1
|
Santos ACFDF, Zamora FV, Al-Sharif L, Sehgal K, Cavalcante DVS, Ferreira SH, da Silva PHCM. Immunosuppressants in women with repeated implantation failure in assisted reproductive techniques: a systematic review and meta-analysis. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2024; 46:e-rbgo70. [PMID: 39380592 PMCID: PMC11460425 DOI: 10.61622/rbgo/2024rbgo70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/14/2024] [Indexed: 10/10/2024] Open
Abstract
Objective To compare outcomes in patients with repeated implantation failure undergoing Intracytoplasmic Sperm Injection/In vitro fertilization (IVF/ICSI) plus immunosuppressants such as prednisolone, prednisone, or cyclosporine A versus the use of IVF/ICSI alone. Data source Databases were systematically searched in PubMed, Cochrane, and Embase databases in September 2023. Study Selection Randomized clinical trials and observational studies with the outcomes of interest were included. Data collect We computed odds ratios (ORs) for binary endpoints, with 95% confidence intervals (CIs). Heterogeneity was assessed using I2 statistics. Data were analyzed using Review Manager 5.4.The main outcomes were live birth, miscarriage, implantation rate, clinical pregnancy, and biochemical pregnancy. Data synthesis Seven studies with 2,829 patients were included. Immunosuppressive treatments were used in 1,312 (46.37%). Cyclosporine A improved implantation rate (OR 1.48; 95% CI 1.01-2.18) and clinical pregnancy (1.89, 95% CI 1.14-3.14). Compared to non-immunosuppressive treatment, prednisolone and prednisone did not improve live birth (OR 1.13, 95% CI 0.88-1.46) and miscarriage (OR 1.49, 95% CI 1.07-2.09). Prednisolone showed no significant effect in patients undergoing IVF/ICSI, clinical pregnancy (OR 1.34; 95% CI 0.76-2.36), or implantation rate (OR 1.36; 95% CI 0.76-2.42). Conclusion Cyclosporine A may promote implantation and clinical pregnancy rates. However, given the limited sample size, it is important to approach these findings with caution. Our results indicate that prednisolone and prednisone do not have any beneficial effects on clinical outcomes of IVF/ICSI patients with repeated implantation failure. PROSPERO CRD42023449655.
Collapse
Affiliation(s)
| | - Fernanda Valeriano Zamora
- Universidade Federal de Minas GeraisBelo HorizonteMGBrazilUniversidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Lubna Al-Sharif
- An-Najah National UniversityNablusWBPalestineAn-Najah National University, Nablus, WB, Palestine.
| | - Kush Sehgal
- Teerthanker Mahaveer UniversityMoradabadUPIndiaTeerthanker Mahaveer University, Moradabad, UP, India.
| | | | - Sarah Hasimyan Ferreira
- Departamento de Ginecologia e ObstetríciaFaculdade de MedicinaUniversidade Federal de GoiásGoiâniaGOBrazilDepartamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Pedro Henrique Costa Matos da Silva
- Departamento de Ginecologia e ObstetríciaFaculdade de MedicinaUniversidade Federal de GoiásGoiâniaGOBrazilDepartamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
2
|
Cuadrado-Torroglosa I, García-Velasco JA, Alecsandru D. Maternal-Fetal Compatibility in Recurrent Pregnancy Loss. J Clin Med 2024; 13:2379. [PMID: 38673652 PMCID: PMC11051463 DOI: 10.3390/jcm13082379] [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: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Nowadays, recurrent pregnancy loss (RPL) is an undesirable condition suffered by many patients of reproductive age. In this scenario, certain immune cell populations and molecules, involved in maternal-fetal compatibility, have emerged as factors related with the pathogenesis of RPL. Among them, uterine Natural Killer cells (uNKs) appear to be of great relevance. These cells are involved in numerous processes during pregnancy, such as the remodeling of uterine spiral arteries or the control of trophoblast invasion. These functions are regulated by the interactions that these cells establish with the extravillous trophoblast, mainly through their Killer Immunoglobulin-like Receptors (KIRs) and the Human Leukocyte Antigen-C (HLA-C) molecules expressed by the embryo. A high level of polymorphism has been reported for both molecules involved in this interaction, with some of the possible KIR-HLA-C combinations being associated with an increased risk of RPL. However, the complexity of the maternal-fetal interface goes beyond this, as other HLA molecules also appear to be related to this reproductive pathology. In this review, we will discuss the role of uNKs in pregnancy, as well as the polymorphisms and clinical implications of KIR-HLA-C binding. We will also address the involvement of other, different HLA molecules in RPL, and the current advice on the appropriate management of patients with 'immunological mismatch', thus covering the main aspects regarding the involvement of maternal-fetal compatibility in RPL.
Collapse
Affiliation(s)
- Isabel Cuadrado-Torroglosa
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
| | - Juan A. García-Velasco
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
- Department of Obstetrics and Gynaecology, Rey Juan Carlos University, Av. de Atenas, s/n, 28922 Alcorcón, Spain
| | - Diana Alecsandru
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
| |
Collapse
|
3
|
Nakagawa K, Sugiyama R. Tacrolimus treatment in women with repeated implantation failures. Reprod Med Biol 2024; 23:e12558. [PMID: 38196832 PMCID: PMC10775185 DOI: 10.1002/rmb2.12558] [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: 07/29/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024] Open
Abstract
Background Tacrolimus is an immunosuppressive drug that works as a calcineurin inhibitor to improve the reproductive outcomes for women who have experienced multiple implantation failures (RIF) and show elevated type 1 helper T (Th1)/Th2 cell ratios. Methods In the first part of this review, we indicate how we re-evaluated the cut-off index for selecting the participants in a tacrolimus regimen via transferred euploid blastocysts. In the second part, we cite cases where tacrolimus has improved the live birth rate for women who have experienced recurrent pregnancy losses (PRL) and we introduce the utility of tacrolimus treatment to prevent obstetrical complications. Main Findings After reconsideration of the cut-off index (Th1/Th2 ≥ 11.8), however, the pregnancy rates of women with tacrolimus were significantly higher than those of women without tacrolimus. The PRL women treated with tacrolimus showed significantly lower rates of biochemical pregnancy, but higher live-birth rates compared with women who were not treated with tacrolimus. Moreover, prior severe obstetrical complications could be controlled via the administration of tacrolimus during pregnancy. Conclusion Tacrolimus has become indispensable in the field of solid-organ transplantation, and in the near future, it should become an essential agent in the reproductive field, as well.
Collapse
Affiliation(s)
- Koji Nakagawa
- Center for Reproductive Medicine and Implantation ResearchSugiyama Clinic ShinjukuTokyoJapan
| | - Rikikazu Sugiyama
- Center for Reproductive Medicine and Implantation ResearchSugiyama Clinic ShinjukuTokyoJapan
| |
Collapse
|
4
|
Li T, Liu X, Yang X, Wang N, Wang Y, Li W, Liang X, Huang R. Comparison of the efficacy of depot GnRH agonist protocol and the GnRH antagonist protocol in patients with repeated IVF failure: a retrospective cohort study. HUM FERTIL 2023; 26:1299-1306. [PMID: 36946058 DOI: 10.1080/14647273.2023.2175267] [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] [Received: 12/21/2021] [Accepted: 09/11/2022] [Indexed: 03/23/2023]
Abstract
The aims of the research were (i) to compare the clinical outcome of IVF using follicular-phase depot gonadotropin-releasing hormone (GnRH) agonist (depot agonist) protocol and GnRH antagonist protocol in patients with repeated IVF failure (RIF), (ii) to discover the optimal ovarian stimulation protocol for this group of low prognosis patients. 801 RIF patients with normal ovarian reserve receiving in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) were included in this retrospective study. Among them, 492 patients were treated with the depot agonist protocol, and the remaining 309 patients with the antagonist protocol. Multivariable logistic regression analysis was used to find the predictor(s) of the chance of live birth. Higher live birth rate (LBR) and clinical pregnancy rate (CPR) in fresh embryo transfer (ET) cycles were associated with the use of depot agonist compared with the antagonist protocols (44.81% vs. 27.27%, 54.64% vs. 34.93%, respectively; both p < 0.01) and depot agonist protocol was a strong predictor of live birth (OR = 2.182, 95% CI 1.355-3.514, p < 0.01). The CPR in thawed ET cycles was not significantly different between the two groups (38.12% vs. 45.26%, p > 0.05). A higher cumulative live birth rate (CLBR) was achieved in the depot agonist group (46.59% vs. 35.21%, p < 0.01). Beneficial endometrial receptivity in the depot agonist protocol contributed to a higher LBR in fresh ET cycles, rendering this protocol the preferred option in the treatment of RIF patients.
Collapse
Affiliation(s)
- Tingting Li
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaoping Liu
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xing Yang
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Ningning Wang
- Department of Statistics, Guangzhou Medical University, Guangzhou, P.R. China
| | - Yanfang Wang
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Wei Li
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaoyan Liang
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Rui Huang
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
5
|
Lédée N, Petitbarat M, Prat-Ellenberg L, Dray G, Vaucoret V, Kazhalawi A, Rodriguez-Pozo A, Habeichi N, Ruoso L, Cassuto NG, Rahmati M. The Next Frontier in ART: Harnessing the Uterine Immune Profile for Improved Performance. Int J Mol Sci 2023; 24:11322. [PMID: 37511080 PMCID: PMC10379072 DOI: 10.3390/ijms241411322] [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: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Assisted reproduction techniques have improved considerably in recent decades, but despite these advances, success rates remain relatively low. Endometrial immune profiling involves the analysis of cytokine biomarkers in the endometrium during the mid-luteal phase. This profiling aims to provide insights into the immune environment of the uterus. The aim is to identify immune disturbances and thus guide the development of personalized therapeutic approaches. The first part of the review looks back at the emergence of innovative concepts, highlighting the specificity of the human uterine environment at the time of implantation. Based on this new knowledge, biomarkers have been selected for endometrial immune profiling. The second part details the results of clinical studies conducted over the last ten years. These clinical results suggest that this approach can increase the rate of live births in patients suffering from repeated implantation failures or repeated pregnancy loss. Uterine immune profiling represents a clinical innovation that can significantly improve the performance of medically assisted reproduction treatments through personalized strategies tailored to the local immune profile. Innovation in personalized medicine for assisted reproduction is crucial to improving the success rates of fertility treatments, while reducing the risks and costs associated with ineffective or unnecessary interventions.
Collapse
Affiliation(s)
- Nathalie Lédée
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 2 Rue Antoine Etex, 94000 Creteil, France
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, 4 Rue Lasson, 75012 Paris, France
| | - Marie Petitbarat
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 2 Rue Antoine Etex, 94000 Creteil, France
| | - Laura Prat-Ellenberg
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, 4 Rue Lasson, 75012 Paris, France
| | - Géraldine Dray
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, 4 Rue Lasson, 75012 Paris, France
| | - Virginie Vaucoret
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, 4 Rue Lasson, 75012 Paris, France
| | - Alaa Kazhalawi
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 2 Rue Antoine Etex, 94000 Creteil, France
| | - André Rodriguez-Pozo
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 2 Rue Antoine Etex, 94000 Creteil, France
| | - Nada Habeichi
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 2 Rue Antoine Etex, 94000 Creteil, France
| | - Lea Ruoso
- Laboratoire Drouot, 21 Rue Drouot, 75010 Paris, France
| | | | - Mona Rahmati
- London Women's Clinic, 113-115 Harley Street, London W1G 6AP, UK
| |
Collapse
|
6
|
Sun Y, Cui L, Lu Y, Tan J, Dong X, Ni T, Yan J, Guan Y, Hao G, Liu JY, Zhang B, Wei D, Hong Y, He Y, Qi J, Xu B, Lu J, Zhang Q, Zhao S, Ji X, Du X, Zhang J, Liu J, Wang J, Huang Y, Huang D, Du Y, Vankelecom H, Zhang H, Chen ZJ. Prednisone vs Placebo and Live Birth in Patients With Recurrent Implantation Failure Undergoing In Vitro Fertilization: A Randomized Clinical Trial. JAMA 2023; 329:1460-1468. [PMID: 37129654 PMCID: PMC10155063 DOI: 10.1001/jama.2023.5302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/18/2023] [Indexed: 05/03/2023]
Abstract
Importance Implantation failure remains a critical barrier to in vitro fertilization. Prednisone, as an immune-regulatory agent, is widely used to improve the probability of implantation and pregnancy, although the evidence for efficacy is inadequate. Objective To determine the efficacy of 10 mg of prednisone compared with placebo on live birth among women with recurrent implantation failure. Design, Setting, and Participants A double-blind, placebo-controlled, randomized clinical trial conducted at 8 fertility centers in China. Eligible women who had a history of 2 or more unsuccessful embryo transfer cycles, were younger than 38 years when oocytes were retrieved, and were planning to undergo frozen-thawed embryo transfer with the availability of good-quality embryos were enrolled from November 2018 to August 2020 (final follow-up August 2021). Interventions Participants were randomized (1:1) to receive oral pills containing either 10 mg of prednisone (n = 357) or matching placebo (n = 358) once daily, from the day at which they started endometrial preparation for frozen-thawed embryo transfer through early pregnancy. Main Outcomes and Measures The primary outcome was live birth, defined as the delivery of any number of neonates born at 28 or more weeks' gestation with signs of life. Results Among 715 women randomized (mean age, 32 years), 714 (99.9%) had data available on live birth outcomes and were included in the primary analysis. Live birth occurred among 37.8% of women (135 of 357) in the prednisone group vs 38.8% of women (139 of 358) in the placebo group (absolute difference, -1.0% [95% CI, -8.1% to 6.1%]; relative ratio [RR], 0.97 [95% CI, 0.81 to 1.17]; P = .78). The rates of biochemical pregnancy loss were 17.3% in the prednisone group and 9.9% in the placebo group (absolute difference, 7.5% [95% CI, 0.6% to 14.3%]; RR, 1.75 [95% CI, 1.03 to 2.99]; P = .04). Of those in the prednisone group, preterm delivery occurred among 11.8% and of those in the placebo group, 5.5% of pregnancies (absolute difference, 6.3% [95% CI, 0.2% to 12.4%]; RR, 2.14 [95% CI, 1.00 to 4.58]; P = .04). There were no statistically significant between-group differences in the rates of biochemical pregnancy, clinical pregnancy, implantation, neonatal complications, congenital anomalies, other adverse events, or mean birthweights. Conclusions and Relevance Among patients with recurrent implantation failure, treatment with prednisone did not improve live birth rate compared with placebo. Data suggested that the use of prednisone may increase the risk of preterm delivery and biochemical pregnancy loss. Our results challenge the value of prednisone use in clinical practice for the treatment of recurrent implantation failure. Trial Registration Chinese Clinical Trial Registry Identifier: ChiCTR1800018783.
Collapse
Affiliation(s)
- 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
| | - Linlin Cui
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 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
| | - Jichun Tan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianxiang Ni
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Yichun Guan
- Reproductive Medicine Center, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guimin Hao
- Department of Reproductive Medicine, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jia-Yin Liu
- Department of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Bo Zhang
- Center for Reproductive Medicine, Maternal and Child Health Hospital/Obstetrics and Gynecology Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Daimin Wei
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Yan Hong
- 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
| | - 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
| | - Bing Xu
- 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
| | - Juanjuan Lu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Qian Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Shanshan Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China
| | - Xiaowei Ji
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaofang Du
- Reproductive Medicine Center, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhang
- Department of Reproductive Medicine, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinyong Liu
- Department of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Jing Wang
- Department of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Yingqin Huang
- Center for Reproductive Medicine, Maternal and Child Health Hospital/Obstetrics and Gynecology Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Dongmei Huang
- Center for Reproductive Medicine, Maternal and Child Health Hospital/Obstetrics and Gynecology Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Yanzhi Du
- 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
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Heping Zhang
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut
| | - Zi-jiang Chen
- 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
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| |
Collapse
|
7
|
Ban Y, Yang X, Xing Y, Que W, Yu Z, Gui W, Chen Y, Liu X. Intrauterine Infusion of Leukocyte-Poor Platelet-Rich Plasma Is an Effective Therapeutic Protocol for Patients with Recurrent Implantation Failure: A Retrospective Cohort Study. J Clin Med 2023; 12:jcm12082823. [PMID: 37109159 PMCID: PMC10146382 DOI: 10.3390/jcm12082823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The clinical application of autologous leukocyte-poor platelet-rich plasma (LP-PRP) in patients with recurrent implantation failure (RIF) is rare. This retrospective observational cohort study aimed to evaluate the efficacy of LP-PRP intrauterine infusion in patients with RIF. METHODS Patients with RIF undergoing frozen embryo transfer (FET) from January 2019 to December 2021 (n = 118) were enrolled, with those undergoing LP-PRP intrauterine infusion as the PRP group (n = 64), and those receiving no LP-PRP treatment as the control group (n = 54). The beta-human chorionic gonadotropin (β-hCG)-positive rate, clinical pregnancy rate (CPR), live birth rate (LBR), and miscarriage rate (MR) per ET cycle were compared. RESULTS The β-hCG-positive rate (57.8% vs. 38.9%, p = 0.041), CPR (45.3% vs. 24.5%, p = 0.022), and LBR per ET cycle (42.2% vs. 18.5%, p = 0.009) were higher in the PRP group than in the control group, and the three variables (62.5% vs. 41.2%, p = 0.040, 47.5% vs. 23.5%, p = 0.033, and 47.5% vs. 20.6%, p = 0.027) in the PRP group transferred with the blastocyst-stage embryos were also higher than those in the control group. The MR was similar in all groups. CONCLUSIONS The LP-PRP treatment could improve the β-hCG-positive rate, CPR, and LBR in RIF patients undergoing FET cycles.
Collapse
Affiliation(s)
- Yanna Ban
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Xiaoliang Yang
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Yan Xing
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Wenjun Que
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Zebo Yu
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Wenwu Gui
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Ying Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Xiru Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| |
Collapse
|
8
|
Vexø LE, Stormlund S, Landersoe SK, Jørgensen HL, Humaidan P, Bergh C, Englund ALM, Klajnbard A, Bogstad JW, Freiesleben NLC, Zedeler A, Prætorius L, Andersen AN, Løssl K, Pinborg A, Nielsen HS. Low-grade inflammation is negatively associated with live birth in women undergoing IVF. Reprod Biomed Online 2023; 46:302-311. [PMID: 36446681 DOI: 10.1016/j.rbmo.2022.10.004] [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] [Received: 07/12/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 11/23/2022]
Abstract
RESEARCH QUESTION Is low-grade inflammation, detected by C-reactive protein (CRP), a marker of IVF outcome addressing both blastocyst quality and pregnancy outcome? DESIGN This sub-study of a multicentre randomized controlled trial included 440 women undergoing IVF treatment with a gonadotrophin-releasing hormone (GnRH) antagonist protocol. Serum CRP was measured on cycle day 2-3 (baseline) and on the day of ovulation triggering. The association between CRP concentrations and reproductive outcomes (number of retrieved oocytes, number of good-quality blastocysts, pregnancy, pregnancy loss and live birth), were analysed, adjusting for relevant confounders. RESULTS A negative association was found between higher baseline CRP concentrations and live birth rate (odds ratio [OR] 0.77, 95% confidence interval [CI] 0.62-0.96, P = 0.02) and higher CRP concentrations at baseline were associated with pregnancy loss among women who conceived (OR 1.37, 95% CI 1.07-1.76, P = 0.01). When testing for a specific cut-off, CRP concentrations above 2.34 (the highest quartile) were more likely to be associated with pregnancy loss (P = 0.02) and a lower chance of live birth (P = 0.04) compared with the lowest quartile. No associations were found between CRP concentrations and pregnancy outcomes on the day of ovulation triggering, and there were no associations between CRP concentrations and the number of good-quality blastocysts. CONCLUSIONS Higher CRP concentrations at cycle day 2-3, before starting ovarian stimulation, are negatively associated with chance of live birth, possibly because of an increased risk of pregnancy loss. No association was found between the number of good-quality blastocysts and CRP concentration. More studies are needed to investigate the impact of low-grade inflammation.
Collapse
Affiliation(s)
- Laura Emilie Vexø
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
| | - Sacha Stormlund
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Selma Kloeve Landersoe
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henrik Løvendahl Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Humaidan
- Fertility Clinic, Skive Regional Hospital and Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Christina Bergh
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University; Reproductive Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anne Lis Mikkelsen Englund
- Department of Obstetrics and Gynaecology, Fertility Clinic, Region Zealand University Hospital, Koege, Denmark
| | - Anna Klajnbard
- Fertility Clinic, Department of Obstetrics and Gynaecology, Herlev University Hospital, Copenhagen, Denmark
| | - Jeanette Wulff Bogstad
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nina la Cour Freiesleben
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Zedeler
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Lisbeth Prætorius
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Anders Nyboe Andersen
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kristine Løssl
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anja Pinborg
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Svarre Nielsen
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Marron K, Harrity C. Correlation of peripheral blood and endometrial immunophenotyping in ART: is peripheral blood sampling useful? J Assist Reprod Genet 2023; 40:381-387. [PMID: 36574140 PMCID: PMC9935767 DOI: 10.1007/s10815-022-02696-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Using a comprehensive flow cytometric panel, simultaneously obtained mid-luteal immunophenotypes from peripheral blood and endometrium were compared and values correlated. Is a peripheral blood evaluation of reproductive immunophenotype status meritorious relative to local endometrial evaluation to directly assess the peri-implantation environment? METHODS Fifty-five patients had a mid-luteal biopsy to assess the local endometrial immunophenotype, while simultaneously providing a peripheral blood sample for analysis. Both samples were immediately assessed using a comprehensive multi-parameter panel, and lymphocyte subpopulations were described and compared. RESULTS Distinct lymphocyte proportions and percentage differences were noted across the two compartments, confirming the hypothesis that they are distinct environments. The ratio of CD4 + to CD8 + T cells were reversed between the two compartments, as were Th1 and Th2-type CD4 + T cell ratios. Despite these differences, some direct relationships were noted. Positive Pearson correlations were found between the levels of CD57 + expressing natural killer cells, CD3 + NK-T cells and CD4 + Th1 cells in both compartments. CONCLUSIONS Flow cytometric evaluation provides a rapid and objective analysis of lymphocyte subpopulations. Endometrial biopsies have become the gold standard technique to assess the uterine immunophenotype in adverse reproductive outcome, but there may still a place for peripheral blood evaluation in this context. The findings demonstrate significant variations in cellular proportions across the two regions, but some positive correlations are present. Immunological assessment of these specific peripheral blood lymphocyte subtypes may provide insight into patients with potential alterations of the uterine immune environment, without the risks and inconveniences associated with an invasive procedure.
Collapse
Affiliation(s)
- Kevin Marron
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin 14, Ireland.
| | - Conor Harrity
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
10
|
Genest G, Banjar S, Almasri W, Beauchamp C, Benoit J, Buckett W, Dzineku F, Gold P, Dahan MH, Jamal W, Jacques Kadoch I, Kadour-Peero E, Lapensée L, Miron P, Shaulov T, Sylvestre C, Tulandi T, Mazer BD, Laskin CA, Mahutte N. Immunomodulation for unexplained recurrent implantation failure: where are we now? Reproduction 2023; 165:R39-R60. [PMID: 36322478 DOI: 10.1530/rep-22-0150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/02/2022] [Indexed: 11/05/2022]
Abstract
In brief Immune dysfunction may contribute to or cause recurrent implantation failure. This article summarizes normal and pathologic immune responses at implantation and critically appraises currently used immunomodulatory therapies. Abstract Recurrent implantation failure (RIF) may be defined as the absence of pregnancy despite the transfer of ≥3 good-quality blastocysts and is unexplained in up to 50% of cases. There are currently no effective treatments for patients with unexplained RIF. Since the maternal immune system is intricately involved in mediating endometrial receptivity and embryo implantation, both insufficient and excessive endometrial inflammatory responses during the window of implantation are proposed to lead to implantation failure. Recent strategies to improve conception rates in RIF patients have focused on modulating maternal immune responses at implantation, through either promoting or suppressing inflammation. Unfortunately, there are no validated, readily available diagnostic tests to confirm immune-mediated RIF. As such, immune therapies are often started empirically without robust evidence as to their efficacy. Like other chronic diseases, patient selection for immunomodulatory therapy is crucial, and personalized medicine for RIF patients is emerging. As the literature on the subject is heterogenous and rapidly evolving, we aim to summarize the potential efficacy, mechanisms of actions and side effects of select therapies for the practicing clinician.
Collapse
Affiliation(s)
- Geneviève Genest
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Shorooq Banjar
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Walaa Almasri
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Coralie Beauchamp
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - Joanne Benoit
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - William Buckett
- McGill University Health Centre Reproductive Centre, Montreal, Quebec, Canada
| | | | - Phil Gold
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Michael H Dahan
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Wael Jamal
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | | | - Einav Kadour-Peero
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, McGill University, Montréal, Quebec, Canada
| | - Louise Lapensée
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - Pierre Miron
- Fertilys Reproductive Center, Laval, Quebec, Canada
| | - Talya Shaulov
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Camille Sylvestre
- Division of Reproductive Endocrinology and Infertility, University of Montreal, Montreal, Quebec, Canada
| | - Togas Tulandi
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Bruce D Mazer
- Department of Pediatrics, McGill University, Division of Allergy Immunology and Clinical Dermatology, Montreal Children's Hospital, McGill University, Montréal, Quebec, Canada
| | - Carl A Laskin
- Deptartments of Medicine and Obstetrics & Gynecology University of Toronto, Toronto, Ontario, Canada
| | - Neal Mahutte
- The Montreal Fertility Centre, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Ma J, Gao W, Li D. Recurrent implantation failure: A comprehensive summary from etiology to treatment. Front Endocrinol (Lausanne) 2023; 13:1061766. [PMID: 36686483 PMCID: PMC9849692 DOI: 10.3389/fendo.2022.1061766] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Implantation is the first step in human reproduction. Successful implantation depends on the crosstalk between embryo and endometrium. Recurrent implantation failure (RIF) is a clinical phenomenon characterized by a lack of implantation after the transfer of several embryos and disturbs approximately 10% couples undergoing in vitro fertilization and embryo transfer. Despite increasing literature on RIF, there is still no widely accepted definition or standard protocol for the diagnosis and treatment of RIF. Progress in predicting and preventing RIF has been hampered by a lack of widely accepted definitions. Most couples with RIF can become pregnant after clinical intervention. The prognosis for couples with RIF is related to maternal age. RIF can be caused by immunology, thrombophilias, endometrial receptivity, microbiome, anatomical abnormalities, male factors, and embryo aneuploidy. It is important to determine the most possible etiologies, and individualized treatment aimed at the primary cause seems to be an effective method for increasing the implantation rate. Couples with RIF require psychological support and appropriate clinical intervention. Further studies are required to evaluate diagnostic method and he effectiveness of each therapy, and guide clinical treatment.
Collapse
Affiliation(s)
- Junying Ma
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, Shenyang, China
- Shengjing Hospital of China Medical University, Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China
| | - Wenyan Gao
- Department of Obstetrics, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, Shenyang, China
- Shengjing Hospital of China Medical University, Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China
| |
Collapse
|
12
|
Cheng R, Liu Q, Zhu Y, Zhao Y, Yang L, Zhang Q. Effectiveness of Jian-Pi-An-Tai formula for the pregnancy outcome of in vitro fertilization and embryo transfer in infertile women: Protocol of a randomized controlled trial. Medicine (Baltimore) 2022; 101:e32419. [PMID: 36595804 PMCID: PMC9794354 DOI: 10.1097/md.0000000000032419] [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] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Infertility has become a global public health issue. In vitro fertilization and embryo transfer (IVF-ET) is widely performed as an infertility treatment. However, a significant number of infertile women continue to experience serial implantation failure, despite the high quality of the transferred embryos. Jian-Pi-An-Tai formula is an experience formula in treating threatened abortion in our team. It has also been used to treat infertile women after embryo transfer and shows superiority compared to single use of conventional western medicine. However, the of evidence on its effective in treating infertile women undergo IVF-ET is lacking. METHODS This randomized controlled trial (RCT) will be carried out in Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University planning to recruit 180 infertile patients undergoing IVF-ET with the type of deficiency of both the spleen and kidney. The control group will be treated by conventional western medicine and the treatment group will use conventional western medicine plus Jian-Pi-An-Tai formula. The primary outcomes will include Embryo implantation rate, Clinical pregnancy rate, Persistent pregnancy rate; and the secondary outcomes will include TCM symptom score and reproductive hormones. Safety evaluation will be recorded during the whole study. All data in this RCT will be analyzed by SPSS 23.0 software. This study has been approved by the Research Ethics Committee of the Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University (2022KY130). DISCUSSION The results of this RCT will contribute to provide scientific and rigorous evidence for the efficacy and safety of Jian-Pi-An-Tai formula in treating infertile women undergo IVF-ET. And the results from this RCT will be published in a relevant journal after finished.
Collapse
Affiliation(s)
- Ran Cheng
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Liu
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuqing Yang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- * Correspondence: Qin Zhang, Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453th Stadium Road, Hangzhou, Zhejiang Province Hangzhou 310007, China (e-mail: )
| |
Collapse
|
13
|
Sacks G, Zhang J. Prednisolone and enoxaparin (clexane) therapy ('the Bondi protocol') for repeated IVF failure. Am J Reprod Immunol 2022; 88:e13616. [PMID: 36067527 PMCID: PMC9788304 DOI: 10.1111/aji.13616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/13/2022] [Accepted: 08/15/2022] [Indexed: 12/30/2022] Open
Abstract
PROBLEM What is the impact of an empirical immune therapy protocol of prednisolone and enoxaparin (clexane) (the 'Bondi protocol') on women with repeated in vitro fertilization (IVF) failure? METHOD OF STUDY This was a retrospective review of live birth outcomes conducted on all transfer cycles performed by a single clinician (GS) at IVFAustralia between February 2016 and April 2020. This study consisted of 1786 transfer cycles, including 460 cycles treated with the Bondi protocol and 1326 without. Women with repeated IVF failure were given the Bondi protocol based on blood NK cell activity. Primary outcome was live birth and statistical analysis was performed with GraphPad Prism software with significance for P-values < .05. RESULTS Overall 'Bondi' and 'normal' protocol cycles had similar rates of IVF/ICSI, fresh/frozen transfers and use of preimplantation genetic testing (PGT). Women given the Bondi protocol were older, had more previous cycles and had higher blood NK cell activity. There was no significant difference in live birth rates (Bondi 26%, normal 28%). Bondi protocol live birth rates per transfer cycle were as high as 40% in patients under 38 years old. The Bondi protocol was more effective as NK activity increased from 'normal' to 'borderline' to 'high'. For high NK cell activity levels, live birth rates were over four times higher for women on the Bondi protocol (28%) than those on normal protocols (6%, P = .0007). CONCLUSION This study describes a simple and relatively safe immune therapy protocol that may improve IVF success rates in women with evidence of immune dysfunction.
Collapse
Affiliation(s)
- Gavin Sacks
- IVFAustraliaSydneyAustralia
- Department of Women’s and Children’s HealthUniversity of New South WalesSydneyAustralia
- St George Hospital and Royal Hospital for WomenSydneyAustralia
| | - Jessica Zhang
- Department of Women’s and Children’s HealthUniversity of New South WalesSydneyAustralia
| |
Collapse
|
14
|
Marron K, Harrity C. Potential utility of a non-invasive menstrual blood immunophenotype analysis in reproductive medicine. REPRODUCTION AND FERTILITY 2022; 3:RAF-22-0047. [PMID: 36173705 PMCID: PMC9641796 DOI: 10.1530/raf-22-0047] [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: 04/19/2022] [Accepted: 09/16/2022] [Indexed: 08/27/2023] Open
Abstract
Purpose Can a comprehensive flow cytometry panel be used to assess immunophenotype profiles in menstrual blood of patients experiencing reproductive failure and age matched controls of proven fertility? Methods 58 recurrent pregnancy loss and repeated implantation failure patients, along with 15 age matched controls of proven fertility, had menstrual blood samples obtained within the first 24 hours of the onset of menstruation to non-invasively assess the local immunophenotype. Using a comprehensive multi-parameter flow panel the lymphocyte sub-populations were described and compared. Results Relative to well established peripheral blood immunophenotyping values, distinct lymphocyte population differences were noted between the subgroups. The ratios of CD4+ and CD8+ T-cells were inverted relative to peripheral blood and uterine NK cells represented by CD56bright were distinctly visualised, emphasising the distinction of menstrual and peripheral blood. Relative to controls there were marked increases in CD3+ve T-cells (p=0.009), CD4:CD8 ratio (p=0.004), CD19 B-cells (p=0.026) and CD56dim NK's (p=0.002) in the reproductive failure cases. Conclusions Flow cytometric evaluation can provide a rapid and objective analysis of lymphocyte subpopulations in many forms of tissue and fluid. The findings show significant variations in cellular composition of immune cells indicating a distinct compartment, with differences between cases and controls. Immunological assessment of the menstrual blood immunophenotype, in clinically appropriate patients, may provide insight into the aetiology of adverse reproductive outcome, without the risks and inconveniences associated with a more invasive endometrial biopsy.
Collapse
Affiliation(s)
| | - Conor Harrity
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
15
|
Boomsma CM, Kamath MS, Keay SD, Macklon NS. Peri-implantation glucocorticoid administration for assisted reproductive technology cycles. Cochrane Database Syst Rev 2022; 6:CD005996. [PMID: 35771604 PMCID: PMC9245898 DOI: 10.1002/14651858.cd005996.pub4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND The use of peri-implantation glucocorticoids has been advocated to improve embryo implantation during assistive reproductive technology (ART) cycles such as in vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI). It has been proposed that glucocorticoids may improve the intrauterine environment by acting as immunomodulators to reduce the uterine natural killer (NK) cell count and activity, normalising the cytokine expression profile in the endometrium and by suppression of endometrial inflammation. OBJECTIVES To evaluate the effectiveness and safety of glucocorticoids versus no glucocorticoids administered around the time of anticipated implantation in women undergoing IVF or ICSI. SEARCH METHODS We searched the Cochrane Gynaecology and Fertility (CGF) Group specialised register, CENTRAL (now also containing output from two trial registers and CINAHL), MEDLINE and Embase, on 20 December 2021, together with reference checking, contact with experts in the field and relevant conference proceedings to identify additional studies. This review is an update of the review first published in 2007 and last updated in 2012. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing the efficacy of supplementary systemic administration of glucocorticoids in the peri-implantation period with a placebo or no glucocorticoids in subfertile women undergoing IVF or ICSI were included. DATA COLLECTION AND ANALYSIS We used standard methodological procedures recommended by Cochrane. The primary review outcomes were live birth rate and multiple pregnancy. MAIN RESULTS We included 16 RCTs (2232 couples analysed). We are uncertain whether glucocorticoids improved live birth rates (odds ratio (OR) 1.37, 95% confidence interval (CI) 0.69 to 2.71; 2 RCTs, n = 366; I2 = 7%; very low-certainty evidence). This suggests that if the chance of live birth following no glucocorticoids/placebo is assumed to be 9%, the chance following glucocorticoids would be between 6% and 21%. We are also uncertain whether there was a difference between peri-implantation glucocorticoids on multiple pregnancy rates per couple (OR 0.86, 95% CI 0.33 to 2.20; 4 RCTs, n = 504; I2 = 53%; very low-certainty evidence). The I2 of 53% may represent moderate statistical heterogeneity and results have to be interpreted with caution. With regard to pregnancy rates, we are uncertain whether there was a difference between ongoing pregnancy rates after glucocorticoids versus no glucocorticoids/placebo (OR 1.19, 95% CI 0.80 to 1.76; 3 RCTs, n = 476; I2 = 0%; very low-certainty evidence) and clinical pregnancy rates after glucocorticoids versus no glucocorticoids/placebo (OR 1.17, 95% CI 0.95 to 1.44; 13 RCTs, n = 1967; I2 = 0%; low-certainty evidence). This suggests that if the chance of clinical pregnancy following no glucocorticoids/placebo is assumed to be 25%, the chance following glucocorticoids would be between 24% and 32%. Furthermore, we are also uncertain whether peri-implantation glucocorticoids influenced miscarriage rates per couple (OR 1.09, 95% CI 0.63 to 1.87; 6 RCTs, n = 821; I2 = 0%; very low-certainty evidence), the incidence of ectopic pregnancies per couple (OR 2.28, 95% CI 0.33 to 15.62; 3 RCTs, n = 320; I2 = 0%; very low-certainty evidence) and ovarian hyperstimulation syndrome (OHSS) per couple (OR 1.07, 95% CI 0.60 to 1.90; 3 RCTs, n = 370; I2 = 0%; very low-certainty evidence) compared to no glucocorticoids/placebo. The evidence was very low to low certainty: the main limitations were serious risk of bias due to poor reporting of study methods, and serious imprecision. AUTHORS' CONCLUSIONS Overall, there was insufficient evidence that administration of peri-implantation glucocorticoids in IVF/ICSI cycles influenced clinical outcomes. These findings were limited to the routine use of glucocorticoids in subfertile women undergoing IVF or ICSI.
Collapse
Affiliation(s)
- Carolien M Boomsma
- Obstetrics and Gynaecology, Bravis Hospital, Bergen op Zoom, Netherlands
| | - Mohan S Kamath
- Department of Reproductive Medicine and Surgery, Christian Medical College, Vellore, India
| | - Stephen D Keay
- Centre for Reproductive Medicine, UHCW NHS Trust, Coventry, UK
| | | |
Collapse
|
16
|
Robertson SA, Moldenhauer LM, Green ES, Care AS, Hull ML. Immune determinants of endometrial receptivity: a biological perspective. Fertil Steril 2022; 117:1107-1120. [PMID: 35618356 DOI: 10.1016/j.fertnstert.2022.04.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/04/2022]
Abstract
Immune cells are essential for endometrial receptivity to embryo implantation and early placental development. They exert tissue-remodeling and immune regulatory roles-acting to promote epithelial attachment competence, regulate the differentiation of decidual cells, remodel the uterine vasculature, control and resolve inflammatory activation, and suppress destructive immunity to paternally inherited alloantigens. From a biological perspective, the endometrial immune response exerts a form of "quality control"-it promotes implantation success when conditions are favorable but constrains receptivity when physiological circumstances are not ideal. Women with recurrent implantation failure and recurrent miscarriage may exhibit altered numbers or disturbed function of certain uterine immune cell populations-most notably uterine natural killer cells and regulatory T cells. Preclinical and animal studies indicate that deficiencies or aberrant activation states in these cells can be causal in the pathophysiological mechanisms of infertility. Immune cells are, therefore, targets for diagnostic evaluation and therapeutic intervention. However, current diagnostic tests are overly simplistic and have limited clinical utility. To be more informative, they need to account for the full complexity and reflect the range of perturbations that can occur in uterine immune cell phenotypes and networks. Moreover, safe and effective interventions to modulate these cells are in their infancy, and personalized approaches matched to specific diagnostic criteria will be needed. Here we summarize current biological understanding and identify knowledge gaps to be resolved before the promise of therapies to target the uterine immune response can be fully realized.
Collapse
Affiliation(s)
- Sarah A Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Lachlan M Moldenhauer
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Ella S Green
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Alison S Care
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - M Louise Hull
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Liu R, Dai M, Gong G, Chen M, Cao C, Wang T, Hou Z, Shi Y, Guo J, Zhang Y, Xia X. The role of extracellular matrix on unfavorable maternal–fetal interface: focusing on the function of collagen in human fertility. JOURNAL OF LEATHER SCIENCE AND ENGINEERING 2022. [DOI: 10.1186/s42825-022-00087-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AbstractExtracellular matrix (ECM) is characterized as widespread, abundant, and pluripotent. Among ECM members, collagen is widely accepted as one of the most prominent components for its essential structural property that can provide a scaffold for other components of ECM and the rich biological functions, which has been extensively used in tissue engineering. Emerging evidence has shown that the balance of ECM degradation and remodeling is vital to regulations of maternal–fetal interface including menstrual cycling, decidualization, embryo implantation and pregnancy maintenance. Moreover, disorders in these events may eventually lead to failure of pregnancy. Although the improvement of assisted conception and embryo culture technologies bring hope to many infertile couples, some unfavorable outcomes, such as recurrent implantation failure (RIF), recurrent pregnancy loss (RPL) or recurrent miscarriage (RM), keep troubling the clinicians and patients. Recently, in vitro three-dimensional (3D) model mimicking the microenvironment of the maternal–fetal interface is developed to investigate the physiological and pathological conditions of conception and pregnancy. The progress of this technology is based on clarifying the role of ECM in the endometrium and the interaction between endometrium and conceptus. Focusing on collagen, the present review summarized the degradation and regulation of ECM and its role in normal menstruation, endometrium receptivity and unsatisfying events occurring in infertility treatments, as well as the application in therapeutic approaches to improve pregnancy outcomes. More investigations about ECM focusing on the maternal–fetal interface interaction with mesenchymal stem cells or local immunoregulation may inspire new thoughts and advancements in the clinical application of infertility treatments.
Graphical abstract
Collapse
|
18
|
OUP accepted manuscript. Hum Reprod Update 2022; 28:435-454. [DOI: 10.1093/humupd/dmac007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/15/2021] [Indexed: 11/13/2022] Open
|
19
|
Kieffer TE, Chin PY, Green ES, Moldenhauer LM, Prins JR, Robertson SA. Prednisolone in early pregnancy inhibits regulatory T cell generation and alters fetal and placental development in mice. Mol Hum Reprod 2021; 26:340-352. [PMID: 32159777 DOI: 10.1093/molehr/gaaa019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/16/2020] [Indexed: 01/01/2023] Open
Abstract
Corticosteroids have been utilised in the assisted reproduction setting with the expectation of suppressing aberrant immune activation and improving fertility in women. However, the effects of corticosteroids on fertility, and on pregnancy and offspring outcomes, are unclear. In this study, mice were administered prednisolone (1 mg/kg) or PBS daily in the pre-implantation phase, and effects on the adaptive immune response, the implantation rate, fetal development and postnatal outcomes were investigated. Prednisolone disrupted the expected expansion of CD4+ T cells in early pregnancy, inhibiting generation of both regulatory T cells (Treg cells) and effector T cells and suppressing IFNG required for T cell functional competence. Prednisolone caused an 8-20% increase in the embryo implantation rate and increased the number of viable pups per litter. In late gestation, fetal and placental weights were reduced in a litter size-dependent manner, and the canonical inverse relationship between litter size and fetal weight was lost. The duration of pregnancy was extended by ~ 0.5 day and birth weight was reduced by ~ 5% after prednisolone treatment. Viability of prednisolone-exposed offspring was comparable to controls, but body weight was altered in adulthood, particularly in male offspring. Thus, while prednisolone given in the pre-implantation phase in mice increases maternal receptivity to implantation and resource investment in fetal growth, there is a trade-off in long-term consequences for fetal development, birth weight and offspring health. These effects are associated with, and likely caused by, prednisolone suppression of the adaptive immune response at the outset of pregnancy.
Collapse
Affiliation(s)
- Tom Ec Kieffer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.,Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peck Y Chin
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ella S Green
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Lachlan M Moldenhauer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jelmer R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarah A Robertson
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
20
|
Cimadomo D, Craciunas L, Vermeulen N, Vomstein K, Toth B. Definition, diagnostic and therapeutic options in recurrent implantation failure: an international survey of clinicians and embryologists. Hum Reprod 2021; 36:305-317. [PMID: 33313697 DOI: 10.1093/humrep/deaa317] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION What is the global variability in recurrent implantation failure (RIF) definition, investigation and therapy, currently offered to patients undergoing IVF? SUMAMRY ANSWER Definitions, diagnostic investigations and treatments offered to RIF patients differ widely amongst assisted reproduction healthcare professionals and clinical guidelines on RIF are urgently needed. WHAT IS KNOWN ALREADY RIF affects around 10% of patients undergoing IVF worldwide. There is no consensus on the definition of RIF, its diagnostic investigations or the therapeutic options, which leads to inconsistencies in clinical practice. STUDY DESIGN, SIZE, DURATION A cross-sectional study of clinicians and embryologists was conducted between May and June 2020. The survey included 43 questions aimed at understanding participants' background and their current practice with regards to defining, investigating and managing RIF. The questions were designed by the European Society of Human Reproduction and Embryology (ESHRE) Special Interest Group (SIG) on implantation and early pregnancy following three consensus meetings. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 8579 ESHRE members from 6916 IVF centers were invited to participate using two global email calls based on their pre-specified interest in implantation and early pregnancy. SurveyMonkey and SPSS were used for data collection and analysis, respectively. Furthermore, differences were reported in the answers of European and non-European professionals, as well as between public and private settings and among clinicians clustered according to the average number of RIF patients treated per year. MAIN RESULTS AND THE ROLE OF CHANCE The final data set included 735 clinicians and 300 embryologist or IVF-biologists. The majority defines RIF based on the number of failed embryo transfers (ETs) with the most common threshold adopted being three ETs both fresh and frozen. More than two-thirds take lifestyle factors into account, mainly drugs, smoking and BMI. The highest consensus on which diagnostic investigations should be performed was reached for anatomical malformations and gynecological aspects focusing on hydrosalpinx, Asherman's syndrome, endometrial thickness and endometriosis. Concerning treatment of RIF patients, the highest consensus was reached for preconceptional therapies, including BMI adjustment, smoking and endometritis followed by therapies during IVF procedures. LIMITATIONS, REASONS FOR CAUTION The response rate was relatively low, but comparable to other surveys. WIDER IMPLICATIONS OF THE FINDINGS A consensus on definition, diagnosis and treatment of RIF would help to reduce costly, time-consuming and poorly validated approaches. STUDY FUNDING/COMPETING INTEREST(S) No external funding was used. B.T. received support from Bayer for Clinical trials concerning endometriosis and Ferring for clinical trials concerning ovarian stimulation. She received reimbursement for travel expenses from Astropharm, Ferring. Dr Kade and is a shareholder of Reprognostics. She is a board member of the Austrian Society for Obstetrics and Gynecology (OEGGG), the associate head of the 'Reproduktionsmedizinische Zentren Baden-Württemberg' (RZBW), a member of guideline group of the German Society for Obstetrics and Gynecology (DGGG) and an editorial board member of the following journals: American Journal of Reproductive Immunology (AJRI), Archives of Gynecology and Obstetrics. All the other authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- D Cimadomo
- Clinica Valle Giulia, Genera Center for Reproductive Medicine, Rome, Italy
| | - L Craciunas
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - K Vomstein
- Department of Gynecological Endocrinology and Reproductive Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - B Toth
- Department of Gynecological Endocrinology and Reproductive Medicine, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Zhou G, Zhou M, Duan X, Li W. Glucocorticoid supplementation improves reproductive outcomes in infertile women with antithyroid autoimmunity undergoing ART: A meta-analysis. Medicine (Baltimore) 2021; 100:e25554. [PMID: 33879707 PMCID: PMC8078294 DOI: 10.1097/md.0000000000025554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Thyroid autoimmune disease (TAI) has been verified to be related to multiple adverse pregnancy outcomes. A growing number of evidences highlight the protective roles of glucocorticoid on the treatments of TAI. This meta-analysis aimed to study whether it is beneficial to add glucocorticoid treatment in infertile women with TAI when they are undergoing assisted reproductive technology (ART). METHODS We conducted a systematic search in PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure (CNKI), WanFang database, Weipu China Science and Technology Journal Databases (VIP database) up to September 10, 2020. The Revman 5.3 software was utilized for data statistics. We used a random-effects model to analyze data and the odds ratio (OR) combining with 95% confidence interval (95% CI) were employed to reveal the results. RESULTS Three publications with 237 antithyroid antibody (ATA)-positive and 384 ATA-negative women were included in the final analysis. Overall, glucocorticoid therapy showed satisfying effects on improving clinical pregnancy rate (OR = 4.63, 95% CI [2.23, 9.58], I2 = 0.0%, P < .0001) and live birth rate (OR = 3.19, 95% CI [1.13, 9.04], I2 = 0.0%, P = .03) of ATA-positive women compared with control group. However, it seems that glucocorticoid showed no significant difference in the abortion rate (OR = 0.62, 95% CI [0.09, 4.32], I2 = 35%, P = .64) and oocyte recovery (OR = 2.26, 95% CI [-1.46, 5.99], I2 = 79%, P < .0001) between the 2 groups. CONCLUSIONS Glucocorticoid may improve the pregnancy outcomes of ART women with ATA positive, but there is no significant reduction in the risk of miscarriage. Due to the limited enrolled references, glucocorticoid adjuvant therapy should be applied after more randomized controlled trials.
Collapse
|
22
|
Koot YEM, Hviid Saxtorph M, Goddijn M, de Bever S, Eijkemans MJC, Wely MV, van der Veen F, Fauser BCJM, Macklon NS. What is the prognosis for a live birth after unexplained recurrent implantation failure following IVF/ICSI? Hum Reprod 2020; 34:2044-2052. [PMID: 31621857 DOI: 10.1093/humrep/dez120] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION What is the cumulative incidence of live birth and mean time to pregnancy (by conception after IVF/ICSI or natural conception) in women experiencing unexplained recurrent implantation failure (RIF) following IVF/ICSI treatment? SUMMARY ANSWER In 118 women who had experienced RIF, the reported cumulative incidence of live birth during a maximum of 5.5 years follow-up period was 49%, with a calculated median time to pregnancy leading to live birth of 9 months after diagnosis of RIF. WHAT IS KNOWN ALREADY Current definitions of RIF include failure to achieve a pregnancy following IVF/ICSI and undergoing three or more fresh embryo transfer procedures of one or two high quality embryos or more than 10 embryos transferred in fresh or frozen cycles. The causes and optimal management of this distressing condition remain uncertain and a range of empirical and often expensive adjuvant therapies is often advocated. Little information is available regarding the long-term prognosis for achieving a pregnancy. STUDY DESIGN, SIZE, DURATION Two hundred and twenty-three women under 39 years of age who had experienced RIF without a known cause after IVF/ICSI treatment in two tertiary referral university hospitals between January 2008 and December 2012 were invited to participate in this retrospective cohort follow up study. PARTICIPANTS/MATERIALS, SETTING, METHODS All eligible women were sent a letter requesting their consent to the anonymous use of their medical file data and were asked to complete a questionnaire enquiring about treatments and pregnancies subsequent to experiencing RIF. Medical files and questionnaires were examined and results were analysed to determine the subsequent cumulative incidence of live birth and time to pregnancy within a maximum 5.5 year follow-up period using Kaplan Meier analysis. Clinical predictors for achieving a live birth were investigated using a Cox hazard model. MAIN RESULTS AND THE ROLE OF CHANCE One hundred and twenty-seven women responded (57%) and data from 118 women (53%) were available for analysis. During the maximum 5.5 year follow up period the overall cumulative incidence of live birth was 49% (95% CI 39-59%). Among women who gave birth, the calculated median time to pregnancy was 9 months after experiencing RIF, where 18% arose from natural conceptions. LIMITATIONS, REASONS FOR CAUTION Since only 57% of the eligible study cohort completed the questionnaire, the risk of response bias limits the applicability of the study findings. WIDER IMPLICATIONS OF THE FINDINGS This study reports a favorable overall prognosis for achieving live birth in women who have previously experienced RIF, especially in those who continue with further IVF/ICSI treatments. However since 51% did not achieve a live birth during the follow-up period, there is a need to distinguish those most likely to benefit from further treatment. In this study, no clinical factors were found to be predictive of those achieving a subsequent live birth. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the University Medical Center Utrecht, in Utrecht and the Academic Medical Centre, in Amsterdam. NSM has received consultancy and speaking fees and research funding from Ferring, MSD, Merck Serono, Abbott, IBSA, Gedion Richter, and Clearblue. During the most recent 5-year period BCJMF has received fees or grant support from the following organizations (in alphabetic order); Actavis/Watson/Uteron, Controversies in Obstetrics & Gynecology (COGI), Dutch Heart Foundation, Dutch Medical Research Counsel (ZonMW), Euroscreen/Ogeda, Ferring, London Womens Clinic (LWC), Merck Serono, Myovant, Netherland Genomic Initiative (NGI), OvaScience, Pantharei Bioscience, PregLem/Gedeon Richter/Finox, Reproductive Biomedicine Online (RBMO), Roche, Teva, World Health Organisation (WHO).None of the authors have disclosures to make in relation to this manuscript.
Collapse
Affiliation(s)
- Y E M Koot
- Department of Reproductive Medicine and Gynaecology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - M Hviid Saxtorph
- Department of Obstetrics and Gynaecology, Zealand University Hospital, Roskilde, Denmark
| | - M Goddijn
- Centre for Reproductive Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - S de Bever
- Centre for Reproductive Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - M J C Eijkemans
- Department of Reproductive Medicine and Gynaecology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - M V Wely
- Centre for Reproductive Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - F van der Veen
- Centre for Reproductive Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - B C J M Fauser
- Department of Reproductive Medicine and Gynaecology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - N S Macklon
- Department of Reproductive Medicine and Gynaecology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Obstetrics and Gynaecology, Zealand University Hospital, Roskilde, Denmark.,London Women's Clinic, London, UK
| |
Collapse
|
23
|
Saxtorph MH, Hallager T, Persson G, Petersen KB, Eriksen JO, Larsen LG, Hviid TV, Macklon N. Assessing endometrial receptivity after recurrent implantation failure: a prospective controlled cohort study. Reprod Biomed Online 2020; 41:998-1006. [PMID: 32978074 DOI: 10.1016/j.rbmo.2020.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022]
Abstract
RESEARCH QUESTION What is the prevalence of disrupted markers of endometrial function among women experiencing recurrent implantation failure (RIF), and does the prevalence differ from a control cohort? DESIGN Prospective controlled cohort study. In total, 86 women with a history of RIF and 37 women starting their first fertility treatment were recruited for this study. Endometrial and blood profiling were carried out in a hormone-substituted cycle using oestradiol and progesterone. Endometrial biopsies were analysed by histology, immune cell profiling, and the endometrial receptivity array (ERA®) test (Igenomix, Valencia, Spain). The vaginal microbiome was analysed using a NGS-based technology (ArtPRED, Amsterdam, the Netherlands). Blood tests included oestradiol, progesterone, prolactin, thyroid-stimulating hormone, vitamin D and anti-phospholipid antibody levels. RESULTS Patients who had experienced RIF produced a range of test abnormalities. Compared with controls, women with RIF had a higher prevalence of chronic endometritis (24% versus 6%), a lower vitamin D level and a borderline lower progesterone level. Women who had experienced RIF had a more favourable vaginal microbiome compared with controls. Although the RIF cohort was older than the controls (mean age 33.8 years versus 30.2 years), no differences between the groups were observed in immune cell profiling and the ERA test. CONCLUSION These data demonstrate that a single test or treatment for the endometrial factor in RIF is unlikely to be clinically effective. Diagnosing the endometrium in women with RIF permits targeted rather than blind interventions. Relative vitamin D deficiency, lower mid-luteal progesterone and chronic endometritis are ready targets for treatment. Understanding the role and treatment of an unfavourable vaginal microbiome in RIF needs further investigation.
Collapse
Affiliation(s)
- Malene Hviid Saxtorph
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde/Køge and Department of Clinical Medicine, University of Copenhagen, Lykkebækvej 14, Køge 4600, Denmark; Reprohealth Research Consortium, Sygehusvej 10, 4000 Roskilde, DK.
| | - Trine Hallager
- Department of Pathology, Sygehusvej 9, 4000 Roskilde, DK
| | - Gry Persson
- Reprohealth Research Consortium, Sygehusvej 10, 4000 Roskilde, DK; Department of Clinical Biochemistry, Sygehusvej 10, 4000, Roskilde
| | - Kathrine Birch Petersen
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde/Køge and Department of Clinical Medicine, University of Copenhagen, Lykkebækvej 14, Køge 4600, Denmark; Reprohealth Research Consortium, Sygehusvej 10, 4000 Roskilde, DK; StorkKlinik, Store Kongensgade 40G, 1264 Copenhagen K, DK
| | - Jens Ole Eriksen
- Department of Surgical Pathology Sygehusvej 9, 4000 Roskilde, DK
| | | | - Thomas Vauvert Hviid
- Reprohealth Research Consortium, Sygehusvej 10, 4000 Roskilde, DK; Department of Clinical Biochemistry, Sygehusvej 10, 4000, Roskilde
| | - Nick Macklon
- Department of Gynaecology, Obstetrics, and Fertility, Zealand University Hospital, Roskilde/Køge and Department of Clinical Medicine, University of Copenhagen, Lykkebækvej 14, Køge 4600, Denmark; Reprohealth Research Consortium, Sygehusvej 10, 4000 Roskilde, DK; London Womens Clinic, 113-115 Harley St, Marylebone, London W1G 6AP, Great Britain
| |
Collapse
|
24
|
Recurrent implantation failure in IVF: A Canadian Fertility and Andrology Society Clinical Practice Guideline. Reprod Biomed Online 2020; 41:819-833. [PMID: 32962928 DOI: 10.1016/j.rbmo.2020.08.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/16/2020] [Accepted: 08/08/2020] [Indexed: 12/23/2022]
Abstract
Recurrent implantation failure (RIF) after IVF is a challenging topic for clinicians and can be a devastating reality for some patients with infertility. The purpose of this guideline from the Canadian Fertility and Andrology Society (CFAS) is to provide the most relevant evidence to date for the assessment and management of RIF. This guideline was developed using the GRADE (Grading of Recommendations, Assessment, Development and Evaluation) approach. This guideline recognizes the presence of heterogeneity in the definition of RIF. Recommendations are offered here on the investigation of RIF and management options that may increase the chance of a live birth.
Collapse
|
25
|
Lu Y, Yan J, Liu J, Tan J, Hong Y, Wei D, Chen ZJ, Sun Y. Prednisone for patients with recurrent implantation failure: study protocol for a double-blind, multicenter, randomized, placebo-controlled trial. Trials 2020; 21:719. [PMID: 32807234 PMCID: PMC7430107 DOI: 10.1186/s13063-020-04630-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/26/2020] [Indexed: 12/19/2022] Open
Abstract
Background Recurrent implantation failure (RIF) brings great challenges to clinicians and causes deep frustration to patients. Previous data has suggested that prednisone may play a promising role in the establishment of pregnancy and help improve the pregnancy outcome in women with RIF. But there is insufficient evidence from randomized clinical trials that had adequate power to determine if prednisone can enhance live births as the primary outcome. Methods/design This trial is a prospective, multicenter, randomized, double-blind, placebo-controlled clinical trial (1:1 ratio of prednisone versus placebo). Infertile patients with RIF who intend to undergo frozen-thawed embryo transfer (FET) after in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) or pre-implantation genetic testing for aneuploidy (PGT-A) will be enrolled and randomly assigned to two parallel groups. Participants will be given the treatment of prednisone or placebo from the start of endometrial preparation till the end of the first trimester of pregnancy if pregnant. The primary outcome is live birth rate. Discussion The results of this study will provide evidence for the effect of prednisone on pregnancy outcomes in patients with RIF. Trial registration Chinese Clinical Trial Registry, ChiCTR1800018783. Registered on 9 October 2018.
Collapse
Affiliation(s)
- Yao Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 845 Lingshan Road, Pudong New District, Shanghai, 200135, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Jiayin Liu
- Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jichun Tan
- Reproductive Medical Center, Obstetrics and Gynecology Department, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yan Hong
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 845 Lingshan Road, Pudong New District, Shanghai, 200135, China
| | - Daimin Wei
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 845 Lingshan Road, Pudong New District, Shanghai, 200135, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 845 Lingshan Road, Pudong New District, Shanghai, 200135, China.
| |
Collapse
|
26
|
Plowden TC, Connell MT, Hill MJ, Mendola P, Kim K, Nobles CJ, Kuhr DL, Galai N, Gibbins KJ, Silver RM, Wilcox B, Sjaarda L, Perkins NJ, Schisterman EF, Mumford SL. Family history of autoimmune disease in relation to time-to-pregnancy, pregnancy loss, and live birth rate. J Transl Autoimmun 2020; 3:100059. [PMID: 32743539 PMCID: PMC7388372 DOI: 10.1016/j.jtauto.2020.100059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/27/2022] Open
Abstract
Several autoimmune conditions have adverse effects on reproductive outcomes, but the relationship between family history of autoimmune disease in women without these conditions and pregnancy is uncertain. The objective of this study was to determine if there is an association between a family history of an autoimmune condition and time-to-pregnancy (TTP), pregnancy loss, and live birth. This was a prospective cohort study from a RCT of 1228 adult women ages 18-40, who were healthy, had no history of infertility, were actively attempting to conceive, and had one or two prior pregnancy losses. Of these, 1172 women had data available regarding family history of autoimmune conditions. Women with an affected first-degree relative had similar TTP when compared to those without a FHx (fecundability odds ratio 0.90, 95% confidence interval [CI] 0.70, 1.15). Women with an affected first-degree relative had a lower likelihood of live birth (relative risk [RR] 0.83, 95% CI 0.69, 0.99). Among women who achieved pregnancy, FHx of autoimmune disease was associated with a higher likelihood of pregnancy loss (RR 1.49, 95% CI 1.10, 2.03). Women who had a first-degree relative with an autoimmune disease had a similar TTP as unaffected women but a lower likelihood of live birth and higher risk of pregnancy loss. This information may encourage clinicians to evaluate women with a family history of autoimmune conditions prior to pregnancy and highlights the need for further studies to ascertain the effects of autoimmunity and pregnancy.
Collapse
Affiliation(s)
- Torie C Plowden
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Matthew T Connell
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Micah J Hill
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Pauline Mendola
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Keewan Kim
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Carrie J Nobles
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Daniel L Kuhr
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Department of Obstetrics and Gynecology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Mail Stop MAC 5034, Cleveland, OH 44106, United States
| | - Noya Galai
- Department of Statistics, University of Haifa, Mt Carmel, Haifa, 31905, Israel
| | - Karen J Gibbins
- OB/GYN, University of Utah, Salt Lake City, UT, United States
| | - Robert M Silver
- OB/GYN, University of Utah, Salt Lake City, UT, United States
| | - Brian Wilcox
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Room 2B200 SOM, 50 North Medical Drive, Salt Lake City, UT 84132, United States
| | - Lindsey Sjaarda
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Neil J Perkins
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Enrique F Schisterman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Sunni L Mumford
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| |
Collapse
|
27
|
Liu S, Yu Y, Zhang S, Cole JB, Tenesa A, Wang T, McDaneld TG, Ma L, Liu GE, Fang L. Epigenomics and genotype-phenotype association analyses reveal conserved genetic architecture of complex traits in cattle and human. BMC Biol 2020; 18:80. [PMID: 32620158 PMCID: PMC7334855 DOI: 10.1186/s12915-020-00792-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 05/12/2020] [Indexed: 02/01/2023] Open
Abstract
Background Lack of comprehensive functional annotations across a wide range of tissues and cell types severely hinders the biological interpretations of phenotypic variation, adaptive evolution, and domestication in livestock. Here we used a combination of comparative epigenomics, genome-wide association study (GWAS), and selection signature analysis, to shed light on potential adaptive evolution in cattle. Results We cross-mapped 8 histone marks of 1300 samples from human to cattle, covering 178 unique tissues/cell types. By uniformly analyzing 723 RNA-seq and 40 whole genome bisulfite sequencing (WGBS) datasets in cattle, we validated that cross-mapped histone marks captured tissue-specific expression and methylation, reflecting tissue-relevant biology. Through integrating cross-mapped tissue-specific histone marks with large-scale GWAS and selection signature results, we for the first time detected relevant tissues and cell types for 45 economically important traits and artificial selection in cattle. For instance, immune tissues are significantly associated with health and reproduction traits, multiple tissues for milk production and body conformation traits (reflecting their highly polygenic architecture), and thyroid for the different selection between beef and dairy cattle. Similarly, we detected relevant tissues for 58 complex traits and diseases in humans and observed that immune and fertility traits in humans significantly correlated with those in cattle in terms of relevant tissues, which facilitated the identification of causal genes for such traits. For instance, PIK3CG, a gene highly specifically expressed in mononuclear cells, was significantly associated with both age-at-menopause in human and daughter-still-birth in cattle. ICAM, a T cell-specific gene, was significantly associated with both allergic diseases in human and metritis in cattle. Conclusion Collectively, our results highlighted that comparative epigenomics in conjunction with GWAS and selection signature analyses could provide biological insights into the phenotypic variation and adaptive evolution. Cattle may serve as a model for human complex traits, by providing additional information beyond laboratory model organisms, particularly when more novel phenotypes become available in the near future.
Collapse
Affiliation(s)
- Shuli Liu
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, BARC-East, Beltsville, MD, 20705, USA.,College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ying Yu
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shengli Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - John B Cole
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, BARC-East, Beltsville, MD, 20705, USA
| | - Albert Tenesa
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK.,The Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tara G McDaneld
- US Meat Animal Research Center, Agricultural Research Service, USDA, Clay Center, NE, 68933, USA
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, BARC-East, Beltsville, MD, 20705, USA.
| | - Lingzhao Fang
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, BARC-East, Beltsville, MD, 20705, USA. .,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK. .,Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
28
|
Sacks G, Finkelstein E. Natural killer cells and reproductive success. Am J Reprod Immunol 2020; 85:e13291. [DOI: 10.1111/aji.13291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Gavin Sacks
- IVFAustralia Sydney Australia
- University of New South Wales Sydney Australia
- St George hospital and Royal Hospital for Women Sydney Australia
| | | |
Collapse
|
29
|
Lédée N, Petitbarat M, Prat-Ellenberg L, Dray G, Cassuto GN, Chevrier L, Kazhalawi A, Vezmar K, Chaouat G. Endometrial Immune Profiling: A Method to Design Personalized Care in Assisted Reproductive Medicine. Front Immunol 2020; 11:1032. [PMID: 32582163 PMCID: PMC7287127 DOI: 10.3389/fimmu.2020.01032] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/29/2020] [Indexed: 11/20/2022] Open
Abstract
Objective: To assess the efficiency of the endometrial immune profiling as a method to design personalized care to enhance the pregnancy rate in a large heterogeneous infertile population. We hypothesized that some reproductive failures could be induced by a uterine immune dysregulation which could be identified and corrected with a targeted plan. Design: Prospective cohort study. Setting: Multicentric study. Intervention(s) and Main outcome measure(s): One thousand and seven hundred thirty-eight infertile patients had an immune profiling on a timed endometrial biopsy between 2012 and 2018. This test documented the absence or the presence of an endometrial immune dysregulation and identified its type. In case of dysregulation, a targeted personalized plan was suggested to the treating clinician aiming to supply the anomaly. One year after the test, the clinician was contacted to provide the outcome of the subsequent embryo transfer with the applied suggested plan. Result(s): After testing, 16.5% of the patients showed no endometrial immune dysregulation, 28% had a local immune under-activation, 45% had a local immune over-activation, and 10.5% had a mixed endometrial immune profile. In patients with a history of repeated implantation failures (RIF) or recurrent miscarriages (RM), the pregnancy rate was significantly higher if an endometrial dysregulation was found and the personalized plan applied, compared to the patients with an apparent balanced immune profile (respectively 37.7 and 56% vs. 26.9 and 24%, p < 0.001). In contrast, in good prognosis IVF (in vitro fertilization) subgroup and patients using donor eggs, this difference was not significant between dysregulated and balanced subgroups, but higher pregnancy rates were observed in absence of dysregulation. For patients with immune over-activation, pregnancy rates were significantly higher for patients who had a test of sensitivity, regarding the type of immunotherapy introduced, when compared to the ones who did not (51 vs. 39.9%, p = 0.012). Conclusion(s): Local endometrial immunity appears to be a new and important parameter able to influence the prognosis of pregnancy. Targeted medical care in case of local immune dysregulation resulted in significantly higher pregnancy rates in RIF and RM patients.
Collapse
Affiliation(s)
- Nathalie Lédée
- MatriceLAB Innove, Pépinière Paris Santé Cochin, Hôpital Cochin, Paris, France
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, Paris, France
| | - Marie Petitbarat
- MatriceLAB Innove, Pépinière Paris Santé Cochin, Hôpital Cochin, Paris, France
| | - Laura Prat-Ellenberg
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, Paris, France
| | - Géraldine Dray
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, Paris, France
| | - Guy N. Cassuto
- Centre d'Assistance Médicale à la Procréation, Hôpital des Bluets, Paris, France
- Laboratoire Drouot, Paris, France
| | - Lucie Chevrier
- MatriceLAB Innove, Pépinière Paris Santé Cochin, Hôpital Cochin, Paris, France
| | - Alaa Kazhalawi
- MatriceLAB Innove, Pépinière Paris Santé Cochin, Hôpital Cochin, Paris, France
| | - Katia Vezmar
- MatriceLAB Innove, Pépinière Paris Santé Cochin, Hôpital Cochin, Paris, France
| | | |
Collapse
|
30
|
Persson G, Bork JBS, Isgaard C, Larsen TG, Bordoy AM, Bengtsson MS, Hviid TVF. Cytokine stimulation of the choriocarcinoma cell line JEG-3 leads to alterations in the HLA-G expression profile. Cell Immunol 2020; 352:104110. [PMID: 32387976 DOI: 10.1016/j.cellimm.2020.104110] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/16/2020] [Accepted: 04/05/2020] [Indexed: 12/13/2022]
Abstract
The checkpoint molecule human leukocyte antigen (HLA)-G has restricted tissue expression, and plays a role in the establishment of maternal tolerance to the semi-allogenic fetus during pregnancy by expression on the trophoblast cells in the placenta. HLA-G exists in at least seven well-described mRNA isoforms, of which four are membrane-bound and three soluble. Regulation of the tissue expression of HLA-G and its isoforms is relatively unknown. Therefore, it is important to understand the regulation of HLA-G, and the HLA-G+ choriocarcinoma cell line JEG-3 is a widely used cellular model. We hypothesized that cytokines present in the microenvironment can regulate the HLA-G expression profile. In the present study, we systematically stimulated JEG-3 cells with various concentrations of IL-2, IL-4 IL-6, IL-10, IL-12, IL-15, IL-17A, TGF-β1, TNF-α and IFN-γ1b. The results suggest that IFN-γ plays a role in maintenance of HLA-G expression, while IL-10 might be involved in regulation of the isoform profile.
Collapse
Affiliation(s)
- Gry Persson
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Julie Birgit Siig Bork
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Cecilie Isgaard
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Tine Graakjær Larsen
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Anna Maria Bordoy
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Meghan Sand Bengtsson
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Thomas Vauvert Faurschou Hviid
- Center for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and the Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
31
|
Ali S, Majid S, Niamat Ali M, Taing S. Evaluation of T cell cytokines and their role in recurrent miscarriage. Int Immunopharmacol 2020; 82:106347. [PMID: 32143004 DOI: 10.1016/j.intimp.2020.106347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 02/08/2020] [Accepted: 02/23/2020] [Indexed: 01/03/2023]
Abstract
Recurrent miscarriage (RM) is defined as two or more consecutive pregnancy losses that affect approximately 5% of conceived women worldwide. RM is a multi-factorial reproductive problem and has been associated with parental chromosomal abnormalities, embryonic chromosomal rearrangements, uterine anomalies, autoimmune disorders, endocrine dysfunction, thrombophilia, life style factors, and maternal infections. However, the exact cause is still undecided in remaining 50% of cases. Immunological rejection of the embryo due to exacerbated maternal immune reaction against paternal embryonic antigens has been set forth as one of the significant reason for RM. The accurate means that shield the embryo during normal pregnancy from the attack of maternal immune network and dismissal are inadequately implicit. However, it is suggested that the genetically irreconcilable embryo escapes maternal immune rejection due to communication among many vital cytokines exuded at maternal-embryonic interface both by maternal and embryonic cells. Previous investigations suggested the Th1/Th2 dominance in altered immunity of RM patients, according to which the allogenic embryo flees maternal T cell reaction by inclining the Th0 differentiation toward Th2 pathway resulting into diminished pro-inflammatory Th1 immunity. However, recently pro-inflammatory Th17 cells and immunoregulatory Treg cells have been discovered as essential immune players in RM besides Th1/Th2 components. Cytokines are believed to develop a complicated regulatory network so as to establish a state of homeostasis between the semi-allogenic embryo and the maternal immune system. However, an adverse imbalance among cytokines at maternal-embryonic interface perhaps due to their gene polymorphisms may render immunoregulatory means not enough to re-establish homeostasis and thus may collapse pregnancy.
Collapse
Affiliation(s)
- Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, 190006 Srinagar, J&K, India
| | - Sabhiya Majid
- Department of Biochemistry, Government Medical College, Srinagar, J&K, India
| | - Md Niamat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, 190006 Srinagar, J&K, India.
| | - Shahnaz Taing
- Department of Obstetrics and Gynaecology, Government Medical College Associated Lalla Ded Hospital, Srinagar, J&K, India
| |
Collapse
|
32
|
Min Y, Wang X, Chen H, Yin G. The exploration of Hashimoto's Thyroiditis related miscarriage for better treatment modalities. Int J Med Sci 2020; 17:2402-2415. [PMID: 33029083 PMCID: PMC7532476 DOI: 10.7150/ijms.48128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/21/2020] [Indexed: 12/25/2022] Open
Abstract
Hashimoto's thyroiditis (HT) is the most prevalent autoimmune thyroid disease (ATD) worldwide and is strongly associated with miscarriage and even recurrent miscarriage (RM). Moreover, with a deepening understanding, emerging evidence has shown that immune dysfunctions caused by HT conditions, including imbalanced subsets of CD4+ T-helper cells, B regulatory (Breg) cells, high expression levels of CD56dim natural killer (NK) cells, and cytokines, possibly play an important role in impairing maternal tolerance to the fetus. In recent years, unprecedented progress has been made in recognizing the specific changes in immune cells and molecules in patients with HT, which will be helpful in exploring the mechanism of HT-related miscarriage. Based on these findings, research investigating some potentially more effective treatments, such as selenium (Se), vitamin D3, and intravenous immunoglobulin (IVIG), has been well developed over the past few years. In this review, we highlight some of the latest advances in the possible immunological pathogenesis of HT-related miscarriage and focus on the efficacies of treatments that have been widely introduced to clinical trials or practice described in the most recent literature.
Collapse
Affiliation(s)
- Yu Min
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Rd, Yuzhong Dist, Chongqing 404100, China
| | - Xing Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Rd, Yuzhong Dist, Chongqing 404100, China
| | - Hang Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Rd, Yuzhong Dist, Chongqing 404100, China
| | - Guobing Yin
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Rd, Yuzhong Dist, Chongqing 404100, China
| |
Collapse
|
33
|
Ehrlich R, Hull ML, Walkley J, Sacks G. Intralipid Immunotherapy for Repeated IVF Failure. FERTILITY & REPRODUCTION 2019. [DOI: 10.1142/s2661318219500178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The intravenous fat emulsion, intralipid, has been hypothesised to be an effective and safe treatment for repeated in vitro fertilisation (IVF), implantation failure and pregnancy loss. This exploratory, retrospective cohort study determined pregnancy outcomes and documented adverse events associated with intralipid use. Ninety-three women were identified as having received intralipid for a history of repeated unsuccessful IVF cycles and pre-viable pregnancy loss in two Australian IVF units that independently recruited between October 2014 and July 2016. Pregnancy outcomes and adverse events were recorded in fresh and frozen embryo transfer cycles in which the infusion was administered. The 93 women who received intralipid had a clinical pregnancy rate of 40.0%, compared with 35.0% in 651 age-matched controls undergoing IVF, which was not significantly different. The intralipid group had a livebirth rate of 35.7%. Apart from flushing, which was experienced by one individual, there were no adverse events associated with intralipid use. As a prelude to definitive evidence of benefit, we did not identify a safety concern or reduced pregnancy rates in intralipid users compared to controls. Indeed, these outcomes were better than expected in a poor prognosis group. This data supports an argument for large, randomised controlled trials to determine the benefit of intralipid in the treatment of recurrent implantation failure or miscarriage.
Collapse
Affiliation(s)
- Romy Ehrlich
- University of New South Wales, Sydney, Australia
| | - M. Louise Hull
- Robinson Research Institute, University of Adelaide, Frome Road, Adelaide, South Australia, Australia
- FertilitySA, Adelaide, South Australia, Australia
- Embrace Fertility, Adelaide, South Australia, Australia
| | - Jane Walkley
- FertilitySA, Adelaide, South Australia, Australia
- Embrace Fertility, Adelaide, South Australia, Australia
| | - Gavin Sacks
- University of New South Wales, Sydney, Australia
- IVFAustralia, Bondi, Sydney, Australia
| |
Collapse
|
34
|
Should we stop offering endometrial scratching prior to in vitro fertilization? Fertil Steril 2019; 111:1094-1101. [PMID: 31155116 DOI: 10.1016/j.fertnstert.2019.04.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
|
35
|
Dieamant F, Vagnini LD, Petersen CG, Mauri AL, Renzi A, Petersen B, Mattila MC, Nicoletti A, Oliveira JBA, Baruffi R, Franco Jr. JG. New therapeutic protocol for improvement of endometrial receptivity (PRIMER) for patients with recurrent implantation failure (RIF) - A pilot study. JBRA Assist Reprod 2019; 23:250-254. [PMID: 31091064 PMCID: PMC6724389 DOI: 10.5935/1518-0557.20190035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/04/2019] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To evaluate whether or not one should use a new Protocol for Endometrial Receptivity Improvement (PRIMER) based on platelet-rich plasma (PRP) and granulocyte colony-stimulation factor (G-CSF) to enhance ongoing pregnancy rates in patients with recurrent implantation failure (RIF). METHODS Women undergoing IVF/ICSI were prospectively divided into two groups: - PRIMER/RIF group (n:33): patients with RIF (defined as ≥2embryo transfers (ETs) and at least 5 morphologically good embryos transferred) in which intrauterine PRP injection and subcutaneous G-CSF-injection were performed. - Control group (n:33): patients in their first IVF/ICSI attempt/cycle (without PRP or G-CSF injection). The PRP was prepared using autologous fresh-whole blood processed to increase platelet-concentration in 2 to 4 fold. All patients undergoing the PRP-treatment received 0.7ml of it through intrauterine-injection 48 hours before the ET. G-CSF (300mg/0.5ml) started simultaneously to PRP and was administered subcutaneously every week. RESULTS Regarding implantation, clinical pregnancy and miscarriage rates, we found no statistically significant difference (18.2% versus 17.6%, p=0.90; 36.4% versus 30.3%, p=0.61 and 25.0% versus 9.0%, p=0.43, respectively). The use of PRIMER enabled RIF patients (previous ET µ: 4.0±1.5) to reach similar ongoing pregnancy and live birth rates like those patients who had their first IVF/ICSI cycle attempt (27.3% versus 27.3%, p=0.99). CONCLUSIONS Our results showed, for the first time, evidence that this therapeutic protocol (PRIMER) could be used as a feasible treatment based on biological rationale for patients with RIF, considering its promising outcomes, it is a simple procedure and not associated with patient complications.
Collapse
Affiliation(s)
- Felipe Dieamant
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Laura D. Vagnini
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Claudia G. Petersen
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Ana L. Mauri
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Adriana Renzi
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Bruna Petersen
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | | | - Andreia Nicoletti
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Joao Batista A. Oliveira
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Ricardo Baruffi
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| | - Jose G. Franco Jr.
- Center for Human Reproduction Prof Franco Jr. Ribeirão
Preto, Brazil
- Paulista Center for Diagnosis Research and Training.
Ribeirão Preto, Brazil
| |
Collapse
|
36
|
Yakin K, Oktem O, Urman B. Intrauterine administration of peripheral mononuclear cells in recurrent implantation failure: a systematic review and meta-analysis. Sci Rep 2019; 9:3897. [PMID: 30846784 PMCID: PMC6405957 DOI: 10.1038/s41598-019-40521-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 02/04/2019] [Indexed: 12/20/2022] Open
Abstract
It has been proposed that intrauterine administration of peripheral blood mononuclear cells (PBMCs) modulates maternal immune response through a cascade of cytokines, chemokines and growth factors to favor implantation. We conducted a meta-analysis to verify the effect of intrauterine PBMC administration on the outcome of embryo transfer in women with recurrent implantation failure (RIF). All relevant trials published in PubMed, Web of Science and Cochrane library databases were searched. Two randomized controlled trials and three cohort studies (1173 patients in total) matched the inclusion criteria. No differences in live birth rates were seen between the PBMC-treated patients and controls (OR: 1.65, 95% CI: 0.84–3.25; p = 0.14; I2: 66.3%). The clinical pregnancy rate was significantly higher in women who received intrauterine PBMCs before embryo transfer compared with those who did not (OR: 1.65, 95% CI: 1.30–2.10; p = 0.001, heterogeneity; I2: 60.6%). Subgroup analyses revealed a significant increase in clinical pregnancy rates with the administration of PBMCs in women with ≥3 previous failures compared with controls (OR: 2.69, 95% CI: 1.53–4.72; p = 0.001, I2: 38.3%). In summary, the data did not demonstrate an association between the administration of PBMCs into the uterine cavity before fresh or frozen-thawed embryo transfer and live birth rates in women with RIF. Whether intrauterine PBMC administration significantly changes live birth and miscarriage rates requires further investigation.
Collapse
Affiliation(s)
- Kayhan Yakin
- Department of Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey.
| | - Ozgur Oktem
- Department of Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey
| | - Bulent Urman
- Department of Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
37
|
Altmäe S, Aghajanova L. Growth Hormone and Endometrial Receptivity. Front Endocrinol (Lausanne) 2019; 10:653. [PMID: 31616379 PMCID: PMC6768942 DOI: 10.3389/fendo.2019.00653] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
Administration of growth hormone (GH) during ovarian stimulation has shown beneficial effects on in vitro fertilization (IVF) outcomes. It is generally believed that this improvement is due to the stimulating effect of GH on oocyte quality. However, studies are emerging that show possible positive effect of GH administration on endometrial receptivity, thus suggesting an additional potential benefit at the level of the uterus, especially among women with recurrent implantation failure, thin endometrium, and older normal responders. This review summarizes recent data on GH co-treatment effects on endometrium and endometrial receptivity among infertile women undergoing IVF, and proposes possible mechanisms of GH actions in the endometrium.
Collapse
Affiliation(s)
- Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Competence Centre on Health Technologies, Tartu, Estonia
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- *Correspondence: Signe Altmäe
| | - Lusine Aghajanova
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford School of Medicine, Sunnyvale, CA, United States
| |
Collapse
|
38
|
Moving from peripheral blood to local uterine immunophenotype analysis in patients with poor reproductive history: pilot study of a novel technique. Ir J Med Sci 2018; 188:893-901. [DOI: 10.1007/s11845-018-1933-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/13/2018] [Indexed: 12/29/2022]
|
39
|
Harrity C, Shkrobot L, Walsh D, Marron K. ART implantation failure and miscarriage in patients with elevated intracellular cytokine ratios: response to immune support therapy. FERTILITY RESEARCH AND PRACTICE 2018; 4:7. [PMID: 30349731 PMCID: PMC6192160 DOI: 10.1186/s40738-018-0052-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/27/2018] [Indexed: 11/10/2022]
Abstract
Background The origins of adverse reproductive outcome can be multifactorial, but the contribution of the maternal immune system is considered debatable. Elevated intracellular cytokine ratios have been proposed, although not universally supported, as a marker for immunological dysfunction in implantation and early pregnancy. Poor patient selection or inadequate treatment or testing may be confounding factors. Specific immunomodulation, in carefully selected sub-populations of ART patients with poor reproductive history, despite transfer of good quality blastocysts, may potentially improve clinical outcomes. Methods Intracellular cytokine ratios (CKR) were prospectively assessed in 337 patients presenting with a history of implantation failure and/or pregnancy loss, prior to further treatment, and were found to be elevated in 150 (44.5%). Of this group, 134 agreed to initiate a standardised immunotherapy regime (nutraceuticals, prednisolone & intralipids) to evaluate the efficacy of this proposed therapy. Of the intervention population, a small cohort (n = 70) delayed commencing ART for ~ 10 weeks to assess if extended pre-treatment nutraceutical supplementation could normalise CKRs prior to starting ART, and if this conferred additional benefit. Results Baseline assessment in the intervention population (n = 134) identified 160 miscarriages from 180 total pregnancies (89% miscarriage rate, MR), conceived both spontaneously and by assisted reproduction. Post-treatment analysis of subsequent ART cycles revealed a significant improvement in both implantation (OR 3.0, 2.0-4.5) and miscarriage rates (41/97, 42.2% MR, P < 0.001). Interestingly, pre-treatment normalisation of CKRs appeared to impart marginal extra benefit prior to subsequent fertility treatment with immunotherapy. Conclusions Following immunomodulation, significant improvements in both implantation rate and miscarriage rate were seen in this poor prognosis population. This suggests a possible role for both detailed immuno-evaluation of patients with poor reproductive history with good embryo quality, and application of personalised immunotherapy regimes alongside ART in selected cases. Future randomised controlled trials are needed to definitively evaluate this potentially promising therapeutic approach.
Collapse
Affiliation(s)
- Conor Harrity
- 1Royal College of Surgeons Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Lyuda Shkrobot
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin 14, Ireland
| | - David Walsh
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin 14, Ireland
| | - Kevin Marron
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin 14, Ireland
| |
Collapse
|
40
|
Marron K, Walsh D, Harrity C. Detailed endometrial immune assessment of both normal and adverse reproductive outcome populations. J Assist Reprod Genet 2018; 36:199-210. [PMID: 30194617 DOI: 10.1007/s10815-018-1300-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/28/2018] [Indexed: 10/28/2022] Open
Abstract
PURPOSE Using a comprehensive flow cytometric panel, do endometrial immune profiles in adverse reproductive outcomes such as repeat implantation failure (RIF) and repeat pregnancy loss (RPL) differ from each other and male-factor controls? METHODS Six-hundred and twelve patients had an endometrial biopsy to assess the immunophenotype. History on presentation was used to subdivide the population into recurrent implantation failure (RIF) [n = 178], recurrent pregnancy loss (RPL) [n = 155], primary infertility [n = 130] and secondary infertility [n = 114]. A control group was utilised for comparative purposes [n = 35] and lymphocyte subpopulations were described. RESULTS Distinct lymphocyte percentage differences were noted across the populations. Relative to controls and RPL, patients with a history of RIF had significantly raised uterine NKs (53.2 vs 45.2 & 42.9%, p < 0.0001). All sub-fertile populations had increased percentage peripheral type NKs (p = 0.001), and exhibited increased CD69+ activation (p = 0.005), higher levels of B cells (p < 0.001), elevated CD4:CD8 ratio (p < 0.0001), lower T-regs (p = 0.034) and a higher proportion of Th1+ CD4s (p = 0.001). Patient aetiology confers some distinct findings, RPL; pNK, Bcells and CD4 elevated; RIF; uNK and CD56 raised while CD-8 and NK-T lowered. CONCLUSIONS Flow cytometric endometrial evaluation has the ability to provide a rapid and objective analysis of lymphocyte subpopulations. The findings show significant variations in cellular proportions of immune cells across the patient categories relative to control tissue. The cell types involved suggest that a potential differential pro-inflammatory bias may exist in patients with a history of adverse reproductive outcomes. Immunological assessment in appropriate populations may provide insight into the underlying aetiology of some cases of reproductive failure.
Collapse
Affiliation(s)
- Kevin Marron
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin, 14, Ireland.
| | - David Walsh
- Sims IVF Clinic, Clonskeagh Road, Clonskeagh, Dublin, 14, Ireland
| | | |
Collapse
|
41
|
Prins JR, Holvast F, van 't Hooft J, Bos AF, Ganzevoort JW, Scherjon SA, Robertson SA, Gordijn SJ. Development of a core outcome set for immunomodulation in pregnancy (COSIMPREG): a protocol for a systematic review and Delphi study. BMJ Open 2018; 8:e021619. [PMID: 30082354 PMCID: PMC6078247 DOI: 10.1136/bmjopen-2018-021619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION To establish pregnancy, the maternal immune system must adapt to tolerate the semiallogenic fetus. Less than optimal adaptation of the maternal immune system during (early) pregnancy is implicated in several complications of pregnancy. The development of effective immune modulation interventions as preventive or therapeutic strategies for pregnancy complications holds promise. Several studies sought to evaluate the safety and effectiveness of various approaches. However, a limitation is the high variability in clinical and immune outcomes that are reported. We, therefore, aim to develop a core outcome set for application to studies of immune modulation in pregnancy (COSIMPREG). METHODS AND ANALYSIS We will use a stepwise approach to develop a COSIMPREG. First, we will perform a systematic review to identify reported outcomes. For this review, Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines will be followed. Second, we will use the Delphi method to develop a preliminary COSIMPREG. In three rounds, the outcomes of the systematic review will be scored. A panel comprising experts from relevant disciplines and diverse geographical locations will be assembled until a sufficient quality of the panel is reached. We will use predefined decision rules for outcomes. After each round outcomes, including scores, will be returned to the panel for further refinement. The outcomes not excluded after the third round will be taken to a consensus meeting. In this meeting, experts from all relevant disciplines will discuss and finalise the COSIMPREG. ETHICS AND DISSEMINATION For this study ethical approval is not required. The systematic review will be published in an appropriate open access reproductive immunology journal. Once the COSIMPREG is finalised, it will be published in an open access reproductive immunology journal, and disseminated at appropriate international meetings, as well as through relevant research and scientific societies. Experts involved in the Delphi study will be asked to give informed consent.
Collapse
Affiliation(s)
- Jelmer R Prins
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floor Holvast
- Department of General Practice, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janneke van 't Hooft
- Department of Obstetrics and Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arend F Bos
- Division of Neonatology, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Groningen, The Netherlands
| | - Jan Willem Ganzevoort
- Department of Obstetrics and Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sicco A Scherjon
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sanne J Gordijn
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
42
|
Kofod L, Lindhard A, Hviid TVF. Implications of uterine NK cells and regulatory T cells in the endometrium of infertile women. Hum Immunol 2018; 79:693-701. [PMID: 29990511 DOI: 10.1016/j.humimm.2018.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 02/03/2023]
Abstract
A range of studies have shown that the complex process of implantation and an establishment of a pregnancy also involves immune factors. Disturbances in these underlying immune mechanisms might lead to implantation and pregnancy failure and may be involved in the pathogenesis of unexplained infertility. Several studies have reported that imbalances in uterine NK (uNK) cell abundance are associated with infertility; however, controversies exist. An increased amount of CD56+ uNK cells along with a decrease in CD16+ uNK cells have been associated with normal fertility in some studies. Very few studies of FoxP3+ regulatory T cells (Tregs) in the pre-implantation endometrium have been performed. Results are sparse and controversial, studies reporting both increased and decreased numbers of Tregs, respectively, in women suffering from infertility. In conclusion, studies imply that uNK cells, Tregs and HLA-G carry pivotal roles regarding the establishment of a healthy pregnancy, and that abnormal immune mechanisms involving these parameters may be associated with infertility. However, more research in early phases of the reproductive cycle, such as investigating the conditions in the endometrium before implantation, is needed to further clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Louise Kofod
- Centre for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Research Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Anette Lindhard
- The Fertility Clinic, The ReproHealth Research Consortium ZUH, Department of Gynaecology and Obstetrics, Zealand University Hospital, and Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Thomas Vauvert F Hviid
- Centre for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Research Consortium ZUH, Department of Clinical Biochemistry, Zealand University Hospital, and Department of Clinical Medicine, University of Copenhagen, Denmark.
| |
Collapse
|
43
|
Recurrent implantation failure is a pathology with a specific transcriptomic signature. Fertil Steril 2017; 108:9-14. [DOI: 10.1016/j.fertnstert.2017.05.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/30/2022]
|