect cells and tissues handled during ART is crucial for risk mitigation, especially during the implantation window when either endometrial biopsies are often practiced for endometrial receptivity assessment or embryo transfer is performed. To address this question, this review analyzed current knowledge of the field and retrospectively examined the gene expression profiles of SARS-CoV-2-associated receptors and proteases in a cohort of ART candidates using our previous Affymetrix microarray data. Human endometrial tissue under natural and controlled ovarian stimulation cycles and preimplantation embryos were analyzed. A focus was particularly drawn on the renin-angiotensin system, which plays a prominent role in the virus infection, and we compared the gene expression levels of receptors and proteases related to SARS-CoV-2 infection in the samples. High prevalence of genes related to the ACE2 pathway during both cycle phases and mainly during the mid-secretory phase for ACE2 were reported. The impact of COS protocols on endometrial gene expression profile of SARS-CoV-2-associated receptors and proteases is minimal, suggesting no additional potential risks during stimulated ART procedure. In blastocysts, ACE2, BSG, CTSL, CTSA and FURIN were detectable in the entire cohort at high expression level. Specimens from female genital tract should be considered as potential targets for SARS-CoV-2, especially during the implantation window.
Collapse
Affiliation(s)
- Delphine Haouzi
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- IRMB (Institute for Regenerative Medicine & Biotherapy), Univ Montpellier, INSERM, 34295 Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, 34295 Montpellier, France
- Global ART Innovation Network, IRMB, CHU Montpellier, 34295 Montpellier, France
| | - Frida Entezami
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- ART Department, American Hospital of Paris, 92200 Neuilly-Sur-Seine, France
| | - Edward Tuaillon
- CHU Montpellier, Bacteriology-Virology Department, 34295 Montpellier, France;
| | - Anna Gala
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, 34295 Montpellier, France
| | - Alice Ferrières-Hoa
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, 34295 Montpellier, France
| | - Sophie Brouillet
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- IRMB (Institute for Regenerative Medicine & Biotherapy), Univ Montpellier, INSERM, 34295 Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, 34295 Montpellier, France
| | - Alain R. Thierry
- Regional Institute of Cancer of Montpellier, 34090 Montpellier, France;
| | - Samir Hamamah
- Univ Montpellier, INSERM U1203, DEFE, 34295 Montpellier, France; (D.H.); (F.E.); (A.G.); (A.F.-H.); (S.B.)
- IRMB (Institute for Regenerative Medicine & Biotherapy), Univ Montpellier, INSERM, 34295 Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, 34295 Montpellier, France
- Global ART Innovation Network, IRMB, CHU Montpellier, 34295 Montpellier, France
- Correspondence: ; Tel.: +33-04-67-33-64-04; Fax: +33-04-67-33-62-90
| |
Collapse
|
22
|
Zhang WY, Gardner RM, Kapphahn KI, Ramachandran MK, Murugappan G, Aghajanova L, Lathi RB. The impact of estradiol on pregnancy outcomes in letrozole-stimulated frozen embryo transfer cycles. F S Rep 2021; 2:320-326. [PMID: 34553158 PMCID: PMC8441577 DOI: 10.1016/j.xfre.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE To assess the impact of low estradiol (E2) levels in letrozole-stimulated frozen embryo transfer (FET) cycles on pregnancy and neonatal outcomes. DESIGN Retrospective cohort. SETTING University-affiliated fertility center. PATIENTS All patients who underwent letrozole-stimulated FET cycles from January 2017 to April 2020 (n = 217). The "Low E2" group was defined as those with E2 serum levels on the day of trigger <10th percentile level (E2 <91.16 pg/mL, n = 22) and the "Normal E2" group was defined as those with E2 serum levels ≥10th percentile level (E2 ≥91.16 pg/mL, n = 195). INTERVENTIONS None. MAIN OUTCOME MEASURES Pregnancy outcomes including rates of clinical pregnancy, clinical miscarriage, and live birth. Neonatal outcomes including gestational age at delivery, birth weight, and Apgar score. RESULTS The mean ± SD estradiol level was 66.8 ± 14.8 pg/mL for the "Low E2" group compared with 366.3 ± 322.1 pg/mL for the "Normal E2" group. There were otherwise no substantial differences in cycle characteristics such as endometrial thickness on the day of ovulation trigger and progesterone levels in early pregnancy. The "Low E2" group had a significantly higher clinical miscarriage rate (36.4% vs. 8.8%, adjusted odds ratio 8.06) and lower live birth rate (31.8% vs. 57.9%, adjusted odds ratio 0.28). Neonatal outcomes such as gestational age at delivery, mean birth weight, Apgar scores, and incidence of newborn complications were not clinically different between the groups. CONCLUSION Low E2 levels were associated with a significantly higher miscarriage rate and lower live birth rate, suggesting that E2 levels in the follicular phase may have an effect on cycle outcomes. Given the rise in use of FET, further studies are needed to confirm our findings and understand the mechanisms.
Collapse
Affiliation(s)
- Wendy Y. Zhang
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Rebecca M. Gardner
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristopher I. Kapphahn
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Maya K. Ramachandran
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Gayathree Murugappan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Lusine Aghajanova
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Ruth B. Lathi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
23
|
Zhang J, Jin N, Ma Y, Lu J, Wang J, Chen S, Wang X. Ovarian stimulation reduces fetal growth by dysregulating uterine natural killer cells in mice. Mol Reprod Dev 2021; 88:618-627. [PMID: 34409664 DOI: 10.1002/mrd.23528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 05/27/2021] [Accepted: 07/31/2021] [Indexed: 11/08/2022]
Abstract
Ovarian stimulation is associated with a higher risk of low birth weight. However, the precise mechanisms by which ovarian stimulation increases the chances of low birth weight remain unclear. In this mouse model study, in vivo developed blastocysts that were not exposed to gonadotropins were transferred into pseudopregnant females that had mated naturally (the control group), pseudopregnant females that had been administered a low dose of ovulation-stimulating hormone (the L-SO group) and pseudopregnant females that had been administered a high dose of ovulation-stimulating hormone (the H-SO group). The embryo implantation rate and fetal weight were significantly lower in the L-SO and H-SO groups than in the control group. The density of Dolichos biflorus agglutinin (DBA)+ uterine natural killer (uNK) cells in the decidua basalis was significantly lower in the L-SO and H-SO groups than in the control group. Ovarian stimulation also downregulated a variety of cytokines related to uNK cells that are involved in placental angiogenesis and trophoblast invasion. Collectively, our findings indicate that ovarian stimulation impairs DBA+ uNK cell density in the decidua basalis, which may downregulate uNK-related cytokine secretion and influence placental angiogenesis and restrict fetal growth in mice.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China.,Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Ni Jin
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yuan Ma
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jie Lu
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jingjing Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Shuqiang Chen
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
24
|
Pathare ADS, Hinduja I. Endometrial Expression of Cell Adhesion Genes in Recurrent Implantation Failure Patients in Ongoing IVF Cycle. Reprod Sci 2021; 29:513-523. [PMID: 34410653 DOI: 10.1007/s43032-021-00708-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/30/2021] [Indexed: 11/28/2022]
Abstract
Recurrent implantation failure (RIF) is one of the major obstacles in IVF. Transcriptomic literature has revealed the various biological processes involved in endometrial receptivity (ER) under different physiological circumstances, especially in natural cycle. We intended to determine the function-specific ER profile under controlled ovarian stimulation (COS) cycle. This can help to back trace the genomic impairment in RIF patients during the IVF cycle and to validate the genes involved in enriched pathways. In our study, retrospective gene expression microarray dataset was reanalysed after the follow-up, in classic non pregnant RIF (cases) vs fertile women (controls) under COS (n = 5/group). Reanalysis of microarray revealed significant downregulation of cell adhesion function (P:3.11E-05) with the maximum gene count. For validation purpose, downregulation of eight genes (COMP, HABP2, ITGAD, CDH3, COL22A1, MFAP4, THBS1and CD300A) involved in enriched cell adhesion pathway having fold change > 3 were assessed by real-time PCR in independent cohorts of cases and controls (n = 24, each). Downregulation of six out of eight genes (COMP, HABP2, ITGAD, CDH3, MFAP4 and THBS1) were confirmed by real-time PCR (P < 0.05) with fold change > 2. This indicates the importance of analysed genes in the ER mechanism under COS, thus mimicking the fresh embryo transfer. The further analysis in larger cohorts would substantiate the study findings in RIF patients undergoing IVF cycle.
Collapse
Affiliation(s)
- Amruta D S Pathare
- Department of IVF and Research, P.D. Hinduja Hospital and Medical Research Centre, Mumbai, 400016, Maharashtra, India
| | - Indira Hinduja
- Department of IVF and Research, P.D. Hinduja Hospital and Medical Research Centre, Mumbai, 400016, Maharashtra, India.
| |
Collapse
|
25
|
Zuo N, Gao Y, Zhang N, Li D, Wang X. Effects of immediate versus delayed frozen embryo transfer in high responder patients undergoing freeze-all cycles. BMC Pregnancy Childbirth 2021; 21:455. [PMID: 34182954 PMCID: PMC8240376 DOI: 10.1186/s12884-021-03919-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/28/2021] [Indexed: 01/03/2023] Open
Abstract
Background Frozen embryo transfer (FET) can greatly improve the pregnancy outcomes for high responder patients. However, it is not known whether the timing of FET is a risk factor on pregnancy outcomes in high responder patients undergoing freeze-all cycles. Methods A retrospective cohort study to compare the pregnancy outcomes of the immediate and delayed FET groups in high responder patients undergoing freeze-all cycles. The two groups were defined as that FET took place either within the first menstrual cycle following oocyte retrieval or afterwards. Propensity score matching was used to make the potential risk factors of the two groups comparable. Multivariable regression analysis was used to study the effect of the timing of FET on pregnancy outcomes in the entire cohort and propensity score-matched cohort, even in different controlled ovarian hyperstimulation protocol cohorts as subgroup analysis. Results We obtained 1130 patients in immediate FET group and 998 patients in delayed FET group, and the average age of the two groups were 30.30 and 30.63. We showed that the immediate FET group were equivalent to delayed FET group in the entire cohort [clinical pregnancy rate (CPR), 61.0% versus 63.4%, adjusted odd ratio (OR), 0.939, 95% confidence interval (CI), 0.781–1.129; spontaneous abortion rate (SAR), 10.1% versus 12.6%, adjusted OR, 0.831, 95% Cl (0.628–1.098); live birth rate (LBR), 49.9% versus 49.2%, adjusted OR, 1.056, 95% Cl (0.883–1.263)]. The same results were obtained by χ2 test in the propensity score-matched cohort (CPR, 60.5% versus 63.5%; SAR, 11.6% versus 12.3%; LBR, 48% versus 49.3%) (P > 0.05). Subgroup analysis indicated that pregnancy outcomes of immediate FET were no difference to delayed FET in gonadotropin-releasing hormone agonist (GnRH-a) protocol (P > 0.05). The SAR of the immediate FET group were lower than that of the delayed FET group in GnRH antagonist protocol (adjusted OR, 0.645, 95% CI, 0.430–0.966) (P < 0.05), no differences were observed in CPR and LBR (P > 0.05). Conclusions The pregnancy outcomes of immediate FET were no difference to delayed FET in high responder population undergoing freeze-all cycles.
Collapse
Affiliation(s)
- Na Zuo
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yingzhuo Gao
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ningning Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Xiuxia Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
26
|
Kalinderis M, Kalinderi K, Srivastava G, Homburg R. When Should We Freeze Embryos? Current Data for Fresh and Frozen Embryo Replacement IVF Cycles. Reprod Sci 2021; 28:3061-3072. [PMID: 34033111 DOI: 10.1007/s43032-021-00628-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
Recent years have seen a dramatic rise in the number of frozen-thawed embryo replacement (FER) cycles. Along with the advances in embryo cryopreservation techniques, the optimization of endometrial receptivity has resulted in outcomes for FER that are similar to fresh embryo transfer. However, the question of whether the Freeze all strategy is for all is nowadays a hot topic. This review addresses this issue and describes current evidence based on randomized controlled trials and observational studies. To date, it is reasonable to perform FER in cases with a clear indication for the benefits of such strategy including impending ovarian hyperstimulation syndrome (OHSS) or preimplantation genetic testing for aneuploidy (PGT-A); however, this strategy does not fit for all. This review analyses the pros and cons of the freeze all strategy highlighting the need to follow a personalized plan in embryo transfer, avoiding a freeze all methodology for all patients in an unselected manner.
Collapse
Affiliation(s)
| | - Kallirhoe Kalinderi
- 3rd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Roy Homburg
- Homerton Fertility Centre, Homerton University Hospital, London, UK.,Queen Mary University of London, London, UK
| |
Collapse
|
27
|
Supraphysiological estradiol levels on the hCG trigger day are associated with SGA for singletons born from fresh embryo transfer. J Dev Orig Health Dis 2021; 13:244-251. [PMID: 33971999 DOI: 10.1017/s2040174421000234] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The effects of supraphysiological estradiol (E2) on neonatal outcomes and the significance of specific E2 concentrations remain unclear. The purpose of this study was to investigate whether supraphysiological E2 levels on the human chorionic gonadotropin (hCG) trigger day are associated with small size for gestational age (SGA) in singletons born from fresh embryo transfer (ET) cycles. Patients with singleton pregnancies who delivered after the transfer of fresh embryos, during the period from July 2012 to December 2017, at our center were included. We excluded cycles involving a vanishing twin, maternal age >35 years, basal follicle-stimulating hormone ≥10 mIU/ml, or anti-Müllerian hormone ≤1 ng/ml. We then divided all cycles into five groups by E2 level on trigger day: group A, <2000 pg/ml (reference group); group B, 2000 pg/ml≤E2<2999 pg/ml; group C, 3000 pg/ml≤E2<3999 pg/ml; group D, 4000 pg/ml≤E2<4999 pg/ml; and group E, ≥5000 pg/ml. The prevalence of SGA among singletons from fresh ET was the primary outcome. The SGA rate significantly increased when the E2 level was ≥4000 pg/ml, as observed by comparing groups D (odds ratio [OR]: 1·79, 95% confidence interval [CI]: 1·16-2·76, P = 0·01) and E (OR: 1·68, 95% CI: 1·10-2·56, P = 0·02) with the reference group. Multivariate logistic regression indicated that a serum E2 level of at least 4000 pg/ml on the hCG trigger day was associated with increased SGA and with significant differences for groups D (adjusted OR [AOR]: 1·65, 95% CI: 1·05-2·59, P = 0·03) and E (AOR: 1·60, 95% CI: 1·03-2·53, P = 0·04) relative to the reference group. In conclusion, in fresh ET cycles, the supraphysiological E2 ≥4000 pg/ml on the hCG trigger day increases the risk of SGA.
Collapse
|
28
|
Acet F, Hortu I, Sahin G, Goker ENT, Tavmergen E. Is frozen embryo transfer better than fresh embryo transfer in women undergoing intracytoplasmic sperm injection over the age of thirty-five? A single referral centre experience. J OBSTET GYNAECOL 2021; 42:276-280. [PMID: 33913396 DOI: 10.1080/01443615.2021.1882973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Developments in embryo freezing techniques in recent years have increased the chance of pregnancy after frozen embryo transfers. In this study we aimed to compare the pregnancy outcomes between fresh (ET) and frozen-thawed embryo transfer (FET) and to evaluate the benefits of embryo freezing strategy for a woman of advanced age. A total of 513 ET cycles in women over the age of 35 years, including 397 fresh ET cycles and 116 FET cycles were reviewed. Mean age was 37.7 ± 2.91 years in FET cycles and 38 ± 2.8 in fresh cycles (p = .327). The patients undergoing FET cycles had higher oestradiol levels and progesterone levels on the day of trigger in their previous fresh cycles compared to that of fresh ET cycles (1675 pmol/L versus 991 pmol/L; 1.43 pmol/L versus 0.96 pmol/L; p < .005, respectively). Biochemical pregnancy rates (43% versus 32%; p = .048), clinical pregnancy rates (38% versus 29%; p = .030) and live birth rates (30% versus 19.6%; p = .013) were significantly higher in the FET than in the fresh ET over 35 years of age.IMPACT STATEMENTWhat is alreday known on this subject? Ovarian stimulation commonly results in the generation of more embryos than are necessary for the fresh embryo transfer. Therefore, cryopreservation and subsequent replacement of frozen-thawed embryos is an integral part of assisted reproductive technique (ART) programs. As IVF technology improved, embryo freezing was performed to allow subsequent transfer if the fresh cycle was unsuccessful.What do the results of this study add? Clinical pregnancy rates and live birth rates were found to be higher in frozen thawed embryo transfer group compared to fresh embryo transfer group over the age of 35.What are the implications of these findings for clinical practice and/or further research? Frozen thawed embryo transfer seems to be a reasonable and favourable method compared to fresh embryo transfer in patients over 35 years.
Collapse
Affiliation(s)
- Ferruh Acet
- Department of IVF Research and Training Center, Ege University School of Medicine, Izmir, Turkey
| | - Ismet Hortu
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey.,Department of Stem Cell, Ege University Institute of Health Sciences, Izmir, Turkey
| | - Gulnaz Sahin
- Department of IVF Research and Training Center, Ege University School of Medicine, Izmir, Turkey
| | - Ege Nazan Tavmergen Goker
- Department of IVF Research and Training Center, Ege University School of Medicine, Izmir, Turkey.,Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Erol Tavmergen
- Department of IVF Research and Training Center, Ege University School of Medicine, Izmir, Turkey.,Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| |
Collapse
|
29
|
Mackens S, Santos-Ribeiro S, Racca A, Daneels D, Koch A, Essahib W, Verpoest W, Bourgain C, Van Riet I, Tournaye H, Brosens JJ, Lee YH, Blockeel C, Van de Velde H. The proliferative phase endometrium in IVF/ICSI: an in-cycle molecular analysis predictive of the outcome following fresh embryo transfer. Hum Reprod 2021; 35:130-144. [PMID: 31916571 DOI: 10.1093/humrep/dez218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does an early proliferative phase endometrial biopsy harvested during ovarian stimulation harbour information predictive of the outcome following fresh embryo transfer (ET) in that same cycle? SUMMARY ANSWER Transcriptome analysis of the whole-tissue endometrium did not reveal significant differential gene expression (DGE) in relation to the outcome; however, the secretome profile of isolated, cultured and in vitro decidualized endometrial stromal cells (EnSCs) varied significantly between patients who had a live birth compared to those with an implantation failure following fresh ET in the same cycle as the biopsy. WHAT IS KNOWN ALREADY In the majority of endometrial receptivity research protocols, biopsies are harvested during the window of implantation (WOI). This, however, precludes ET in that same cycle, which is preferable as the endometrium has been shown to adapt over time. Endometrial biopsies taken during ovarian stimulation have been reported not to harm the chances of implantation, and in such biopsies DGE has been observed between women who achieve pregnancy versus those who do not. The impact of the endometrial proliferative phase on human embryo implantation remains unclear, but deserves further attention, especially since in luteal phase endometrial biopsies, a transcriptional signature predictive for repeated implantation failure has been associated with reduced cell proliferation, possibly indicating proliferative phase involvement. Isolation, culture and in vitro decidualization (IVD) of EnSCs is a frequently applied basic research technique to assess endometrial functioning, and a disordered EnSC secretome has previously been linked with failed implantation. STUDY DESIGN, SIZE, DURATION This study was nested in a randomized controlled trial (RCT) investigating the effect of endometrial scratching during the early follicular phase of ovarian stimulation on clinical pregnancy rates after IVF/ICSI. Of the 96 endometrial biopsies available, after eliminating those without fresh ET and after extensive matching in order to minimize the risk of potential confounding, 18 samples were retained to study two clinical groups: nine biopsies of patients with a live birth versus nine biopsies of patients with an implantation failure, both following fresh ET performed in the same cycle as the biopsy. We studied the proliferative endometrium by analysing its transcriptome and by isolating, culturing and decidualizing EnSCs in vitro. We applied this latter technique for the first time on proliferative endometrial biopsies obtained during ovarian stimulation for in-cycle outcome prediction, in an attempt to overcome inter-cycle variability. PARTICIPANTS/MATERIALS, SETTING, METHODS RNA-sequencing was performed for 18 individual whole-tissue endometrial biopsies on an Illumina HiSeq1500 machine. DGE was analysed three times using different approaches (DESeq2, EdgeR and the Wilcoxon rank-sum test, all in R). EnSC isolation and IVD was performed (for 2 and 4 days) for a subset of nine samples, after which media from undifferentiated and decidualized cultures were harvested, stored at -80°C and later assayed for 45 cytokines using a multiplex suspension bead immunoassay. The analysis was performed by partial least squares regression modelling. MAIN RESULTS AND THE ROLE OF CHANCE After correction for multiple hypothesis testing, DGE analysis revealed no significant differences between endometrial samples from patients who had a live birth and those with an implantation failure following fresh ET. However secretome analysis after EnSC isolation and culture, showed two distinct clusters that clearly corresponded to the two clinical groups. Upon IVD, the secretome profiles shifted from that of undifferentiated cells but the difference between the two clinical groups remained yet were muted, suggesting convergence of cytokine profiles after decidualization. LIMITATIONS, REASONS FOR CAUTION Caution is warranted due to the limited sample size of the study and the in vitro nature of the EnSC experiment. Validation on a larger scale is necessary, however, hard to fulfil given the very limited availability of in-cycle proliferative endometrial biopsies outside a RCT setting. WIDER IMPLICATIONS OF THE FINDINGS These data support the hypothesis that the endometrium should be assessed not only during the WOI and that certain endometrial dysfunctionalities can probably be detected early in a cycle by making use of the proliferative phase. This insight opens new horizons for the development of endometrial tests, whether diagnostic or predictive of IVF/ICSI treatment outcome. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by Fonds Wetenschappelijk Onderzoek (FWO, Flanders, Belgium, 11M9415N, 1 524 417N), Wetenschappelijk Fonds Willy Gepts (WFWG G160, Universitair Ziekenhuis Brussel, Belgium) and the National Medicine Research Council (NMRC/CG/M003/2017, Singapore). There are no conflicts of interests. TRIAL REGISTRATION NUMBER NCT02061228.
Collapse
Affiliation(s)
- S Mackens
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - S Santos-Ribeiro
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,IVI-RMA Lisboa, Avenida Infante Dom Henrique 333 H 1-9, 1800-282 Lisbon, Portugal
| | - A Racca
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - D Daneels
- Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - A Koch
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - W Essahib
- Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - W Verpoest
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - C Bourgain
- Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Department of Pathology, Imelda Ziekenhuis Bonheiden, Bonheiden, Belgium
| | - I Van Riet
- Department of Hematology and Immunology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - H Tournaye
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - J J Brosens
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, UK
| | - Y H Lee
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore.,Obstetrics & Gynaecology-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - C Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - H Van de Velde
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
30
|
Huang J, Lu X, Lin J, Chen Q, Gao H, Lyu Q, Cai R, Kuang Y. Association between peak serum estradiol level during controlled ovarian stimulation and neonatal birthweight in freeze-all cycles: a retrospective study of 8501 singleton live births. Hum Reprod 2021; 35:424-433. [PMID: 32078675 DOI: 10.1093/humrep/dez262] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
STUDY QUESTION Is there an association between peak serum estradiol (E2) level during controlled ovarian stimulation (COS) and neonatal birthweight in freeze-all cycles? SUMMARY ANSWER Peak serum E2 level during ovarian stimulation is not associated with neonatal birthweight in freeze-all cycles. WHAT IS KNOWN ALREADY Supraphysiologic E2 levels during COS have been demonstrated to generate a suboptimal peri-implantation endometrial environment and thus lead to adverse neonatal outcomes in fresh embryo transfer cycles. Previous experimental studies also suggested a potential influence of superovulation on oocyte epigenetic programming, but whether it translates into altered phenotypes of fetal growth and development remains unclear in clinical practice. By segmenting the process of COS and embryo transfer, the freeze-all policy provides a novel model to investigate the sole impact of ovarian stimulation on oocytes after ruling out the effects of hyperestrogenic milieu on endometrium in fresh cycles. STUDY DESIGN, SIZE, DURATION A retrospective cohort study of 8501 patients who underwent their first COS cycles with a freeze-all strategy and delivered live-born singletons in subsequent frozen-thawed embryo transfer cycles from January 2007 to December 2016 at a tertiary-care academic medical center. PARTICIPANTS/MATERIALS, SETTING, METHODS Patients were categorized into six groups according to E2 level on trigger day in regular increments of 1000 pg/mL: <1000, 1000-1999, 2000-2999, 3000-3999, 4000-4999 and ≥5000 pg/mL. Univariable and multivariable linear regression and logistic regression analysis were performed to assess the independent association between peak E2 level and measures of neonatal birthweight including absolute birthweight, Z-score, low birthweight (LBW) and small-for-gestational age (SGA). MAIN RESULTS AND THE ROLE OF CHANCE The six groups did not differ significantly in birthweight, Z-score or the incidence of LBW and SGA. Compared with the E2 <1000 pg/mL group, the adjusted mean difference (95% confidence interval [CI]) of stratified higher E2 groups was 17.2 (-31.0-65.5), 12.3 (-35.9-60.5), -4.1 (-51.9-43.7), -0.6 (-48.9-47.8) and -3.6 (-50.0-42.8) g for birthweight, and 0 (-0.11-0.10), 0.02 (-0.08-0.12), 0.04 (-0.06-0.14), -0.01 (-0.11-0.10) and -0.04 (-0.14-0.06) for Z-score, respectively. Regarding the outcomes of LBW and SGA, no increased risks were observed in each E2 category, with the adjusted odds ratio (95% CI) being 1.21 (0.68-2.16), 1.0 (0.58-1.90), 0.90 (0.50-1.63), 0.93 (0.51-1.69) and 1.08 (0.61-1.90) for LBW, and 0.97 (0.58-1.64), 1.06 (0.63-1.77), 0.77 (0.46-1.31), 0.71 (0.41-1.22) and 1.00 (0.60-1.65) for SGA, respectively. LIMITATIONS, REASONS FOR CAUTION The study was retrospective in design, and other unknown confounding factors may not be included for adjustment. Furthermore, the generalization of the study finding could be limited to some extent by the majority of double cleavage-stage embryo transfer and difference in birthweight reference percentiles between Chinese and other populations. WIDER IMPLICATIONS OF THE FINDINGS Our observations suggest that the hyperestrogenic milieu during COS does not seem to pose adverse effects on neonatal birthweight after frozen-thawed embryo transfer, which provides reassuring information for high ovarian responders in freeze-all cycles concerning their offspring's health. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the National Key Research and Development Program of China (SQ2018YFC100163) and National Natural Science Foundation of China (81571397, 81771533). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Jialyu Huang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xuefeng Lu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jiaying Lin
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qiuju Chen
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hongyuan Gao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Renfei Cai
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
31
|
Siristatidis C, Stavros S, Drakeley A, Bettocchi S, Pouliakis A, Drakakis P, Papapanou M, Vlahos N. Omics and Artificial Intelligence to Improve In Vitro Fertilization (IVF) Success: A Proposed Protocol. Diagnostics (Basel) 2021; 11:diagnostics11050743. [PMID: 33919350 PMCID: PMC8143333 DOI: 10.3390/diagnostics11050743] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
The prediction of in vitro fertilization (IVF) outcome is an imperative achievement in assisted reproduction, substantially aiding infertile couples, health systems and communities. To date, the assessment of infertile couples depends on medical/reproductive history, biochemical indications and investigations of the reproductive tract, along with data obtained from previous IVF cycles, if any. Our project aims to develop a novel tool, integrating omics and artificial intelligence, to propose optimal treatment options and enhance treatment success rates. For this purpose, we will proceed with the following: (1) recording subfertile couples’ lifestyle and demographic parameters and previous IVF cycle characteristics; (2) measurement and evaluation of metabolomics, transcriptomics and biomarkers, and deep machine learning assessment of the oocyte, sperm and embryo; (3) creation of artificial neural network models to increase objectivity and accuracy in comparison to traditional techniques for the improvement of the success rates of IVF cycles following an IVF failure. Therefore, “omics” data are a valuable parameter for embryo selection optimization and promoting personalized IVF treatment. “Omics” combined with predictive models will substantially promote health management individualization; contribute to the successful treatment of infertile couples, particularly those with unexplained infertility or repeated implantation failures; and reduce multiple gestation rates.
Collapse
Affiliation(s)
- Charalampos Siristatidis
- Second Department of Obstetrics and Gynecology, “Aretaieion Hospital”, Medical School, National and Kapodistrian University of Athens, Vas. Sofias 76, 11528 Athens, Greece; (M.P.); (N.V.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, “Aretaieion Hospital”, Medical School, National and Kapodistrian University of Athens, Vas. Sofias 76, 11528 Athens, Greece
- Correspondence: ; Tel.: +30-69-3229-4994
| | - Sofoklis Stavros
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra Hospital, 80 Vas. Sofias Av. and Lourou str., 11528 Athens, Greece; (S.S.); (P.D.)
| | - Andrew Drakeley
- Hewitt Fertility Centre, Liverpool Women’s NHS Foundation Trust, Crown Street, Liverpool L8 7SS, UK;
| | - Stefano Bettocchi
- Second Unit of Obstetrics and Gynecology, Department of Biomedical and Human Oncologic Science, Policlinico University of Bari, 70124 Bari, Italy;
| | - Abraham Pouliakis
- Second Department of Pathology, National and Kapodistrian University of Athens, “Attikon” University Hospital, Rimini 1, Chaidari, 12642 Athens, Greece;
| | - Peter Drakakis
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra Hospital, 80 Vas. Sofias Av. and Lourou str., 11528 Athens, Greece; (S.S.); (P.D.)
| | - Michail Papapanou
- Second Department of Obstetrics and Gynecology, “Aretaieion Hospital”, Medical School, National and Kapodistrian University of Athens, Vas. Sofias 76, 11528 Athens, Greece; (M.P.); (N.V.)
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynecology, “Aretaieion Hospital”, Medical School, National and Kapodistrian University of Athens, Vas. Sofias 76, 11528 Athens, Greece; (M.P.); (N.V.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, “Aretaieion Hospital”, Medical School, National and Kapodistrian University of Athens, Vas. Sofias 76, 11528 Athens, Greece
| |
Collapse
|
32
|
Wang CH, Hu XQ. A systematic review of clinical efficacy of frozen-thawed embryos and fresh embryos in in-vitro fertilization cycles. Cryobiology 2021; 100:19-25. [PMID: 33872610 DOI: 10.1016/j.cryobiol.2021.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Frozen-thawed embryo (FTE) and fresh embryo (FE) transfer are two common strategies in vitro fertilization (IVF), while the results and findings still vary among studies. METHODS We searched multiple databases for relevant studies comparing the clinical effects of FTE and FE. Meta-analyses were conducted with Review Manager 5.0 to assess the efficacy among included articles. We also analyzed the risk of bias for the reports. RESULTS Nine studies eventually met the inclusion criteria from 2010 to 2018, and 11396 patients were included. The meta-analyses indicated no significant difference in biochemical pregnancy, clinical pregnancy and ongoing pregnancy rates. Meanwhile, the implantation rate and live birth rate in frozen-thawed embryos were much higher than those of fresh embryo. The birth weight in the frozen-thawed ET group was greater than that of the FE group, and the low birth weight rate in FTE was lower than FE group. CONCLUSION Our findings suggested a trend toward favouring frozen-thawed FTE might be a preferred transfer strategy for patients with IVF.
Collapse
Affiliation(s)
- Cui-Hua Wang
- Department of Gynecology and Obstetrics, Wuxi No.2 Chinese Medicine Hospital, Wuxi, China
| | - Xiao-Qing Hu
- Department of Gynecology and Obstetrics, Wuxi No.2 Chinese Medicine Hospital, Wuxi, China.
| |
Collapse
|
33
|
Luteal Support with very Low Daily Dose of Human Chorionic Gonadotropin after Fresh Embryo Transfer as an Alternative to Cycle Segmentation for High Responders Patients Undergoing Gonadotropin-Releasing Hormone Agonist-Triggered IVF. Pharmaceuticals (Basel) 2021; 14:ph14030228. [PMID: 33800021 PMCID: PMC7998839 DOI: 10.3390/ph14030228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
The segmentation of the in vitro fertilization (IVF) cycle, consisting of the freezing of all embryos and the postponement of embryo transfer (ET), has become popular in recent years, with the main purpose of preventing ovarian hyperstimulation syndrome (OHSS) in patients with high response to controlled ovarian stimulation (COS). Indeed cycle segmentation (CS), especially when coupled to a GnRH-agonist trigger, was shown to reduce the incidence of OHSS in high-risk patients. However, CS increases the economic costs and the work amount for IVF laboratories. An alternative strategy is to perform a fresh ET in association with intensive luteal phase pharmacological support, able to overcome the negative effects of the GnRH-agonist trigger on the luteal phase and on endometrial receptivity. In order to compare these two strategies, we performed a retrospective, real-life cohort study including 240 non-polycystic ovarian syndrome (PCO) women with expected high responsiveness to COS (AMH >2.5 ng/mL), who received either fresh ET plus 100 IU daily human chorionic gonadotropin (hCG) as luteal support (FRESH group, n = 133), or cycle segmentation with freezing of all embryos and postponed ET (CS group, n = 107). The primary outcomes were: implantation rate (IR), live birth rate (LBR) after the first ET, and incidence of OHSS. Overall, significantly higher IR and LBR were observed in the CS group than in the FRESH group (42.9% vs. 27.8%, p < 0.05 and 32.7% vs. 19.5%, p < 0.05, respectively); the superiority of CS strategy was particularly evident when 16-19 oocytes were retrieved (LBR 42.2% vs. 9.5%, p = 0.01). Mild OHSS appeared with the same incidence in the two groups, whereas moderate and severe OHSS forms were observed only in the FRESH group (1.5% and 0.8%, respectively). In conclusion, in non-PCO women, high responders submitted to COS with the GnRH-antagonist protocol and GnRH-agonist trigger, CS strategy was associated with higher IR and LBR than the strategy including fresh ET followed by luteal phase support with a low daily hCG dose. CS appears to be advisable, especially when >15 oocytes are retrieved.
Collapse
|
34
|
Time from oocyte retrieval to frozen embryo transfer in the natural cycle does not impact reproductive or neonatal outcomes. Fertil Steril 2021; 115:1232-1238. [PMID: 33589140 DOI: 10.1016/j.fertnstert.2020.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/18/2020] [Accepted: 11/05/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To determine if the time from oocyte retrieval to frozen embryo transfer (FET) in the natural cycle affects reproductive or neonatal outcomes. DESIGN Retrospective cohort. SETTING Not applicable. PATIENT(S) Five hundred and seventy-six consecutive freeze-all cycles from January 2011 to December 2018 followed by natural cycle FET of a single blastocyst. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Primary outcome of live birth; secondary outcomes of preterm delivery (24-37 weeks) and small for gestational age (SGA) with a multivariable logistic regression performed with adjustment for age, infertility diagnosis, ovulatory trigger type, and preimplantation genetic testing (PGT). RESULT(S) Before adjustment for confounding, we found a statistically significantly different live-birth rate (57.7% vs. 48.6%) for natural cycle FET occurring in the first versus second menstrual cycle, respectively. In a multivariate analysis, performing a natural cycle FET of a single blastocyst in the second compared with the first menstrual cycle did not statistically significantly impact the odds of live-birth rate. After adjustment for age, diagnosis, and ovulatory trigger type, only PGT was associated with statistically significantly increased odds of live birth compared with no PGT. There were no differences in the incidence of SGA (male, 6.6% vs. 2.3%; female, 9.8% vs. 11.1%) or preterm delivery (1.6% vs. 5.6%) between both groups. CONCLUSION(S) Performing a natural cycle FET of a single blastocyst in the second compared with the first menstrual cycle after ovarian stimulation did not statistically significantly impact the odds of live birth or neonatal outcomes.
Collapse
|
35
|
Zaat T, Zagers M, Mol F, Goddijn M, van Wely M, Mastenbroek S. Fresh versus frozen embryo transfers in assisted reproduction. Cochrane Database Syst Rev 2021; 2:CD011184. [PMID: 33539543 PMCID: PMC8095009 DOI: 10.1002/14651858.cd011184.pub3] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND In vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI) treatments conventionally consist of a fresh embryo transfer, possibly followed by one or more cryopreserved embryo transfers in subsequent cycles. An alternative option is to freeze all suitable embryos and transfer cryopreserved embryos in subsequent cycles only, which is known as the 'freeze all' strategy. This is the first update of the Cochrane Review on this comparison. OBJECTIVES To evaluate the effectiveness and safety of the freeze all strategy compared to the conventional IVF/ICSI strategy in women undergoing assisted reproductive technology. SEARCH METHODS We searched the Cochrane Gynaecology and Fertility Group Trials Register, CENTRAL, MEDLINE, Embase, PsycINFO, CINAHL, and two registers of ongoing trials from inception until 23 September 2020 for relevant studies, checked references of publications found, and contacted study authors to obtain additional data. SELECTION CRITERIA Two review authors (TZ and MZ) independently selected studies for inclusion, assessed risk of bias, and extracted study data. We included randomised controlled trials comparing a 'freeze all' strategy with a conventional IVF/ICSI strategy including a fresh embryo transfer in women undergoing IVF or ICSI treatment. DATA COLLECTION AND ANALYSIS The primary outcomes were cumulative live birth rate and ovarian hyperstimulation syndrome (OHSS). Secondary outcomes included effectiveness outcomes (including ongoing pregnancy rate and clinical pregnancy rate), time to pregnancy and obstetric, perinatal and neonatal outcomes. MAIN RESULTS We included 15 studies in the systematic review and eight studies with a total of 4712 women in the meta-analysis. The overall evidence was of moderate to low quality. We graded all the outcomes and downgraded due to serious risk of bias, serious imprecision and serious unexplained heterogeneity. Risk of bias was associated with unclear blinding of investigators for preliminary outcomes of the study during the interim analysis, unit of analysis error, and absence of adequate study termination rules. There was an absence of high-quality evidence according to GRADE assessments for our primary outcomes, which is reflected in the cautious language below. There is probably little or no difference in cumulative live birth rate between the 'freeze all' strategy and the conventional IVF/ICSI strategy (odds ratio (OR) 1.08, 95% CI 0.95 to 1.22; I2 = 0%; 8 RCTs, 4712 women; moderate-quality evidence). This suggests that for a cumulative live birth rate of 58% following the conventional strategy, the cumulative live birth rate following the 'freeze all' strategy would be between 57% and 63%. Women might develop less OHSS after the 'freeze all' strategy compared to the conventional IVF/ICSI strategy (OR 0.26, 95% CI 0.17 to 0.39; I2 = 0%; 6 RCTs, 4478 women; low-quality evidence). These data suggest that for an OHSS rate of 3% following the conventional strategy, the rate following the 'freeze all' strategy would be 1%. There is probably little or no difference between the two strategies in the cumulative ongoing pregnancy rate (OR 0.95, 95% CI 0.75 to 1.19; I2 = 31%; 4 RCTs, 1245 women; moderate-quality evidence). We could not analyse time to pregnancy; by design, time to pregnancy is shorter in the conventional strategy than in the 'freeze all' strategy when the cumulative live birth rate is comparable, as embryo transfer is delayed in a 'freeze all' strategy. We are uncertain whether the two strategies differ in cumulative miscarriage rate because the evidence is very low quality (Peto OR 1.06, 95% CI 0.72 to 1.55; I2 = 55%; 2 RCTs, 986 women; very low-quality evidence) and cumulative multiple-pregnancy rate (Peto OR 0.88, 95% CI 0.61 to 1.25; I2 = 63%; 2 RCTs, 986 women; very low-quality evidence). The risk of hypertensive disorders of pregnancy (Peto OR 2.15, 95% CI 1.42 to 3.25; I2 = 29%; 3 RCTs, 3940 women; low-quality evidence), having a large-for-gestational-age baby (Peto OR 1.96, 95% CI 1.51 to 2.55; I2 = 0%; 3 RCTs, 3940 women; low-quality evidence) and a higher birth weight of the children born (mean difference (MD) 127 g, 95% CI 77.1 to 177.8; I2 = 0%; 5 RCTs, 1607 singletons; moderate-quality evidence) may be increased following the 'freeze all' strategy. We are uncertain whether the two strategies differ in the risk of having a small-for-gestational-age baby because the evidence is low quality (Peto OR 0.82, 95% CI 0.65 to 1.05; I2 = 64%; 3 RCTs, 3940 women; low-quality evidence). AUTHORS' CONCLUSIONS We found moderate-quality evidence showing that one strategy is probably not superior to the other in terms of cumulative live birth rate and ongoing pregnancy rate. The risk of OHSS may be decreased in the 'freeze all' strategy. Based on the results of the included studies, we could not analyse time to pregnancy. It is likely to be shorter using a conventional IVF/ICSI strategy with fresh embryo transfer in the case of similar cumulative live birth rate, as embryo transfer is delayed in a 'freeze all' strategy. The risk of maternal hypertensive disorders of pregnancy, of having a large-for-gestational-age baby and a higher birth weight of the children born may be increased following the 'freeze all' strategy. We are uncertain if 'freeze all' strategy reduces the risk of miscarriage, multiple pregnancy rate or having a small-for-gestational-age baby compared to conventional IVF/ICSI.
Collapse
Affiliation(s)
- Tjitske Zaat
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| | - Miriam Zagers
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| | - Femke Mol
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| | - Mariëtte Goddijn
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| | - Madelon van Wely
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| | - Sebastiaan Mastenbroek
- Amsterdam UMC, University of Amsterdam, Center for Reproductive Medicine, Amsterdam Reproduction & Development Research Institute, Amsterdam, Netherlands
| |
Collapse
|
36
|
Haouzi D, Entezami F, Torre A, Innocenti C, Antoine Y, Mauries C, Vincens C, Bringer-Deutsch S, Gala A, Ferrieres-Hoa A, Ohl J, Gonzalez Marti B, Brouillet S, Hamamah S. Customized Frozen Embryo Transfer after Identification of the Receptivity Window with a Transcriptomic Approach Improves the Implantation and Live Birth Rates in Patients with Repeated Implantation Failure. Reprod Sci 2021; 28:69-78. [PMID: 32725589 PMCID: PMC7782404 DOI: 10.1007/s43032-020-00252-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/04/2020] [Accepted: 06/30/2020] [Indexed: 01/10/2023]
Abstract
The aim of this prospective study was to evaluate outcome benefits expected in repeated implantation failure (RIF) patients (n = 217) after customized embryo transfer based upon identification of the receptivity window by transcriptomic approach using the Win-Test. In this test, the expression of 11 endometrial genes known to be predictive of endometrial receptivity is assessed by RT-PCR in biopsies collected during the implantation window (6-9 days after the spontaneous luteinizing hormone surge during natural cycles, 5-9 days after progesterone administration during hormone replacement therapy cycles). Then, patients underwent either customized embryo transfer (cET, n = 157 patients) according to the Win-Test results or embryo transfer according to the classical procedure (control group, n = 60). Pregnancy and live birth rates were compared in the two groups. The Win-Test showed that in 78.5% of women, the receptivity window lasted less than 48 h, although it could be shorter (< 24 h, 9.5%) or longer (> 48 h, 12%). This highlighted that only in 20% of patients with RIF the endometrium would have been receptive if the classical embryo transfer protocol was followed. In the other 80% of patients, the receptivity window was delayed by 1-3 days relative to the classical timing. This suggests that implantation failure could be linked to inadequate timing of embryo transfer. In agreement, both implantation (22.7% vs. 7.2%) and live birth rates per patient (31.8% vs. 8.3%) were significantly higher in the cET group than in the control group. cET on the basis of the Win-Test results could be proposed to improve pregnancy and live birth rates.ClinicalTrials.gov ID: NCT04192396; December 5, 2019, retrospectively registered.
Collapse
Affiliation(s)
- Delphine Haouzi
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France.
- IRMB, Univ Montpellier, INSERM, Montpellier, France.
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France.
| | - Frida Entezami
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- ART Department, American Hospital of Paris, Neuilly-Sur-Seine, France
- Laboratoire Eylau-UNILABS-La Muette, Clinique de La Muette-Ramsay-Générale de Santé, Paris, France
| | - Antoine Torre
- Division of Child Health, Obstetrics & Gynaecology Department, University of Nottingham, Nottingham, UK
| | - Charlène Innocenti
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| | - Yannick Antoine
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | - Charlotte Mauries
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| | - Claire Vincens
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| | | | - Anna Gala
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| | - Alice Ferrieres-Hoa
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| | - Jeanine Ohl
- Department of reproductive medicine, CMCO, Schiltigheim, France
| | - Beatriz Gonzalez Marti
- ART Department, American Hospital of Paris, Neuilly-Sur-Seine, France
- Laboratoire Eylau-UNILABS-La Muette, Clinique de La Muette-Ramsay-Générale de Santé, Paris, France
| | - Sophie Brouillet
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
- Univ Grenoble-Alpes, INSERM U1036, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
| | - Samir Hamamah
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PGD Department, Arnaud de Villeneuve Hospital, Montpellier, France
| |
Collapse
|
37
|
Li T, Greenblatt EM, Shin ME, Brown TJ, Chan C. Cargo small non-coding RNAs of extracellular vesicles isolated from uterine fluid associate with endometrial receptivity and implantation success. Fertil Steril 2020; 115:1327-1336. [PMID: 33272614 DOI: 10.1016/j.fertnstert.2020.10.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/04/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To optimize a method of isolating extracellular vesicles (EVs) from uterine fluid and to characterize small non-coding RNAs (sncRNAs) from the EVs, with the goal of identifying novel receptivity-associated biomarkers. DESIGN Longitudinal study comparing sncRNA expression profiles from endometrial EVs. SETTING University-affiliated, hospital-based fertility clinic. PATIENT(S) Healthy volunteers with no history of infertility (Group A) and women receiving controlled ovarian stimulation (COS)-in vitro fertilization treatment (Group B). INTERVENTIONS(S) In Group A, EVs were isolated from uterine fluid obtained on luteinizing hormone (LH)+2 and LH+7 in one natural menstrual cycle. In Group B, EVs were isolated from uterine fluid obtained on human chorionic gonadotropin (hCG)+2 and hCG+7 in one COS cycle. RNAs extracted from EVs were profiled using next-generation sequencing. MAIN OUTCOME MEASURE(S) Differential EV-sncRNAs between LH+2 and LH+7 (Group A), between hCG+2 and hCG+7 (Group B), and between pregnant and nonpregnant in vitro fertilization cycles (Group B). RESULT(S) Ultracentrifugation was validated as the most efficient method to isolate EVs from uterine fluid. We identified 12 endometrial EV-sncRNAs (11 microRNAs and 1 piwi-interacting RNA) as receptivity-associated transcripts conserved in both natural and COS cycles. These sncRNAs were associated strongly with biological functions related to immune response, extracellular matrix, and cell junction. Within COS cycles, we also identified a group of EV-sncRNAs that exhibited differential expression in patients who conceived versus those who did not, with hsa-miR-362-3p most robustly overexpressed in the nonpregnant patients. CONCLUSION(S) This study is the first to profile comprehensively sncRNAs in endometrial EVs from uterine fluid and identify sncRNA biomarkers of endometrial receptivity and implantation success.
Collapse
Affiliation(s)
- Tiantian Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Ellen M Greenblatt
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada; Mount Sinai Fertility, Sinai Health System, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | | | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Crystal Chan
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada; Mount Sinai Fertility, Sinai Health System, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Hernández-Vargas P, Muñoz M, Domínguez F. Identifying biomarkers for predicting successful embryo implantation: applying single to multi-OMICs to improve reproductive outcomes. Hum Reprod Update 2020; 26:264-301. [PMID: 32096829 DOI: 10.1093/humupd/dmz042] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/08/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Successful embryo implantation is a complex process that requires the coordination of a series of events, involving both the embryo and the maternal endometrium. Key to this process is the intricate cascade of molecular mechanisms regulated by endocrine, paracrine and autocrine modulators of embryonic and maternal origin. Despite significant progress in ART, implantation failure still affects numerous infertile couples worldwide and fewer than 10% of embryos successfully implant. Improved selection of both the viable embryos and the optimal endometrial phenotype for transfer remains crucial to enhancing implantation chances. However, both classical morphological embryo selection and new strategies incorporated into clinical practice, such as embryonic genetic analysis, morphokinetics or ultrasound endometrial dating, remain insufficient to predict successful implantation. Additionally, no techniques are widely applied to analyse molecular signals involved in the embryo-uterine interaction. More reliable biological markers to predict embryo and uterine reproductive competence are needed to improve pregnancy outcomes. Recent years have seen a trend towards 'omics' methods, which enable the assessment of complete endometrial and embryonic molecular profiles during implantation. Omics have advanced our knowledge of the implantation process, identifying potential but rarely implemented biomarkers of successful implantation. OBJECTIVE AND RATIONALE Differences between the findings of published omics studies, and perhaps because embryonic and endometrial molecular signatures were often not investigated jointly, have prevented firm conclusions being reached. A timely review summarizing omics studies on the molecular determinants of human implantation in both the embryo and the endometrium will help facilitate integrative and reliable omics approaches to enhance ART outcomes. SEARCH METHODS In order to provide a comprehensive review of the literature published up to September 2019, Medline databases were searched using keywords pertaining to omics, including 'transcriptome', 'proteome', 'secretome', 'metabolome' and 'expression profiles', combined with terms related to implantation, such as 'endometrial receptivity', 'embryo viability' and 'embryo implantation'. No language restrictions were imposed. References from articles were also used for additional literature. OUTCOMES Here we provide a complete summary of the major achievements in human implantation research supplied by omics approaches, highlighting their potential to improve reproductive outcomes while fully elucidating the implantation mechanism. The review highlights the existence of discrepancies among the postulated biomarkers from studies on embryo viability or endometrial receptivity, even using the same omic analysis. WIDER IMPLICATIONS Despite the huge amount of biomarker information provided by omics, we still do not have enough evidence to link data from all omics with an implantation outcome. However, in the foreseeable future, application of minimally or non-invasive omics tools, together with a more integrative interpretation of uniformly collected data, will help to overcome the difficulties for clinical implementation of omics tools. Omics assays of the embryo and endometrium are being proposed or already being used as diagnostic tools for personalised single-embryo transfer in the most favourable endometrial environment, avoiding the risk of multiple pregnancies and ensuring better pregnancy rates.
Collapse
Affiliation(s)
- Purificación Hernández-Vargas
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Manuel Muñoz
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Francisco Domínguez
- Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| |
Collapse
|
39
|
The freeze-all strategy after IVF: which indications? Reprod Biomed Online 2020; 42:529-545. [PMID: 33384269 DOI: 10.1016/j.rbmo.2020.11.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/09/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022]
Abstract
The freeze-all strategy is gaining popularity worldwide as an alternative to the conventional fresh embryo transfer. It consists of cryopreservation of the entire embryo cohort and the embryo transfer in a subsequent cycle that takes place separately from ovarian stimulation. The freeze-all strategy was initially a 'rescue' strategy for women at high risk of ovarian hyperstimulation syndrome; however, this approach has been extended to other indications as a scheduled strategy to improve implantation rates. This assumes that ovarian stimulation can alter endometrial receptivity in fresh cycles owing to the effect of supraphysiological levels of steroids on endometrial maturation. The procedure, however, has not been associated with increased live birth rates in all infertile couples, and concerns have been raised about the occurrence of several adverse perinatal outcomes. It is, therefore, crucial to identify in which subgroups of patients a freeze-all strategy could be beneficial. The aim of this review is to summarize current scientific research in this field to highlight potential indications for this strategy and to guide clinicians in their daily practice.
Collapse
|
40
|
Jwa SC, Seto S, Takamura M, Kuwahara A, Kajihara T, Ishihara O. Ovarian stimulation increases the risk of ectopic pregnancy for fresh embryo transfers: an analysis of 68,851 clinical pregnancies from the Japanese Assisted Reproductive Technology registry. Fertil Steril 2020; 114:1198-1206. [PMID: 33081977 DOI: 10.1016/j.fertnstert.2020.06.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the risk of ectopic pregnancies (EPs) for fresh cycles according to different ovarian stimulation protocols. DESIGN Registry-based retrospective cohort study. SETTING Not applicable. PATIENT(S) A total of 68,851 clinical pregnancies after fresh single embryo transfer between 2007 and 2015. INTERVENTION (S) None MAIN OUTCOME MEASURE(S): Ectopic pregnancies. Odds ratios and 95% confidence intervals for EPs were calculated by using generalized estimating equations adjusted for potential maternal and treatment characteristics. RESULT(S) Among 68,851 clinical pregnancies, 1,049 (1.46%) cases of EP were reported. Compared with natural cycles, all ovarian stimulation protocols were associated with a significantly increased risk of EP. Ovarian stimulation with clomiphene (CC) demonstrated the highest odds ratios for EPs. Significant associations between ovarian stimulation protocols and EP compared with natural cycles were prominent when the number of retrieved oocytes was low (1-3) to moderate (4-7), but there were no significant associations when the number of retrieved oocytes was high (≥8). CONCLUSION(S) Ovarian stimulation protocols were significantly associated with an increased risk of EP. In particular, ovarian stimulation with CC had the highest risk of EP compared with other stimulation protocols. Further studies are essential to investigate possible confounding factors for different ovarian stimulation protocols, especially CC, and the risk of EP.
Collapse
Affiliation(s)
- Seung Chik Jwa
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan.
| | - Sachie Seto
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan
| | - Masashi Takamura
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan
| | - Akira Kuwahara
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takeshi Kajihara
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan
| | - Osamu Ishihara
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan
| |
Collapse
|
41
|
Hviid Saxtorph M, Persson G, Hallager T, Birch Petersen K, Eriksen JO, Larsen LG, Macklon N, Hviid TVF. Are different markers of endometrial receptivity telling us different things about endometrial function? Am J Reprod Immunol 2020; 84:e13323. [PMID: 33245608 DOI: 10.1111/aji.13323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/23/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022] Open
Abstract
PROBLEM To what extent do endocrine, immunological, gene expression and histological markers of endometrial receptivity correlate? METHOD OF STUDY Between November 2017 and September 2019, 121 women referred to a University Hospitals Fertility Clinic consented to inclusion in this cohort study. The women underwent timed endometrial biopsy followed by blood samples in a hormone-substituted cycle. Of these, 37 women had just started IVF treatment, and the remaining 84 had experienced recurrent implantation failure following IVF/ICSI. The hormone-substituted cycle consisted of initiation with oral oestradiol followed by addition of vaginal progesterone treatment for five full days. Endometrial biopsies were subject to histological examination, immune cell markers by immunohistochemistry (CD56+ , CD16+ , CD163+ , FoxP3) and gene expression microarray analyses with the endometrial receptivity array (ERA® ) test (Igenomix). Plasma progesterone and oestradiol were measured on the day of biopsy. RESULTS CD56+ uterine natural killer (uNK) cell counts correlate with transcriptional markers of endometrial receptivity assessed by the ERA test. Endometrial maturation, receptivity and immunological markers were not correlated with mid-luteal blood plasma progesterone level. Mid-luteal serum oestradiol level correlated with markers of endometrial maturation and receptivity. The tests were carried out during a standard hormone substitution cycle, and the findings may not apply in the natural cycle. CONCLUSION CD56+ uNK cell counts and endometrial receptivity assessed by the ERA test appear to be linked. Mid-luteal progesterone levels were not correlated to the tested markers of endometrial receptivity. In contrast, mid-luteal oestradiol level was inversely related to markers of endometrial receptivity and maturation.
Collapse
Affiliation(s)
- Malene Hviid Saxtorph
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde, Denmark.,ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gry Persson
- ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Biochemistry, Centre for Immune Regulation and Reproductive Immunology, Zealand University Hospital, Roskilde, Denmark
| | - Trine Hallager
- ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Kathrine Birch Petersen
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde, Denmark.,ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Stork Fertility Clinic, VivaNeo/The Fertility Partnership, Roskilde, Denmark
| | - Jens O Eriksen
- ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Lise Grupe Larsen
- ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Nick Macklon
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde, Denmark.,ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,London Womens Clinic, London, UK
| | - Thomas Vauvert F Hviid
- ReproHealth Research Consortium, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Centre for Immune Regulation and Reproductive Immunology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
42
|
Lattes K, López S, Checa MA, Brassesco M, García D, Vassena R. A freeze-all strategy does not increase live birth rates in women of advanced reproductive age. J Assist Reprod Genet 2020; 37:2443-2451. [PMID: 32876800 DOI: 10.1007/s10815-020-01934-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/25/2020] [Indexed: 11/29/2022] Open
Abstract
RESEARCH QUESTION Does a freeze-all strategy improve live birth rates in women of different age groups? DESIGN Retrospective cohort study of 1882 first embryo transfer cycles, performed between January 2013 and December 2015. Reproductive outcomes between fresh (FRESH) or frozen (FROZEN) embryo transfers were compared in patients stratified by age: < 35, between 35 and 38, or > 38 years. Student's t test for independent samples and χ2 analyses were used as needed. A multivariable logistic regression analysis was performed adjusting for age, triggering drug, number of retrieved oocytes, number of transferred embryos, and percentage of top-quality embryos. MAIN RESULTS AND THE ROLE OF CHANCE Live birth rates (LBR) were significantly higher for FROZEN in the < 35 years group (43.7% vs 24%; p < 0.001). In both the 35-38 and > 38 years groups, LBR for FROZEN vs FRESH were not statistically different (30.9% in the FROZEN group vs 29.3% in the FRESH group, p = 0.70, and 19.8% in the FROZEN group vs 12.7% in the FRESH group, p = 0.07, respectively). The multivariate analysis found a significantly positive effect of performing FROZEN on LBR in the younger group (OR 2.46, 95% CI 1.31-4.62; p = 0.005) but had no impact in women between 35 and 38 years (OR 1.01, 95% CI 0.55-1.83; p = 0.98) or older (OR 0.96, 95% CI 0.43-2.13; p = 0.92). CONCLUSIONS Performing a freeze-all strategy seems to result in better reproductive outcomes when compared with a fresh ET in women under 35 years, with no significant impact on older women.
Collapse
Affiliation(s)
- K Lattes
- Centro de Infertilidad y Reproducción Humana (CIRH), 08017, Barcelona, Spain
| | - S López
- Centro de Infertilidad y Reproducción Humana (CIRH), 08017, Barcelona, Spain
| | - M A Checa
- Department of Obstetrics and Gynecology, Parc de Salut Mar, Universitat Autònoma de Barcelona, 08003, Barcelona, Spain.,Barcelona Infertility Research Group (GRI-BCN), 08005, Barcelona, Spain
| | - M Brassesco
- Centro de Infertilidad y Reproducción Humana (CIRH), 08017, Barcelona, Spain
| | | | | |
Collapse
|
43
|
Drissennek L, Baron C, Brouillet S, Entezami F, Hamamah S, Haouzi D. Endometrial miRNome profile according to the receptivity status and implantation failure. HUM FERTIL 2020; 25:356-368. [DOI: 10.1080/14647273.2020.1807065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Loubna Drissennek
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | - Chloé Baron
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | - Sophie Brouillet
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PDG department, Arnaud de Villeneuve Hospital, Montpellier, Montpellier, France
- Univ Grenoble-Alpes, INSERM 1036, Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l’Infection (BCI), Grenoble, France
| | - Frida Entezami
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- American Hospital of Paris, IVF department, Neuilly-Sur-Seine, France
| | - Samir Hamamah
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PDG department, Arnaud de Villeneuve Hospital, Montpellier, Montpellier, France
| | - Delphine Haouzi
- Univ Montpellier, INSERM U1203, EmbryoPluripotency, Montpellier, France
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- CHU Montpellier, ART/PDG department, Arnaud de Villeneuve Hospital, Montpellier, Montpellier, France
| |
Collapse
|
44
|
Marin L, Vitagliano A. Whether immediate frozen-thawed embryo transfer improves IVF outcome in non-elective freeze all policy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:979. [PMID: 32953779 PMCID: PMC7475503 DOI: 10.21037/atm-2020-97] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Loris Marin
- Department of Women's and Children's Health, University of Padua, Salus Pueri, Padova, Italy
| | - Amerigo Vitagliano
- Department of Women's and Children's Health, University of Padua, Salus Pueri, Padova, Italy
| |
Collapse
|
45
|
Bosdou JK, Venetis CA, Tarlatzis BC, Grimbizis GF, Kolibianakis EM. Higher probability of live-birth in high, but not normal, responders after first frozen-embryo transfer in a freeze-only cycle strategy compared to fresh-embryo transfer: a meta-analysis. Hum Reprod 2020; 34:491-505. [PMID: 30689865 DOI: 10.1093/humrep/dey388] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/31/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Does the outcome of the comparison of live birth rates between the first frozen embryo transfer (ET) (in a freeze-only cycles strategy, i.e. frozen ET group) and a fresh embryo transfer (fresh ET group) differ considering the type of ovarian response? SUMMARY ANSWER Α significantly higher probability of live birth is present in high, but not normal, responders, after the first frozen ET in a freeze-only cycle strategy as compared to a fresh ET. WHAT IS KNOWN ALREADY It has been hypothesised that freezing all good embryos in a fresh in-vitro fertilisation (IVF) cycle and deferring embryo transfer in subsequent cycles may provide a more physiological endometrial environment for embryo implantation when compared to a fresh ET. However, currently, three relevant meta-analyses have been published with conflicting results, while none of them has taken into consideration the type of ovarian response. Recently, the publication of additional, large relevant randomised controlled trials (RCTs) in patients with different types of ovarian response makes possible the comparative evaluation of the first frozen ET (in a freeze-only cycle strategy) versus fresh ET, considering the type of ovarian response. STUDY DESIGN, SIZE, DURATION A systematic review and meta-analysis was performed aiming to identify RCTs comparing the first frozen ET (in a freeze-only cycle strategy) to a fresh ET. The main outcome was live birth, while secondary outcomes included ongoing pregnancy, clinical pregnancy, moderate/severe ovarian hyperstimulation syndrome (OHSS) and miscarriage. PARTICIPANTS/MATERIALS, SETTING, METHODS We identified eight eligible RCTs, including 5265 patients, which evaluated the first frozen ET in a freeze-only cycle strategy versus a fresh ET either in high responders (n = 4) or in normal responders (n = 4). No relevant RCTs were present in poor responders. Meta-analysis of weighted data using fixed and random effects model was performed. Results are reported as relative risk (RR) with 95% confidence interval (CI). MAIN RESULTS AND THE ROLE OF CHANCE Eligible RCTs were published between 2011 and 2018. Four RCTs (n = 3255 patients) compared the first frozen ET (in a freeze-only cycle strategy) to a fresh ET in normal responders and four RCTs (n = 2010 patients) did the comparison in high responders. In high responders, a significantly higher probability of live birth was observed in the frozen ET group when compared with the fresh ET group (RR: 1.18, 95% CI: 1.06-1.31; fixed effects model; heterogeneity: I2 = 0%; three studies; n = 3398 patients). However the probability of live birth was not significantly different between the frozen ET group and the fresh ET group in normal responders (RR: 1.13, 95% CI: 0.90-1.41; random effects model; heterogeneity: I2 = 77%; three studies; n = 1608 patients). The risk of moderate/severe OHSS was significantly lower in the frozen ET group when compared with the fresh ET group both in high (RR: 0.19, 95% CI: 0.10-0.37; fixed effects model; heterogeneity: not applicable; a single study; n = 1508 patients) and normal responders (RR: 0.39, 95% CI: 0.19-0.80; fixed effects model; heterogeneity: I2 = 0%; two studies; n = 2939 patients). LIMITATIONS, REASONS FOR CAUTION Considerable heterogeneity was present among the studies, regarding ovarian stimulation protocols and the triggering signal used for inducing final oocyte maturation as well as the cryopreservation methods, while the quality of evidence was poor for the live birth rate in high responders. Moreover, the analysis did not apply a standard for determining 'high' or 'normal' responders since the type of ovarian response followed the characterisation of populations as reported by the authors of the eligible studies. WIDER IMPLICATIONS OF THE FINDINGS A freeze-only cycle strategy should be the preferred option in high responders since it enhances the probability of live birth, while reducing the chance of moderate/severe OHSS. In normal responders, the same strategy could be applied, in the interest of patient safety or clinic convenience, without compromising the chances of live birth. STUDY FUNDING/COMPETING INTEREST(S) No external funding was used and there were no competing interests. PROSPERO REGISTRATION NUMBER PROSPERO registration number: CRD42018099389.
Collapse
Affiliation(s)
- J K Bosdou
- Aristotle University of Thessaloniki, Medical School, Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Thessaloniki, Greece
| | - C A Venetis
- University of New South Wales, Centre for Big Data Research in Health & School of Women's and Children's Health, UNSW Medicine, Sydney, Australia
| | - B C Tarlatzis
- Aristotle University of Thessaloniki, Medical School, Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Thessaloniki, Greece
| | - G F Grimbizis
- Aristotle University of Thessaloniki, Medical School, Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Thessaloniki, Greece
| | - E M Kolibianakis
- Aristotle University of Thessaloniki, Medical School, Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Thessaloniki, Greece
| |
Collapse
|
46
|
Wang F, Liu Y. Identification of key genes, regulatory factors, and drug target genes of recurrent implantation failure (RIF). Gynecol Endocrinol 2020; 36:448-455. [PMID: 31646911 DOI: 10.1080/09513590.2019.1680622] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective: Recurrent implantation failure (RIF) exacerbates the physical trauma of infertile women that undergone in vitro fertilization-embryo transfer (IVF-ET). We aimed to identify the key genes, regulatory factors, and drug target genes involved in the RIF.Methods: The dataset GSE58144 that obtained from the Gene Expression Omnibus mainly contained 43 RIF and 72 control endometrial samples. Differently expressed genes (DEGs) between RIF and control groups were firstly analyzed, followed by the pathway and Gene Ontology (GO) enrichment analysis. Then, protein-protein interaction (PPI) network and miRNA-transcript factor (TF)-DEGs network were established. Finally, a drug-target interaction network was constructed.Results: A total of 399 DEGs were identified between the RIF and controls. In the PPI and key module network, UBE2I, PLK4, XPO1, AURKB, and NUP107 were identified as the hub genes, which mainly enriched in RNA transport and cell division cycle-related pathways and GO items. In the miRNA-TF-DEGs network, E2F4, SIN3A, miRNA489, miRNA199A, miRNA369-3P, miRNA422, and miRNA522 were considered as the key regulatory factors during RIF. In addition, HTR1A, NR3C1, and GABRA3 were the main targets of the drugs annotated in DrugBank.Conclusion: The effects of PLK4, XPO1, AURKB, and NUP107 on the RIF may be via affecting the proliferation and differentiation of endometrial stromal cells. Besides, SIN3A and miRNA199A may be crucial for embryo implantation. In addition, NR3C1 may be used as a possible target for the clinical therapy of RIF.
Collapse
Affiliation(s)
- Fang Wang
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yaofang Liu
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
47
|
Abstract
Recurrent implantation failure (RIF) is an uncommon, imprecisely defined clinical disorder characterized by failure to achieve pregnancy after repeated embryo transfers. The diverse etiologies and incomplete understanding of RIF provide significant diagnostic and therapeutic challenges to patients and providers. Careful clinical evaluation prior to assisted reproduction can uncover many treatable causes, including thyroid dysfunction, submucosal myomas, and tobacco use. The more-subtle causes often require a more-targeted assessment. Undetected, small polyps or small areas of intrauterine synechiae are relatively common and easily treated contributors to RIF. Molecular and cellular abnormalities pose a greater therapeutic challenge. Putative causes of RIF, including progesterone resistance, shifted window of receptivity, decreased integrin expression, and immunologic disturbances, should be considered in the evaluation of a patient with otherwise unexplained RIF. It may also be true that a more complex and standardized definition of RIF would be helpful in these cases. In this paper, we review the diagnostic and therapeutic approaches to RIF, with emphasis on disorders of endometrial receptivity.
Collapse
Affiliation(s)
- Sarah Moustafa
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven L Young
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
48
|
Chibelean CB, Petca RC, Radu DC, Petca A. State of the Art in Fertility Preservation for Female Patients Prior to Oncologic Therapies. ACTA ACUST UNITED AC 2020; 56:medicina56020089. [PMID: 32102169 PMCID: PMC7073829 DOI: 10.3390/medicina56020089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/07/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022]
Abstract
Quality of life improvement stands as one of the main goals of the medical sciences. Increasing cancer survival rates associated with better early detection and extended therapeutic options led to the specific modeling of patients’ choices, comprising aspects of reproductive life that correlated with the evolution of modern society, and requires better assessment. Of these, fertility preservation and ovarian function conservation for pre-menopause female oncologic patients pose a contemporary challenge due to procreation age advance in evolved societies and to the growing expectations regarding cancer treatment. Progress made in cell and tissue-freezing technologies brought hope and shed new light on the onco-fertility field. Additionally, crossing roads with general fertility and senescence studies proved highly beneficial due to the enlarged scope and better synergies and funding. We here strive to bring attention to this domain of care and to sensitize all medical specialties towards a more cohesive approach and to better communication among caregivers and patients.
Collapse
Affiliation(s)
- Călin Bogdan Chibelean
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu-Mures, 540139 Targu-Mures, Romania;
- Mureș County Hospital, 540136 Targu-Mures, Romania
| | - Răzvan-Cosmin Petca
- “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania;
- Department of Urology, “Prof. Dr. Th. Burghele” Clinical Hospital, 050659 Bucharest, Romania
- Correspondence: ; Tel.: +40-722-224492
| | | | - Aida Petca
- “Carol Davila” University of Medicine and Pharmacy, 050471 Bucharest, Romania;
- Department of Obstetrics and Gynecology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| |
Collapse
|
49
|
Robert CA. Endometrial receptivity array for individualized determination of endometrial receptivity. Hum Reprod Open 2020; 2020:hoaa014. [PMID: 32363238 PMCID: PMC7187693 DOI: 10.1093/hropen/hoaa014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/07/2019] [Indexed: 11/17/2022] Open
Affiliation(s)
- Chris Andrea Robert
- Department of Obstetrics and Gynecology, Sunrise Hospital, Solapur, Maharashtra, 413002, India
| |
Collapse
|
50
|
He Y, Zheng H, Du H, Liu J, Li L, Liu H, Cao M, Chen S. Delayed frozen embryo transfer failed to improve live birth rate and neonatal outcomes in patients requiring whole embryo freezing. Reprod Biol Endocrinol 2020; 18:1. [PMID: 31924215 PMCID: PMC6953147 DOI: 10.1186/s12958-019-0560-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/27/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Controlled ovarian stimulation (COS) has a negative effect on the endometrial receptivity compared with natural menstrual cycle. Whether it's necessary to postpone the first frozen embryo transfer (FET) following a freeze-all strategy in order to avoid any residual effect on endometrial receptivity consequent to COS was inconclusive. OBJECTIVE The purpose of this retrospective study was to explore whether the delayed FET improve the live birth rate and neonatal outcomes stratified by COS protocols after a freeze-all strategy. METHODS A total of 4404 patients who underwent the first FET cycle were enrolled in this study between April 2014 to December 2017, and were divided into immediate (within the first menstrual cycle following withdrawal bleeding) or delayed FET (waiting for at least one menstrual cycle and the transferred embryos were cryopreserved for less than 6 months). Furthermore, each group was further divided into two subgroups according to COS protocols, and the pregnancy and neonatal outcomes were analyzed between the immediate and delayed FET following the same COS protocol. RESULTS When FET cycles following the same COS protocol, there was no significant difference regarding the rates of live birth, implantation, clinical pregnancy, multiple pregnancy, early miscarriage, premature birth and stillbirth between immediate and delayed FET groups. Similarly, no significant differences were found for the mean gestational age, the mean birth weight, and rates of low birth weight and very low birth weight between the immediate and delayed FET groups. The sex ratio (male/female) and the congenital anomalies rate also did not differ significantly between the two FET groups stratified by COS protocols. CONCLUSION Regardless of COS protocols, FET could be performed immediately after a freeze-all strategy for delaying FET failed to improve reproductive and neonatal outcomes.
Collapse
Affiliation(s)
- Yuxia He
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, Guangzhou, Guangdong, China
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, Guangdong, China
| | - Haiyan Zheng
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Hongzi Du
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Jianqiao Liu
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Lei Li
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Haiying Liu
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Mingzhu Cao
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China
| | - Shiping Chen
- Department of Reproductive Medicine, the Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, Guangdong, China.
- Key Laboratory of Reproductive Medicine of Guangdong Province, Guangzhou, Guangdong, China.
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, Guangdong, China.
| |
Collapse
|