1
|
Ma W, Ye S, Tian L, Liu M, Wang R, Yang X, Wang M, Fu F, Ren W, Dang L, Wang T, Wang W, Wang S, Sun Y, Li Y. Association of LHCGR rs2293275 genotype with ovarian aging in Chinese women: a multicenter population-based study. Reprod Biol Endocrinol 2025; 23:41. [PMID: 40089715 PMCID: PMC11909885 DOI: 10.1186/s12958-025-01375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
OBJECTIVE To evaluate the association between the LHCGR rs2293275 (N312S) genotype and ovarian aging phenotypes in Han Chinese women, focusing on diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI). STUDY DESIGN This multicenter population-based study included 1,240 women aged 18-40 years diagnosed with DOR (n = 711) or POI (n = 529), alongside 72,846 ethnically and regionally matched controls from the Han Chinese Genomes Database (PGG.Han). Genotyping of rs2293275 was performed, and clinical data (menstrual history, hormonal profiles, maternal menopause age, and ART outcomes) were analyzed. MAIN RESULTS The AA genotype frequency in the ovarian aging cohort (1.85%) was significantly higher than in the general Han population (0.62%, OR 3.04, 95% CI 1.99-4.64, p < 0.001). AA carriers exhibited earlier POI diagnosis (25.5 ± 6.4 vs. 32.0 ± 5.1 years in GG carriers, p < 0.001) and maternal menopause (41.6 ± 3.3 vs. 47.8 ± 4.1 years, p < 0.001). In controlled ovarian stimulation cycles, AA carriers demonstrated reduced ovarian sensitivity (OSI: 3.59 vs. 1.21 in GG, p = 0.019) despite comparable gonadotropin doses. CONCLUSIONS The LHCGR rs2293275 AA genotype is strongly associated with accelerated ovarian aging in Han Chinese women, highlighting its potential as a biomarker for early identification of high-risk individuals. While these findings underscore genetic contributions to ovarian dysfunction, further mechanistic studies are needed to establish causality and optimize clinical translation. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT05665010, registered on 2022-11-30.
Collapse
Affiliation(s)
- Wenqing Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuangmei Ye
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lifeng Tian
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Women's Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, 330000, China
| | - Min Liu
- Changsha Maternal and Child Health Hospital, Changsha, 410000, China
| | - Rui Wang
- Henan Maternal and Child Health Hospital, Zhengzhou, 450000, China
| | - Xuezhou Yang
- Department of Reproductive Center, Affiliated Hospital of Hubei, Xiangyang Central Hospital, University of Arts and Science, Xiangyang, 441000, China
| | - Man Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wu Ren
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Dang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Sun
- Fujian Maternal and Child Health Hospital, Fuzhou, 350001, China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Conforti A, Di Girolamo R, Guida M, Alviggi C, Casarini L. Pharmacogenomic of LH and its receptor: are we ready for clinical practice? Reprod Biol Endocrinol 2025; 23:29. [PMID: 40001128 PMCID: PMC11863420 DOI: 10.1186/s12958-025-01359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Luteinizing hormone (LH) is fundamental to support development and reproduction. It acts through a receptor expressed in the gonads, modulating mitogenic, anti-apoptotic, and steroidogenic signals. LH is also marketed as a drug for controlled ovarian stimulation (COS), where it is administered to women to support the action of follicle-stimulating hormone and can lead to specific responses, depending on the individual genetic background. These concepts underline the relevance of a pharmacogenetic approach to COS, in the attempt to optimize clinical outcomes and avoid adverse events. However, knowledge is currently limited by the paucity of clinical studies. This review aims to provide a comprehensive overview of LH and its receptor activity, starting from the description of their molecular pathways from in vitro studies. Data on LH action from in vivo studies were described, as well as the impact of LH and LH/choriogonadotropin (hCG) receptor genetic variants on folliculogenesis and its association with infertility or polycystic ovarian syndrome. Finally, evidence from clinical studies evaluating genetic polymorphisms in the context of assisted reproductive technology treatments and its implications for a pharmacogenomic approach were discussed.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 5, Napoli, 80131, Italy.
| | | | - Maurizio Guida
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
3
|
Hochberg A, Dahan MH, Yarali H, Vuong LN, Esteves SC. Effect of follicle-stimulating hormone dose on the risk of being classified as suboptimal responders according to the POSEIDON criteria. J Assist Reprod Genet 2024; 41:3387-3398. [PMID: 39422825 PMCID: PMC11707116 DOI: 10.1007/s10815-024-03296-2] [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: 03/27/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
PURPOSE The purpose of this study is to investigate the impact of daily follicle-stimulating-hormone (FSH) dose on the likelihood of suboptimal response to ovarian stimulation (OS) for in vitro fertilization (IVF) according to POSEIDON's criteria. METHODS A tri-center retrospective cohort study (2015-2017) including women with normal anti-Müllerian hormone (AMH ≥ 1.2 ng/mL) and antral follicle count (AFC ≥ 5) values per POSEIDON's criteria, undergoing their first IVF/ICSI cycle using conventional OS (FSH ≥ 150 IU/day). Suboptimal response was the retrieval of 4-9 oocytes. In previous research, we detected an AMH ≤ 2.97 ng/mL and AFC ≤ 12 as the optimal cut-offs predicting suboptimal response. Therefore, we examined the effect of daily FSH dose (≤ 300 IU versus > 300 IU) on suboptimal response risk for each AMH and AFC value within these thresholds (AMH between 1.20 and 2.97 ng/mL, by 0.01 ng/mL increments; and an AFC between 5 and 12, by unit increments). Analysis involved contingency tables and multivariable logistic regression. RESULTS Included were 4005 patients with AMH and AFC values in the specific range, among whom 2131 (53.2%) were suboptimal responders. Among 177 AMH groups analyzed, apart from three distributed irregularly, daily FSH doses > 300 IU versus lower doses (≤ 300 IU) did not decrease suboptimal response risk; similarly, higher doses did not decrease risk at the eight AFC values examined (p > 0.05 for all). Using multivariable logistic regression, FSH doses were not associated with suboptimal response risk. Conversely, female age, AMH, AFC, and gonadotropin type were associated with suboptimal response. CONCLUSIONS In women with AMH values between 1.20 and 2.97 ng/mL and/or AFC between 5 and 12, FSH dose increase did not decrease suboptimal response risk. Individualizing the gonadotropin regimen and considering LH activity supplementation to FSH may mitigate risks.
Collapse
Affiliation(s)
- Alyssa Hochberg
- Department of Obstetrics and Gynecology, McGill University, 845 Rue Sherbrooke, O, Montreal, QC, 3HA 0G4, Canada.
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Michael H Dahan
- Department of Obstetrics and Gynecology, McGill University, 845 Rue Sherbrooke, O, Montreal, QC, 3HA 0G4, Canada
| | - Hakan Yarali
- Anatolia IVF, Ankara, Turkey
- Department of Obstetrics and Gynecology, Hacettepe University, Ankara, Turkey
| | - Lan N Vuong
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- IVFMD, My Duc Hospital, Ho Chi Minh City, Vietnam
- HOPE Research Center, My Duc Hospital, Ho Chi Minh City, Vietnam
| | - Sandro C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil
| |
Collapse
|
4
|
Zhang C, Nie Y, Xu B, Mu C, Tian GG, Li X, Cheng W, Zhang A, Li D, Wu J. Luteinizing Hormone Receptor Mutation (LHR N316S) Causes Abnormal Follicular Development Revealed by Follicle Single-Cell Analysis and CRISPR/Cas9. Interdiscip Sci 2024; 16:976-989. [PMID: 39150470 PMCID: PMC11512921 DOI: 10.1007/s12539-024-00646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Abnormal interaction between granulosa cells and oocytes causes disordered development of ovarian follicles. However, the interactions between oocytes and cumulus granulosa cells (CGs), oocytes and mural granulosa cells (MGs), and CGs and MGs remain to be fully explored. Using single-cell RNA-sequencing (scRNA-seq), we determined the transcriptional profiles of oocytes, CGs and MGs in antral follicles. Analysis of scRNA-seq data revealed that CGs may regulate follicular development through the BMP15-KITL-KIT-PI3K-ARF6 pathway with elevated expression of luteinizing hormone receptor (LHR). Because internalization of the LHR is regulated by Arf6, we constructed LHRN316S mice by CRISPR/Cas9 to further explore mechanisms of follicular development and novel treatment strategies for female infertility. Ovaries of LHRN316S mice exhibited reduced numbers of corpora lutea and ovulation. The LHRN316S mice had a reduced rate of oocyte maturation in vitro and decreased serum progesterone levels. Mating LHRN316S female mice with ICR wild type male mice revealed that the infertility rate of LHRN316S mice was 21.4% (3/14). Litter sizes from LHRN316S mice were smaller than those from control wild type female mice. The oocytes from LHRN316S mice had an increased rate of maturation in vitro after progesterone administration in vitro. Furthermore, progesterone treated LHRN316S mice produced offspring numbers per litter equivalent to WT mice. These findings provide key insights into cellular interactions in ovarian follicles and provide important clues for infertility treatment.
Collapse
Affiliation(s)
- Chen Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Hematology, Tangdu Hospital, Xi'an, 710032, China
| | - Yongqiang Nie
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bufang Xu
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunlan Mu
- School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Geng G Tian
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyong Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weiwei Cheng
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Aijun Zhang
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
- School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
5
|
Nenonen H, Kondic A, Henic E, Hjelmér I. Recurrent implantation failure and inflammatory markers in serum and follicle fluid of women undergoing assisted reproduction. J Reprod Immunol 2024; 162:104209. [PMID: 38310681 DOI: 10.1016/j.jri.2024.104209] [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: 06/30/2023] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
It has been shown previously that the immune system plays a role in implantation and embryo development. The objective was therefore to evaluate cytokine levels and Th1/Th2 ratio in women with recurrent implantation failure in this nested case-control study. Women with no implantation after ≥ 3 embryo transfers were included in the recurrent implantation failure group (n = 29) and were compared with women with successful pregnancy after the first embryo transfer, with an indication of male factor (n = 26). Cytokines analyzed with the Meso scale discovery (MSD) technology Proinflammatory Human Kit 1 and calculated Th1/Th2 ratios were the main outcome measures. In serum there was a difference between the recurrent implantation failure group and the control group in ratios for IFN-γ/IL-10 (p = 0.01), IL-1β/IL-10 (p = 0.04), IL-2/IL-10 (p = 0.00), TNF-α/IL-10 (p = 0.02), IFN-γ/IL-13 (p = 0.01), IL-12/IL-13 (p = 0.02), IL-2/IL-13 (p = 0.00), and TNF-α/IL-13 (p = 0.00), where the control group had higher ratios, i.e. a shift towards a Th1 pro-inflammatory profile before treatment start. In follicular fluid there were differences in ratios between IL-2/IL-10 (p = 0.02), IL-8/IL-10 (p = 0.02), TNF-α/IL-10 (p = 0.02), IFN-γ/IL-13 (p = 0.01), and TNF-α/IL-13 (p = 0.03). The recurrent implantation failure group had higher ratios except for IFN-γ/IL-13, indicating a shift towards a Th1 pro-inflammatory profile in their follicular fluid. Pro-inflammatory activity in both serum and follicle fluid differs in recurrent implantation failure patients and patients with successful assisted reproduction treatment. Women at risk of immune-related recurrent implantation failure could be identified proactively. Because it is taken at a timepoint closer to implantation, ratios in follicular fluid are specifically interesting as risk markers.
Collapse
Affiliation(s)
- Hannah Nenonen
- Department of Translational Medicine, Lund University, Jan Waldenströms gata 35, 214 28 Malmö, Sweden
| | - Alexandra Kondic
- Department of Translational Medicine, Lund University, Jan Waldenströms gata 35, 214 28 Malmö, Sweden
| | - Emir Henic
- Scanian University Hospital Malmö, Reproductive Medicine Centre, Östra Varvsgatan 11F, 205 02 Malmö, Sweden; Department of Translational Medicine, Lund University, Jan Waldenströms gata 35, 214 28 Malmö, Sweden
| | - Ida Hjelmér
- Department of Translational Medicine, Lund University, Jan Waldenströms gata 35, 214 28 Malmö, Sweden.
| |
Collapse
|
6
|
Li Q, Zhou X, Ye B, Tang M, Zhu Y. Ovarian response determines the luteinizing hormone suppression threshold for patients following the gonadotrophin releasing hormone antagonist protocol: A retrospective cohort study. Heliyon 2024; 10:e23933. [PMID: 38187350 PMCID: PMC10767281 DOI: 10.1016/j.heliyon.2023.e23933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/09/2024] Open
Abstract
Background Ovarian reactivity to gonadotrophin stimulation varies, and individual adjustments to the timing and dose of gonadotrophin-releasing hormone (GnRH) antagonist administration are necessary to prevent excessive increases and decreases in luteinizing hormone (LH) levels in patients with different ovarian response following the GnRH antagonist (GnRH-A) protocol. The present study aims to investigate optimal LH suppression thresholds for patients with normal ovarian response (NOR), high ovarian response (HOR), and poor ovarian response (POR) following the GnRH-A protocol respectively. Methods A total of 865 in vitro fertilization (IVF) cycles using a flexible or fixed GnRH-A protocol were included. Patients were categorized into the HOR, NOR, or POR group according to their anti-Müllerian hormone (AMH) levels. Then, patients in each group were stratified into one of four subgroups according to the quartile (Q1-Q4) of the basal LH level to LH on triggering day ratio (bLH/hLH). The primary outcomes were the clinical pregnancy and live birth rates, and the secondary outcomes were the number of oocytes retrieved, MII oocytes, two pronucleus (2PN) embryos, and good-quality embryos. Results There were 526 patients with NOR, 180 with HOR, and 159 with POR. Basal LH level, LH on triggering day and bLH/hLH were identified as independent predictors of clinical pregnancy rate and live birth rate by logistics regression analysis. Compared to those with NOR, patients with POR had the lowest embryo implantation rate (22.6% vs. 32.8%, P < 0.05), clinical pregnancy rate (32.3% vs. 47.3%, P < 0.05) and live birth rate (22.6 vs. 37.8%, P < 0.05) of fresh embryo transfer (ET). The embryo implantation, clinical pregnancy and live birth rates of frozen embryo transfer (FET) were not significantly different among the three groups. In the subgroup analysis, patients with HOR had the highest embryo implantation rate (51.6%, P < 0.05), clinical pregnancy rate (68.4%, P < 0.05) and live birth rate (52.6%, P < 0.05) of ET in Q3, with a bLH/hLH ratio of 2.40-3.69. In the NOR group, the embryo implantation rate (41.9%, P < 0.05), clinical pregnancy rate (61.5%, P < 0.05) and live birth rate (50.8%, P < 0.05) of ET and live birth rate (53.1%, P < 0.05) of FET were highest in Q2, with a bLH/hLH ratio of 1.29-2.05. Patients with POR had the highest clinical pregnancy rate (57.1%, P < 0.05) and live birth rate (42.9%, P < 0.05) of ET in Q2, with a bLH/hLH ratio of 0.86-1.35. Conclusions In the present study, the bLH/hLH ratio represented the LH suppression threshold. The subgroup analysis of HOR, NOR and POR showed that, the LH suppression threshold varies according to ovarian response. We recommend LH suppression thresholds of 2.40-3.69 for HOR, 1.29-2.05 for NOR, and 0.86-1.35 for POR to obtain the highest clinical pregnancy rate and live birth rate. This study provides comprehensive and precise references for clinicians to monitor LH levels individually during controlled ovarian stimulation (COS) according to the patient's ovarian response following the GnRH-A protocol.
Collapse
Affiliation(s)
- Qingfang Li
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Xiaoqian Zhou
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Bingru Ye
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Minyue Tang
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Yimin Zhu
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
7
|
Lee IT, Berger DS, Koelper N, Senapati S, Mainigi M. Race, ovarian responsiveness, and live birth after in vitro fertilization. Fertil Steril 2023; 120:1023-1032. [PMID: 37549835 DOI: 10.1016/j.fertnstert.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
OBJECTIVE To determine if ovarian responsiveness to gonadotropin stimulation differs by race/ethnicity and whether this predicts live birth rates (LBRs) in non-White patients undergoing in vitro fertilization (IVF). DESIGN Retrospective cohort study. SETTING Academic infertility center. PATIENT(S) White, Asian, Black, and Hispanic patients undergoing ovarian stimulation for IVF. INTERVENTION(S) Self-reported race and ethnicity. MAIN OUTCOME MEASURE(S) The primary outcome was ovarian sensitivity index (OSI), defined as (the number of oocytes retrieved ÷ total gonadotropin dose) × 1,000 as a measure of ovarian responsiveness, adjusting for age, body mass index, infertility diagnosis, and cycle number. Secondary outcomes included live birth and clinical pregnancy after first retrievals, adjusting for age, infertility diagnosis, and history of fibroids, as well as miscarriage rate per clinical pregnancy, adjusting for age, body mass index, infertility diagnosis, duration of infertility, history of fibroids, and use of preimplantation genetic testing for aneuploidy. RESULT(S) The primary analysis of OSI included 3,360 (70.2%) retrievals from White patients, 704 (14.7%) retrievals from Asian patients, 553 (11.6%) retrievals from Black patients, and 168 (3.5%) retrievals from Hispanic patients. Black and Hispanic patients had higher OSIs than White patients after accounting for those with multiple retrievals and adjusting for confounders (6.08 in Black and 6.27 in Hispanic, compared with 5.25 in White). There was no difference in OSI between Asian and White patients. The pregnancy outcomes analyses included 2,299 retrievals. Despite greater ovarian responsiveness, Black and Hispanic patients had lower LBRs compared with White patients, although these differences were not statistically significant after adjusting for confounders (adjusted odds ratio, 0.83; 95% confidence interval [CI], 0.63-1.09, for Black; adjusted odds ratio, 0.93; 95% CI, 0.61-1.43, for Hispanic). Ovarian sensitivity index was modestly predictive of live birth in White and Asian patients but not in Black (area under the curve, 0.51; 95% CI, 0.38-0.64) and Hispanic (area under the curve, 0.50; 95% CI, 0.37-0.63) patients. CONCLUSION(S) Black and Hispanic patients have higher ovarian responsiveness to stimulation during IVF but do not experience a consequent increase in LBR. Factors beyond differences in responsiveness to ovarian stimulation need to be explored to address the racial/ethnic disparity established in prior literature.
Collapse
Affiliation(s)
- Iris T Lee
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Dara S Berger
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nathanael Koelper
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Nagulapalli A, Vembu R, Pandurangi M, Nellepalli SR. Luteinizing Hormone/Human Chorionic Gonadotropin Receptor N312S Single-Nucleotide Polymorphism and Its Impact on Clinical and Reproductive Outcomes in Assisted Reproductive Technology: A Prospective Cohort Study. Cureus 2023; 15:e47217. [PMID: 38022167 PMCID: PMC10652146 DOI: 10.7759/cureus.47217] [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] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The aim of this study was to determine the genotypic distribution of luteinizing hormone/human chorionic gonadotropin receptor (LHCGR) N312S single-nucleotide polymorphism (SNP) and to investigate its impact on clinical and reproductive outcomes in infertile Indian women undergoing assisted reproductive technology (ART). Study design and settings This was a prospective cohort study conducted at a tertiary care university hospital. Subjects and methods Infertile women aged between 21 and 40 years undergoing ART with an antagonist protocol were enrolled in this study. A 2-ml sample of peripheral venous blood was collected from each woman and genotyped for the LHCGR N312S SNP. Participants were divided into three groups based on their SNP: NN, NS, and SS. All subjects underwent controlled ovarian hyperstimulation (COH) through a gonadotropin-releasing hormone (GnRH) antagonist protocol and intracytoplasmic sperm injection (ICSI). Of the 140 women recruited based on selection criteria, 128 underwent embryo transfer. We compared the genotypic distribution of the LHCGR N312S SNP, baseline characteristics, clinical outcomes, and reproductive outcomes in ART among the three groups. Data were analyzed using IBM SPSS Statistics for Windows, Version 29 (Released 2022; IBM Corp., Armonk, New York, United States). The chi-square test and Fisher-Irwin test were employed to evaluate significant differences among the qualitative categorical variables. A p-value of less than 0.05 was considered statistically significant. Results Among the test subjects, 19.3% were homozygous for the LHCGR N312 SNP (NN group), 38.6% were heterozygous (NS group), and 42.1% were homozygous for the LHCGR S312 SNP (SS group). Baseline characteristics were similar among the three groups. In terms of ovarian reserve tests, significantly lower anti-Müllerian hormone (AMH) levels were observed in the SS group compared to the NS and NN groups (2.8 ± 2.1 vs. 3.2 ± 2.5 vs. 4.3 ± 3.3; p=0.03). No significant differences were observed in COH outcomes such as duration of stimulation, total gonadotropin requirement, oocyte yield, or the number of good-quality embryos among the three groups. The cumulative pregnancy rate (82.9% vs. 50.0% vs. 38.2%, p=0.0005), cumulative clinical pregnancy rate (78.8% vs. 44.7% vs. 34.5%, p = 0.0005), and cumulative live birth rate (50.0% vs. 20.2% vs. 20.0%, p=0.005) were significantly higher in the NN group than in the NS and SS groups. Conclusion The study's findings suggest that LHCGR N312 may help predict reproductive outcomes in ART, which may aid in providing better counseling to infertile couples. We need more studies on individualized/personalized COH using pharmacogenomics for follicle-stimulating hormone (FSH) and luteinizing hormone (LH) supplementation based on combined FSH and LH receptor SNP and to assess their effects on ART outcomes.
Collapse
Affiliation(s)
- Amulya Nagulapalli
- Department of Reproductive Medicine and Surgery, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Radha Vembu
- Department of Reproductive Medicine and Surgery, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Monna Pandurangi
- Department of Reproductive Medicine and Surgery, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Sanjeeva R Nellepalli
- Department of Reproductive Medicine and Surgery, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
9
|
Jin H, Yang H, Zheng J, Zhou J, Yu R. Post-trigger luteinizing hormone concentration to positively predict oocyte yield in the antagonist protocol and its association with genetic variants of LHCGR. J Ovarian Res 2023; 16:189. [PMID: 37691102 PMCID: PMC10494325 DOI: 10.1186/s13048-023-01271-6] [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: 04/10/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND The concentration of human chorionic gonadotropin (hCG)/ luteinizing hormone (LH) after triggering is generally accepted as a predictor of the normal ovarian response to the trigger, but few studies have explored the distribution model of concentration and its impact on oocyte yield. Genetic variations in LHCGR, known as a receptor for hCG and LH, also play a role in oocyte maturation and retrieval. The objective of the study was to investigate the impact of concentrations of hCG/LH after triggering on oocyte yield and its association with genetic variants of LHCGR. METHODS A retrospective cohort study including 372 antagonist IVF cycles, in which 205 received the recombinant hCG trigger and 167 received the gonadotropin-releasing hormone agonist (GnRH-a) trigger, was conducted. The post-trigger concentrations of hCG/LH and the LHCGR N312S (rs2293275) genotype were evaluated in patients to analyse the impact of these factors on oocyte yield. RESULTS The oocyte retrieval rate (ORR) increased significantly among the low-, medium- and high-hCG-concentration groups (0.91 ± 0.25, 0.99 ± 0.23 and 1.08 ± 0.19, P < 0.001) and among the low-, medium- and high-LH-concentration groups (0.80 ± 0.29, 0.95 ± 0.21 and 1.07 ± 0.19, P < 0.001). The Pearson correlation coefficient between the post-trigger hCG concentration and ORR was 0.242 (P < 0.001), and that between the LH concentration and ORR was 0.454 (P < 0.001). After adjustment for confounding factors, high post-trigger LH concentrations remained associated with the significantly higher ORRs (adjusted R2 = 0.541, P < 0.001). Patients with the AG genotype of LHCGR N312S were more likely to have low post-trigger LH concentrations (46.10 IU/L versus 60.91 IU/L, P < 0.001) and a significantly lower ORR (0.85 versus 0.96, P = 0.042) than patients with the GG genotype after the GnRH-a trigger. CONCLUSIONS The post-trigger LH concentration can positively predict oocyte yield in antagonist IVF cycles, and patients with the AG genotype of LHCGR rs2293275 could have a suboptimal oocyte yield using the GnRH-a trigger.
Collapse
Affiliation(s)
- Hao Jin
- The Urological Surgical Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haiyan Yang
- The Reproductive Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China, No. 96, Fuxue Road, Lucheng District
| | - Jiujia Zheng
- The Reproductive Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China, No. 96, Fuxue Road, Lucheng District
| | - Jiechun Zhou
- The Reproductive Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China, No. 96, Fuxue Road, Lucheng District
| | - Rong Yu
- The Reproductive Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China, No. 96, Fuxue Road, Lucheng District.
| |
Collapse
|
10
|
Alviggi C, Longobardi S, Papaleo E, Santi D, Alfano S, Vanni VS, Campitiello MR, De Rosa P, Strina I, Huhtaniemi I, Pursiheimo JP, D'Hooghe T, Humaidan P, Conforti A. Genetic Variants of Gonadotropins and Their Receptors Could Influence Controlled Ovarian Stimulation: IVF Data from a Prospective Multicenter Study. Genes (Basel) 2023; 14:1269. [PMID: 37372449 DOI: 10.3390/genes14061269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Specific polymorphisms might influence controlled ovarian stimulation in women undergoing assisted reproductive technologies (ARTs). Data regarding possible interactions of these polymorphisms are still scanty. The aim of this analysis was to evaluate the effect of polymorphisms of gonadotropins and their receptors in women undergoing ART. METHODS A total of 94 normogonadotropic patients from three public ART units were enrolled. Patients underwent a gonadotropin releasing hormone (GnRH) long down-regulation protocol with a starting dose of 150 IU of recombinant follicular stimulating hormone (FSH) daily. Eight polymorphisms were genotyped. RESULTS A total of 94 women (mean age 30.71 ± 2.61) were recruited. Fewer fertilized and mature oocytes were retrieved in homozygous carriers of luteinizing hormone/choriogonadotropin receptor (LHCGR) 291 (T/T) than in heterozygous C/T carriers (p = 0.035 and p = 0.05, respectively). In FSH receptor (FSHR) rs6165 and FSHR rs6166 carriers, the ratio between total gonadotropin consumption and number of oocytes retrieved differed significantly among three genotypes (p = 0.050), and the ratio was lower in homozygous A/A carriers than in homozygous G/G and heterozygous carriers. Women who co-expressed allele G in FSHR-29 rs1394205 and FSHR rs6166 and allele C LHCGR 291 rs12470652 are characterized by an increased ratio between total FSH dosage and number of oocytes collected after ovarian stimulation (risk ratio: 5.44, CI 95%: 3.18-7.71, p < 0.001). CONCLUSIONS Our study demonstrated that specific polymorphisms affect the response to ovarian stimulation. Despite this finding, more robust studies are required to establish the clinical utility of genotype analysis before ovarian stimulation.
Collapse
Affiliation(s)
- Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
| | | | | | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Simona Alfano
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
| | | | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
| | - Pasquale De Rosa
- Azienda Ospedaliera Universitaria Federico II di Napoli, 80131 Naples, Italy
| | - Ida Strina
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
| | - Ilpo Huhtaniemi
- Faculty of Medicine, Department of Surgery and Cancer, London W12 0NN, UK
| | | | - Thomas D'Hooghe
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Peter Humaidan
- Fertility Clinic at Skive Regional Hospital, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
| | - Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
| |
Collapse
|
11
|
Hartanto S, Budiyanto A, Widayanti R, Setyawan EMN, Prasetya ID. Characterization of polymorphisms in the follicle-stimulating hormone receptor and insulin-like growth factor-1 genes and their association with fertility traits in Jawa-Brebes cows. Vet World 2023; 16:711-716. [PMID: 37235159 PMCID: PMC10206960 DOI: 10.14202/vetworld.2023.711-716] [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: 11/28/2022] [Accepted: 02/27/2023] [Indexed: 05/28/2023] Open
Abstract
Background and Aim The availability of fertility markers is crucial for maintaining, protecting, and improving the genetics of Jawa-Brebes (Jabres) cows. Follicle-stimulating hormone receptor (FSHR) and insulin-like growth factor-1 (IGF-1) play critical roles in female reproductive physiology. The single-nucleotide polymorphisms (SNPs) FSHR G-278A and IGF-1 C-512T correlate with cows' fertility traits. This study aimed to identify these SNPs and their potential associations with fertility parameters in Jabres cows. Materials and Methods Samples were collected from 45 heads of multiparous Jabres cows aged 3-10 years with body condition scores of 2.5-5.0 on a 5-point scale in Brebes Regency, Java, Indonesia. These cows were assigned to fertile (n = 16) and infertile groups (n = 29). Polymerase chain reaction (PCR) was carried out for DNA amplification of FSHR G-278A and IGF-1 C-512T fragments. Restriction fragment length polymorphism-PCR with the restriction enzymes FaqI for the product of FSHR G-278A and SnaBI for the product of IGF-1 C-512T was used to identify SNPs. Results The FaqI enzyme cut the 211 bp DNA fragment of FSHR G-278A in all samples into two bands of 128 bp and 83 bp (GG genotype). Meanwhile, the genotyping of amplicon products of IGF-1 C-512T generated a single 249 bp fragment (CC genotype) in both groups. Conclusion The results showed that the FSHR G-278A/FaqI and IGF-1 C-512T/SnaBI loci were monomorphic in Jabres cows. Thus, neither FSHR G-278A/FaqI nor IGF-1 C-512T/SnaBI is a possible genetic marker for fertility in Jabres cows.
Collapse
Affiliation(s)
- Slamet Hartanto
- Department of Reproduction, Obstetrics, and Gynecology, Faculty of Veterinary Medicine, Gadjah Mada University, Yogyakarta, Indonesia
- National Research and Innovation Agency (BRIN), Jakarta, Indonesia
| | - Agung Budiyanto
- Department of Reproduction, Obstetrics, and Gynecology, Faculty of Veterinary Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| | - Rini Widayanti
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Erif Maha Nugraha Setyawan
- Department of Reproduction, Obstetrics, and Gynecology, Faculty of Veterinary Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| | - Imawan Daru Prasetya
- Directorate General of Livestock and Animal Health, Ministry of Agriculture, Jakarta, Indonesia
| |
Collapse
|
12
|
Bøtkjær JA, Kristensen SG, Olesen HØ, Larsson P, Mannaerts B, Andersen CY. Dose-dependent stimulation of human follicular steroidogenesis by a novel rhCG during ovarian stimulation with fixed rFSH dosing. Front Endocrinol (Lausanne) 2022; 13:1004596. [PMID: 36339420 PMCID: PMC9632659 DOI: 10.3389/fendo.2022.1004596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Choriogonadotropin (CG) beta (FE 999302), a novel recombinant human (h)CG produced by a human cell line, has a longer half-life and higher potency than CG alfa produced by a Chinese hamster ovary cell line. hCG augments steroid production, but the extent of which CG beta treatment during ovarian stimulation (OS) increases steroidogenesis is unknown. Objective To explore how increasing doses of CG beta during OS augment follicular steroidogenesis and change gene expression in cumulus cells. Study design This study is part of a randomized, double-blind, placebo-controlled trial to investigate the efficacy and safety of CG beta plus recombinant follicle-stimulating hormone (rFSH) in women undergoing OS during a long gonadotrophin-releasing hormone agonist protocol. The study primary endpoint was intrafollicular steroid concentrations after CG beta administration. Secondary outcomes were gene expression of FSHR , LHR, CYP19a1, and androgen receptor (AR). Participants/methods 619 women with anti-Müllerian hormone levels 5-35 pmol/L were randomized to receive placebo or 1, 2, 4, 8, or 12 µg/day CG beta from Day 1 of OS plus rFSH. Follicular fluid (FF) (n=558), granulosa (n=498) and cumulus cells (n=368) were collected at oocyte retrieval. Steroid FF hormones were measured using enzyme-linked immunosorbent assays, gene expression was analyzed in cumulus cells by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and single nucleotide polymorphism (SNP) analysis was performed in granulosa cells. Results 17-OH-progesterone, androstenedione, testosterone, and estradiol concentrations significantly increased in a CG-beta dose-dependent manner during OS (p<0.0001), reaching up to 10 times higher values in the highest dose group versus placebo. There was no difference between CG beta dose groups and placebo for progesterone. Expression levels of CYP19a1 increased significantly in the highest dose group of CG beta (p=0.0325) but levels of FSHR , LHR and AR were not affected by CG beta administration. There were no differences between the FSHR (307) or LHR(312) SNP genotypes for dose-dependent effects of CG beta in relation to number of oocytes, intrafollicular steroid hormone levels, or gene expression levels. Conclusions These results reflect the importance of the combined effect of FSH and hCG/LH during OS on granulosa cell activity, follicle health and potentially oocyte quality. Trial Registration number 2017-003810-13 (EudraCT Number). Trial Registration date 21 May 2018. Date of first patient’s enrolment 13 June 2018. Presented at the 38th Annual Meeting of the European Society of Human Reproduction and Embryology, P-567, 2022.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Hanna Ørnes Olesen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Per Larsson
- Global Biometrics, Ferring Pharmaceuticals A/S, Copenhagen, Denmark
| | - Bernadette Mannaerts
- Reproductive Medicine & Maternal Health, Ferring Pharmaceuticals A/S, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Surcel M, Doroftei B, Neamtiu IA, Muresan D, Caracostea G, Goidescu I, Staicu A, Nemeti G, Bloom MS, Zlatescu-Marton C. Impact of Follicle Stimulating Hormone Receptor (FSHR) Polymorphism on the Efficiency of Co-Treatment with Growth Hormone in a Group of Infertile Women from Romania. Diagnostics (Basel) 2022; 12:diagnostics12102371. [PMID: 36292058 PMCID: PMC9600645 DOI: 10.3390/diagnostics12102371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
“Poor responders” (PR) are an important category of infertile women who experience a modest response to controlled ovarian stimulation. In this study, we evaluated response to growth hormone (GH) administration among PR patient subtypes stratified by follicle stimulation hormone receptor (FSHR) polymorphism (c.2039A > G p.Asn680Ser). We conducted a cohort study of 125 women with poor ovarian response, 58 of whom received GH in addition to the standard treatment, and 67 of whom received the standard treatment only. The Ala307Thr polymorphism genotypes were analyzed using a polymerase chain reaction-restriction fragment length polymorphism method, and the FSHR gene polymorphism was analyzed using a predesigned TaqMan SNP Genotyping Assay (rs6166). A comparative analysis detected statistically significant differences in mean mature follicles (p = 0.0002), metaphase-II oocytes (p = 0.0005), progesterone levels (p = 0.0036), and IGF levels (follicle IGF1, p = 0.0004) between GH-treated and non-GH-treated participants with the FSHR (Ser/Ser) polymorphism. However, the differences were modest among participants with the other two FSHR polymorphisms (Ser/Asn and Asn/Asn). The subcategory of patients with the FSHR Asn680Ser (Ser/Ser) polymorphism showed a stronger response when GH was added to the IVF protocol.
Collapse
Affiliation(s)
- Mihai Surcel
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Bogdan Doroftei
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Iulia Adina Neamtiu
- Health Department, Environmental Health Center, 58 Busuiocului Street, 400240 Cluj-Napoca, Romania
- Faculty of Environmental Science and Engineering, Babes-Bolyai University, 30 Fantanele Street, 400294 Cluj-Napoca, Romania
- Correspondence: ; Tel.: +40-264432979; Fax: +40-264534404
| | - Daniel Muresan
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Gabriela Caracostea
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Iulian Goidescu
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Adelina Staicu
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Georgiana Nemeti
- 1st Department of Obstetrics and Gynecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Michael S. Bloom
- Department of Global and Community Health, George Mason University, 4400 University Drive, MS 5B7, Fairfax, VA 22030, USA
| | | |
Collapse
|
14
|
Pirtea P, de Ziegler D, Marin D, Sun L, Tao X, Ayoubi JM, Franasiak J, Scott RT. Gonadotropin receptor polymorphisms (FSHR N680S and LHCGR N312S) are not predictive of clinical outcome and live birth in assisted reproductive technology. Fertil Steril 2022; 118:494-503. [PMID: 35842313 DOI: 10.1016/j.fertnstert.2022.06.011] [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: 03/21/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To study the consequences of specific genotype profiles of follicle-stimulating hormone receptor (FSHR) and luteinizing hormone choriogonadotropin receptor (LHCGR) on assisted reproductive technology outcomes when preimplantation genetic testing for aneuploidy is used for controlling the embryo ploidy status. The most common reported single-nucleotide polymorphisms in the amino acid position for the FSHR (N680S; N: asparagine, S: serine; [rs6166]) and the LHCGR (N312S variant; N: asparagine, S: serine [rs2293275]) were chosen for this study. DESIGN Retrospective cohort study. SETTING Private Fertility Clinic. PATIENT(S) All women aged 18-40 years undergoing their first assisted reproductive technology cycle with aneuploidy screening between 2006 and 2017 with body mass index of >18 and <40 kg/m2 were included. INTERVENTION(S) All patients received both recombinant follicle-stimulating hormone and human menopausal gonadotropin or low dose human chorionic gonadotropin. Genomic DNA was isolated from patients' blood. Genotyping of the FSHR and LHCGR polymorphisms was performed using TaqMan genotyping assays. Associations between both receptor genotypes and clinical outcomes were assessed using generalized regression and ANOVA. MAIN OUTCOMES MEASURE(S) Live birth rate was the primary outcome. Secondary outcomes included oocyte yield, mature oocytes, blastulation rate, usable blastocyst rate, and implantation rate. RESULT(S) A total of 1,183 patients met the inclusion criteria and generated reliable genotype results. The overall genotype frequencies in the study population for the FSHR gene were as follows: 21.7% homozygous for S in codon 680, 29.2% homozygous for N680, and 48.1% heterozygous (N680S). As for the LHCGR, 15.6% were homozygous for N312, 38.5% homozygous for S312 and 45.9% heterozygous (N312S). Our study population consisted of 53.8% non-Hispanic white; 6.1% Hispanic white; 4.1% Afro-American; 15.4% Asian; and 20.6% other or unknown. No significant association was found with any of the studied variables (oocyte yield, usable blastocyst rate, implantation rate, live birth) when genotypes were analyzed per receptor or in combination with one another. There was a statistically significant but clinically irrelevant difference in the rate of mature oocytes across different variant combinations. CONCLUSION(S) Our findings suggest that the presence of gonadotropin receptor polymorphisms in both FSHR N680S and LHCGR N312S are not associated with assisted reproductive technology outcomes; therefore, these variants should not be considered reproductive predictors.
Collapse
Affiliation(s)
- Paul Pirtea
- Hôpital FOCH, Suresnes, France; Instituto Valenciano de Infertilidad Reproductive Medicine Associates, Basking Ridge, New Jersey.
| | | | - Diego Marin
- Instituto Valenciano de Infertilidad Reproductive Medicine Associates, Basking Ridge, New Jersey; Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Li Sun
- Foundation for Embryonic Competence, Basking Ridge, New Jersey
| | - Xin Tao
- Foundation for Embryonic Competence, Basking Ridge, New Jersey
| | | | - Jason Franasiak
- Instituto Valenciano de Infertilidad Reproductive Medicine Associates, Basking Ridge, New Jersey; Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Richard T Scott
- Instituto Valenciano de Infertilidad Reproductive Medicine Associates, Basking Ridge, New Jersey; Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Conforti A, Tüttelmann F, Alviggi C, Behre HM, Fischer R, Hu L, Polyzos NP, Chuderland D, Rama Raju GA, D’Hooghe T, Simoni M, Sunkara SK, Longobardi S. Effect of Genetic Variants of Gonadotropins and Their Receptors on Ovarian Stimulation Outcomes: A Delphi Consensus. Front Endocrinol (Lausanne) 2022; 12:797365. [PMID: 35178027 PMCID: PMC8844496 DOI: 10.3389/fendo.2021.797365] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/20/2023] Open
Abstract
Background A Delphi consensus was conducted to evaluate the influence of single nucleotide polymorphisms (SNPs) in genes encoding gonadotropin and gonadotropin receptors on clinical ovarian stimulation outcomes following assisted reproductive technology (ART) treatment. Methods Nine experts plus two Scientific Coordinators discussed and amended statements plus supporting references proposed by the Scientific Coordinators. The statements were distributed via an online survey to 36 experts, who voted on their level of agreement or disagreement with each statement. Consensus was reached if the proportion of participants agreeing or disagreeing with a statement was >66%. Results Eleven statements were developed, of which two statements were merged. Overall, eight statements achieved consensus and two statements did not achieve consensus. The statements reaching consensus are summarized here. (1) SNP in the follicle stimulating hormone receptor (FSHR), rs6166 (c.2039A>G, p.Asn680Ser) (N=5 statements): Ser/Ser carriers have higher basal FSH levels than Asn/Asn carriers. Ser/Ser carriers require higher amounts of gonadotropin during ovarian stimulation than Asn/Asn carriers. Ser/Ser carriers produce fewer oocytes during ovarian stimulation than Asn/Asn or Asn/Ser carriers. There is mixed evidence supporting an association between this variant and ovarian hyperstimulation syndrome. (2) SNP of FSHR, rs6165 (c.919G>A, p.Thr307Ala) (N=1 statement): Few studies suggest Thr/Thr carriers require a shorter duration of gonadotropin stimulation than Thr/Ala or Ala/Ala carriers. (3) SNP of FSHR, rs1394205 (-29G>A) (N=1 statement): Limited data in specific ethnic groups suggest that A/A allele carriers may require higher amounts of gonadotropin during ovarian stimulation and produce fewer oocytes than G/G carriers. (4) SNP of FSH β-chain (FSHB), rs10835638 (-211G>T) (N=1 statement): There is contradictory evidence supporting an association between this variant and basal FSH levels or oocyte number. (5) SNPs of luteinizing hormone β-chain (LHB) and LH/choriogonadotropin receptor (LHCGR) genes (N=1 statement): these may influence ovarian stimulation outcomes and could represent potential future targets for pharmacogenomic research in ART, although data are still very limited. Conclusions This Delphi consensus provides clinical perspectives from a diverse international group of experts. The consensus supports a link between some variants in gonadotropin/gonadotropin receptor genes and ovarian stimulation outcomes; however, further research is needed to clarify these findings.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - Hermann M. Behre
- Center for Reproductive Medicine and Andrology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Robert Fischer
- Department of Gynecological Endocrinology and Reproductive Medicine, Fertility Center Hamburg, Hamburg, Germany
| | - Liang Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Nikolaos P. Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
| | - Dana Chuderland
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Thomas D’Hooghe
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Darmstadt, Germany
- Research Group Reproductive Medicine, Department of Development and Regeneration, Organ Systems, Group Biomedical Sciences, KU Leuven (University of Leuven), Leuven, Belgium
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sesh K. Sunkara
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Salvatore Longobardi
- Global Clinical Development, Research and Development, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
16
|
Di Segni N, Busnelli A, Secchi M, Cirillo F, Levi-Setti PE. Luteinizing hormone supplementation in women with hypogonadotropic hypogonadism seeking fertility care: Insights from a narrative review. Front Endocrinol (Lausanne) 2022; 13:907249. [PMID: 35979440 PMCID: PMC9377693 DOI: 10.3389/fendo.2022.907249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
The management of infertile women affected by hypogonadotropic hypogonadism (HH) or conditions mimicking it is particularly challenging. In the present narrative review, we aimed to synthesize the available evidence on the benefit (if any) of exogenous luteinizing hormone (LH) supplementation in this group of patients. Available data support LH supplementation in women with organic or functional HH. On the contrary, the benefit of exogenous LH on reproductive outcomes both in advanced maternal age patients and in cases of depletion of FSH and LH levels induced by GnRH analogues has not been demonstrated. unfortunately, the inhomogeneous study populations as well as the methodological heterogeneity between studies focused on women affected by conditions mimicking HH do not allow reliable conclusions to be drawn.
Collapse
Affiliation(s)
- Noemi Di Segni
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Andrea Busnelli
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Matteo Secchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Federico Cirillo
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Paolo Emanuele Levi-Setti
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- *Correspondence: Paolo Emanuele Levi-Setti,
| |
Collapse
|
17
|
Liu S, Lv Y, Liu M, Han S, Liu X, Zhao Z, Cui W, Yang A, Li Y. Luteinizing hormone-based modified GnRH antagonist protocol in normal responders undergoing in vitro fertilization treatment: A multi-center randomized controlled trial. Front Endocrinol (Lausanne) 2022; 13:922950. [PMID: 36034460 PMCID: PMC9403177 DOI: 10.3389/fendo.2022.922950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE To study the clinical efficacy and cost-effectiveness of a modified gonadotrophin-releasing hormone (GnRH) antagonist protocol based on luteinizing hormone (LH) levels through one complete assisted reproductive technology (ART) cycle in normal responders. DESIGN Non-inferiority, multicenter randomized controlled trial. SETTING University-based hospitals and an academic medical center. PATIENTS A total of 372 patients fulfilled the inclusion criteria and were eligible to participate. INTERVENTIONS Participants were randomized at a 1:1 ratio and stimulated with the conventional flexible GnRH antagonist protocol (control group) or LH-based modified GnRH antagonist protocol (study group). MAIN OUTCOME MEASURES The primary outcome was the cumulative ongoing pregnancy rate per aspiration. The secondary outcomes were number of oocytes retrieved, number of good quality embryos, cumulative positive βhCG rate, cumulative clinical pregnancy rate, pregnancy loss rate, moderate and severe ovarian hyperstimulation syndrome (OHSS), and financial expenditure. RESULTS The cumulative ongoing pregnancy rate was 65.1% in the study group and 70.1% in the control group (odds ratio, 0.79; 95% confidence interval, 0.50-1.26; P = 0.33). The multivariate regression analyses results showed that the number of retrieved oocytes was positively associated with the odds for a higher cumulative ongoing pregnancy rate (adjusted odds ratio, 1.11, 95% confidence interval, 1.06-1.17, P < 0.001). The treatment protocol, female age, and body mass index were not independent predictors. The incremental cost-effectiveness ratio for luteinizing hormone-based gonadotrophin releasing hormone antagonist protocol versus the conventional flexible gonadotrophin releasing hormone antagonist protocol was estimated at 3568.6 USD for each additional ongoing pregnancy. CONCLUSION The luteinizing hormone-based gonadotrophin releasing hormone antagonist protocol had clinical efficacy similar to the conventional flexible gonadotrophin releasing hormone antagonist protocol in normal responders undergoing in vitro fertilization treatment but was more cost-effective considering the cumulative ongoing pregnancy rate in the entire assisted reproductive technology cycle. CLINICAL TRIAL REGISTRATION www.chictr.org.cn, identifier: ChiCTR1800018077. URL OF THE REGISTRATION SITE http://www.chictr.org.cn/edit.aspx?pid=27389&htm=4. TRIAL REGISTRATION DATE 29 August 2018. DATE OF FIRST PATIENT ENROLLMENT 1 September 2018.
Collapse
Affiliation(s)
- Shan Liu
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yasu Lv
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Minghui Liu
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuo Han
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoqun Liu
- Center of Reproductive Medicine, Research Institute of Family Planning of Hebei Province, Shijiazhuang, China
| | - Zhiming Zhao
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Cui
- Center of Reproductive Medicine, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Aijun Yang
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Yuan Li
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yuan Li,
| |
Collapse
|
18
|
Koloda YA, Denisova YV, Podzolkova NM. Genetic polymorphisms of reproductive hormones and their receptors in assisted reproduction technology for patients with polycystic ovary syndrome. Drug Metab Pers Ther 2021; 37:111-122. [PMID: 34851566 DOI: 10.1515/dmpt-2021-0123] [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: 03/25/2021] [Accepted: 08/31/2021] [Indexed: 11/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies in women of childbearing, which is defined by the accumulation of multiple, small fluid-filled ovarian cysts without the selection of a single dominant follicle. Most PCOS phenotypes are characterized by the absence of spontaneous ovulation, resistance toward ovulation inductors, the production of a large immature oocytes number, and the high prevalence of ovarian hyperstimulation syndrome, resulting in reduced assisted reproductive technologies (ART) programs effectiveness. The review analyses current data about the relationship between polymorphism genotypes of KISS genes, follicle stimulating hormone (FSH), luteinizing hormone (LH), anti-Müllerian hormone (AMH) and their receptors genes, gonadotropin-releasing hormone (GnRH), estrogen, and progesterone receptors genes, the PCOS risk and the features of ovarian response to stimulation during ART cycles. The use of single nucleotide polymorphisms (SNPs) as prognostic markers of ART programs outcomes would provide a personalized approach to the drugs and doses choice for ovarian stimulation and significantly increase the chance of pregnancy.
Collapse
Affiliation(s)
- Yulia A Koloda
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Yulia V Denisova
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Natalia M Podzolkova
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|
19
|
Recombinant luteinizing hormone supplementation in assisted reproductive technology: a review of literature. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Luteinizing hormone (LH) has the main role in ovarian function in both natural and artificial cycles. A normal LH concentration during controlled ovarian hyperstimulation is positively correlated to the number and quality of retrieved oocytes and resulting embryos.
Main body of the abstract
In this study, we reviewed whether rLH administration, adjunct to the ovarian stimulation regimen, could improve clinical outcomes. The literature review showed that rLH supplementation improves assisted reproductive technology (ART) outcomes among women with hypogonadotropic hypogonadism, and hyporesponsive women to follicle-stimulating hormone monotherapy. Besides, rLH supplementation has advantages for poor responder women 36–39 years of age. Even though the data suggested no priority regarding the LH source for improving ART outcome, women with different LH polymorphisms who did not respond similarly to ovarian stimulation may benefit from adjuvant rLH therapy.
Conclusion
rLH usage for improving ART outcome should be scrutinized via well-designed studies considering the subgroups of infertile women who benefit the most from rLH adjuvant therapy, the type of ovarian stimulation protocol to which rLH would be added, and also the exact dosage, as well as the proper timing (during or prior to a cycle).
Collapse
|
20
|
Ali ARA, Abdul-Rasheed OF, Al-Kawaz UM. The Impact of Luteinizing Hormone/Chorionic Gonadotropin Hormone Receptor Gene Polymorphism rs68073206 in Men with Non-obstructive Azoospermia: A Case-control Study. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: The functional consequences of the luteinizing hormone/chorionic gonadotropin hormone receptor (LHCGR) gene single nucleotide polymorphism (rs68073206) on male infertility in patients with non-obstructive azoospermia (NOA) is not clear.
Objective: To examine whether the presence of LHCGR gene; rs68073206 single nucleotide polymorphisms (SNPs) can be associated with incidence of non-obstructive azoospermia.
Materials and methods: A case-control study comprised of a total of 70 unrelated Iraqi infertile men with non-obstructive azoospermia (zero sperm in semen) whose were on two groups: Group I that were diagnosed to have NOA but didn’t receive infertility treatment yet (33 patient with age of 31.58±1.059 year) and group II that were receiving injectable gonadotropin treatment (37 patient with age of 33.46±1.173 year). In addition to 34 age and BMI matched healthy fertile normozoospermic men (according to the parameters of WHO, 2010). The study population was genotyped by TaqMan assay for LHCGR gene single nucleotide polymorphism (rs68073206). The level of each hormone was estimated by immunoassay technique while the sperm analyses were conducted in accordance with the World Health Organization criteria.
Results: The study revealed a statistically significant higher hormonal level of serum inhibin B in infertile group I patients with wild GG genotype (246.445±224.106 pg/ml), and the p-value is (0.0439) as compared to that hormone levels of GT and TT genotypes carriers that were (85.969±71.685 pg/ml) and (56.420±23.988 pg/ml) respectively. ). The genotyping variations of patients, whether carrying the homozygous GG, heterozygous GT or homozygous TT genotype, did not reveal a statistically significant difference in distribution as compared to control individuals.
Conclusions: The LHCGR gene rs68073206 polymorphisms in our population having non-obstructive azoospermia can be suggested to have a modulating potential in variable gonadotropin sensitivity. The detected non-significant difference in genotypic prevalence can be attributable to the limited sample size.
Collapse
|
21
|
Guo C, Yu H, Feng G, Lv Q, Liu X, Liu X. Associations of FSHR and LHCGR gene variants with ovarian reserve and clinical pregnancy rates. Reprod Biomed Online 2021; 43:561-569. [PMID: 34391684 DOI: 10.1016/j.rbmo.2021.06.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 01/18/2023]
Abstract
RESEARCH QUESTION Are there any associations between the variants of FSHR c.2039 G>A (p. Ser680Asn, rs6166) and LHCGR c.935A>G (p. Asn312Ser, rs2293275) and ovarian reserve, ovarian response, clinical pregnancy rate and POSEIDON group? DESIGN A total of 210 infertile women were enrolled in this prospective study. The gene variants were analysed by the Sanger method. The clinical parameters were analysed based on genotypes. RESULTS The frequency of heterozygous and homozygous G allele for FSHR c.2039 G>A in the low prognosis group was significantly higher than that in other response groups (P = 0.034); there was no significant association between LHCGR c.935 A>G and ovarian response. Moreover, the serum anti-Müllerian hormone (AMH) concentration, antral follicle count (AFC), oocytes retrieved, metaphase II (MII) oocytes and two-pronuclear (2PN) oocytes in patients with AG genotype for FSHR c.2039 G>A were significantly lower than those with AA genotype. The serum LH concentrations and clinical pregnancy rate of fresh embryo transfer in patients with GG genotype for LHCGR c.935 A>G were significantly higher than that of the AG genotype. In POSEIDON analysis, the low prognosis women with AA genotype for FSHR c.2039 G>A were more likely to appear in subgroup 1 (P = 0.038). CONCLUSION The FSHR c.2039 G>A variant has a significant beneficial influence on ovarian reserve and ovarian response. The LHCGR c.935 A>G variant is associated with increased clinical pregnancy rate of fresh embryo transfer in infertile women. In addition, the low prognosis women with AA genotype for FSHR c.2039 G>A tend to show better ovarian reserve and prognosis.
Collapse
Affiliation(s)
- Chun Guo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | | | - Guimei Feng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Qun Lv
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Xiaoqi Liu
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital; Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital; School of Medicine, University of Electronic Science and Technology of China.
| | - Xiangqin Liu
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
22
|
Boudry L, Racca A, Tournaye H, Blockeel C. Type and dose of gonadotropins in poor ovarian responders: does it matter? Ther Adv Reprod Health 2021; 15:26334941211024203. [PMID: 34263173 PMCID: PMC8243085 DOI: 10.1177/26334941211024203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Infertile patients with a diminished ovarian reserve, also referred to as
poor ovarian responders, constitute a substantial and increasing
population of patients undergoing in vitro
fertilization. The management of patients with poor ovarian response
is still a controversial issue. Almost a century has passed since the
introduction of the first gonadotropin. A broad collection of urinary
and recombinant gonadotropins, including biosimilars, is commercially
available now. Despite great advances in assisted reproductive
technology, there remains uncertainty about the optimal treatment
regimen for ovarian stimulation in poor ovarian responders. Although
oocyte donation is the most successful and ultimate remedy for poor
ovarian responders, most patients persist on using their own oocytes
in several attempts, to achieve the desired pregnancy. The aim of this
review is twofold: first, to provide an overview of the commercially
available gonadotropins and summarize the available evidence
supporting the use of one or another for ovarian stimulation in poor
ovarian responders, and second, to address the controversies on the
dosage of gonadotropins for this specific in vitro
fertilization population.
Collapse
Affiliation(s)
- Liese Boudry
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, 1090, Brussels, Belgium
| | - Annalisa Racca
- Department of Obstetrics, Gynecology and Reproduction, Hospital Universitari Dexeus, Barcelona, Spain
| | - Herman Tournaye
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium; Department of Obstetrics, Gynecology, Perinatology and Reproduction, Institute of Professional Education, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Christophe Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium; Department of Obstetrics and Gynaecology, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
23
|
Involvement of single nucleotide polymorphisms in ovarian poor response. J Assist Reprod Genet 2021; 38:2405-2413. [PMID: 34050449 DOI: 10.1007/s10815-021-02242-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE Unpredictability in acquiring an adequate number of high-quality oocytes following ovarian stimulation is one of the major complications in controlled ovarian hyperstimulation (COH). Genetic predispositions of variations could alter the immunological profiles and consequently be implicated in the variability of ovarian response to the stimulation. DESIGN Uncovering the influence of variations in AMHR2, LHCGR, MTHFR, PGR, and SERPINE1 genes with ovarian response to gonadotrophin stimulation in COH of infertile women. METHODS Blood samples of the women with a good ovarian response (GOR) or with a poor ovarian response (POR) were collected. Genomic DNA was extracted, and gene variations were genotyped by TaqMan SNP Genotyping Assays using primer-probe sets or real-time PCR Kit. RESULTS Except for PGR (rs10895068), allele distributions demonstrate that the majority of POR patients carried minor alleles of AMHR2 (rs2002555, G-allele), LHCGR (rs2293275, G-allele), MTHFR (rs1801131, C-allele, and rs1801133, T-allele), and SERPINE1 (rs1799889, 4G allele) genes compared to the GOR. Similarly, genotypes with a minor allele in AMHR2, LHCGR, MTHFR, and SERPINE1 genes had a higher prevalence among POR patients with the polymorphic genotypes. However, further genotype stratification indicated that the minor alleles of these genes are not associated with poor response. Multivariate logistic analysis of clinical-demographic factors and polymorphic genotypes demonstrated a correlation between FSH levels and polymorphic genotypes of SERPINE1 in poor response status. CONCLUSIONS Despite a higher prevalence of AMHR2, LHCGR, MTHFR, and SERPINE1 variations in the patients with poor ovarian response, it seems that these variations are not associated with the ovarian response.
Collapse
|
24
|
Bosch E, Alviggi C, Lispi M, Conforti A, Hanyaloglu AC, Chuderland D, Simoni M, Raine-Fenning N, Crépieux P, Kol S, Rochira V, D'Hooghe T, Humaidan P. Reduced FSH and LH action: implications for medically assisted reproduction. Hum Reprod 2021; 36:1469-1480. [PMID: 33792685 PMCID: PMC8129594 DOI: 10.1093/humrep/deab065] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Luteinizing hormone (LH) and follicle-stimulating hormone (FSH) play complementary roles in follicle development and ovulation via a complex interaction in the hypothalamus, anterior pituitary gland, reproductive organs, and oocytes. Impairment of the production or action of gonadotropins causes relative or absolute LH and FSH deficiency that compromises gametogenesis and gonadal steroid production, thereby reducing fertility. In women, LH and FSH deficiency is a spectrum of conditions with different functional or organic causes that are characterized by low or normal gonadotropin levels and low oestradiol levels. While the causes and effects of reduced LH and FSH production are very well known, the notion of reduced action has received less attention by researchers. Recent evidence shows that molecular characteristics, signalling as well as ageing, and some polymorphisms negatively affect gonadotropin action. These findings have important clinical implications, in particular for medically assisted reproduction in which diminished action determined by the afore-mentioned factors, combined with reduced endogenous gonadotropin production caused by GnRH analogue protocols, may lead to resistance to gonadotropins and, thus, to an unexpected hypo-response to ovarian stimulation. Indeed, the importance of LH and FSH action has been highlighted by the International Committee for Monitoring Assisted Reproduction Technologies (ICMART) in their definition of hypogonadotropic hypogonadism as gonadal failure associated with reduced gametogenesis and gonadal steroid production due to reduced gonadotropin production or action. The aim of this review is to provide an overview of determinants of reduced FSH and LH action that are associated with a reduced response to ovarian stimulation.
Collapse
Affiliation(s)
| | - C Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - M Lispi
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - A Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - A C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - D Chuderland
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany
| | - M Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - N Raine-Fenning
- Department of Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham, UK
| | - P Crépieux
- Physiologie de la Reproduction et des Comportements, UMR INRA 085, CNRS 7247, Université de Tours, Nouzilly, France
| | - S Kol
- IVF Unit, Elisha Hospital, Haifa, Israel
| | - V Rochira
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - T D'Hooghe
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,Department of Development & Regeneration, University of Leuven (KU Leuven), Leuven, Belgium.,Department of Obstetrics and Gynecology, Yale University, New Haven, CT, USA
| | - P Humaidan
- Fertility Clinic, Skive Regional Hospital, and the Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Effect of polymorphisms of MTHFR in controlled ovarian stimulation: a systematic review and meta-analysis. J Assist Reprod Genet 2021; 38:2237-2249. [PMID: 34032987 DOI: 10.1007/s10815-021-02236-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE Although several studies have reported a potential impact of methylenetetrahydrofolate reductase (MTHFR) polymorphisms on controlled ovarian stimulation (COS), the results remain controversial. The aim of the systematic review and meta-analysis was to evaluate the effect of MTHFR polymorphism on COS outcomes. METHODS PubMed, Web of Science, Embase, and Cochrane Central Register of Controlled Trials databases were searched up to December 2, 2020. COS clinical outcomes based on gene polymorphisms were included. Two reviewers independently extracted the data. The primary outcome was the number of oocytes retrieved. The secondary outcomes were the number of metaphase II (MII) oocytes, stimulation duration, basal follicle-stimulating hormone (FSH) level, FSH dosage, positive pregnancy test, ongoing pregnancy rate, clinical pregnancy rate, miscarriage rate, and live birth rate. Meta-analysis was performed using a fixed-effect model or random-effect model with Review Man 5.3.5. Mean difference (MD) with 95% confidence intervals (95%CIs) was calculated for continuous outcomes. The quality assessment of included studies was evaluated by using the Newcastle-Ottawa Scale. RESULTS Eleven studies were included in the systematic review, and seven studies with 2015 participants were included in the meta-analysis. Basal FSH level was significantly lower in CC homozygotes than TT homozygotes (four studies, 867 participants, MD - 0.54, 95%CI - 0.85 to - 0.23, P = 0.0006; I2 = 0%) of MTHFR (rs1801133). FSH dose was significantly fewer in CC homozygotes compared with CT heterogeneous (three studies, 949 participants, MD - 75.78, 95%CI - 135.23 to - 16.33, P = 0.01; I2 = 32%) or CT/TT model (three studies, 1097 participants, MD - 80.18, 95%CI - 135.54 to - 24.81, P = 0.005; I2 = 42%). Differences in the oocytes retrieved and stimulation duration were insignificant. Gene variants on MTHFR (rs1801133) and MTHFR (rs1801131) were reported in ongoing pregnancy rate, clinical pregnancy rate, and live birth rate. CONCLUSION Studies to date indicate that polymorphisms of MTHFR could influence basal FSH level and FSH dose. The results could be useful to promote clinical practice on COS protocols. Further studies are needed to evaluate the clinical relevance of the multigene combination on COS.
Collapse
|
26
|
Liu S, Liu M, Li L, Li H, Qu D, Ren H, Su H, Zhang Y, Li Y. Patients With Deep Ovarian Suppression Following GnRH Agonist Long Protocol May Benefit From a Modified GnRH Antagonist Protocol: A Retrospective Cohort Study. Front Endocrinol (Lausanne) 2021; 12:618580. [PMID: 34326810 PMCID: PMC8314175 DOI: 10.3389/fendo.2021.618580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/29/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To verify if patients with deep ovarian suppression following gonadotropin releasing hormone (GnRH) agonist long protocol may benefit from a modified GnRH antagonist protocol based on luteinizing hormone (LH) levels. DESIGN Retrospective cohort study. SETTING University-based hospital. PATIENTS 110 patients exhibited ultra-low LH levels during ovarian stimulation using GnRH agonist long protocol. INTERVENTIONS As all the embryos in the first cycle were exhausted without being pregnant, these patients proposed to undergo a second cycle of ovarian stimulation. 74 of them were treated with a modified GnRH antagonist protocol based on LH levels. Other 36 patients were still stimulated following GnRH agonist long protocol. MAIN OUTCOME MEASURE The primary outcome was live birth rate (LBR). The second outcomes were biochemical pregnancy rate, clinical pregnancy rate (CPR), ongoing pregnancy rate (OPR) and cancellation rate. RESULTS Reproductive outcomes were much better in the modified GnRH antagonist protocol. The OPR and LBR were much higher in the GnRH antagonist protocol group than in the GnRH agonist long protocol group [odds ratio (OR) 3.82, 95% confidence interval (CI) 1.47, 10.61, P=0.018; OR 4.33, 95% CI 1.38, 13.60, P=0.008; respectively]. Meanwhile, the cancellation rate was much lower in the GnRH antagonist protocol group (OR 0.13, 95% CI 0.02, 0.72; P=0.014). Mean LH level during stimulation did not have a predictive value on live birth. However, it was independently associated with the occurrence of ongoing pregnancy (OR 2.70, 95% CI 1.25, 5.85; P=0.01). The results of sensitivity analyses were consistent with the data mentioned above. The patients got completely different and excellent clinical outcomes in their second cycles stimulated with the modified GnRH antagonist protocol. CONCLUSION Patients with deep ovarian suppression following GnRH agonist long protocol may benefit from a modified GnRH antagonist protocol based on LH levels.
Collapse
|
27
|
GA R, Cheemakurthi R, Kalagara M, Prathigudupu K, Balabomma KL, Mahapatro P, Thota S, Kommaraju AL, Muvvala SPR. Effect of LHCGR Gene Polymorphism (rs2293275) on LH Supplementation Protocol Outcomes in Second IVF Cycles: A Retrospective Study. Front Endocrinol (Lausanne) 2021; 12:628169. [PMID: 34046009 PMCID: PMC8147863 DOI: 10.3389/fendo.2021.628169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Infertility is a major concern for couples wanting to have progeny. Despite recent advances in the field of IVF, success rates still need improvement. Understanding the patient's variability and addressing it with personalized interventions may improve the success rate of fertilization and live births. This study examined the impact of a personalized pharmacogenomic approach on LH supplementation on the pregnancy and live birth rate outcomes in comparison with the traditional approaches. 193 patients undergoing a second IVF cycle in Krishna IVF Clinic received LH supplementation either as per the conventional methods or based on N312S (rs2293275) LHCGR gene polymorphism. Results showed a significant increase in pregnancy rate (P-value: 0.049) and a trend showing improvement in live birth rates (P-value: 0.082) when r-hLH supplementation protocol was decided as per the genotypes A/A, A/G, and G/G of the N312S variant in the respective patients. This stimulation regimen helped in providing optimum levels of r-hLH supplementation to patients with impaired hormone-receptor interacting activity, to achieve higher success in pregnancy and live birth rates.
Collapse
Affiliation(s)
- Ramaraju GA
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
- Department of Biotechnology, Institute of Science, Gitam (Deemed to be) University, Visakhapatnam, India
- *Correspondence: Ramaraju GA,
| | | | - Madan Kalagara
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | | | | | - Pranati Mahapatro
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | - Sivanarayana Thota
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | - Aruna Lakshmi Kommaraju
- Department of Biotechnology, Institute of Science, Gitam (Deemed to be) University, Visakhapatnam, India
| | | |
Collapse
|
28
|
Drakopoulos P, Bardhi E, Boudry L, Vaiarelli A, Makrigiannakis A, Esteves SC, Tournaye H, Blockeel C. Update on the management of poor ovarian response in IVF: the shift from Bologna criteria to the Poseidon concept. Ther Adv Reprod Health 2020; 14:2633494120941480. [PMID: 32844159 PMCID: PMC7416136 DOI: 10.1177/2633494120941480] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Despite the considerate progress to which assisted reproduction technology (ART)
has been subject since 1978, some issues remain unresolved. Notably, the
clinical management of patients with a poor ovarian response is still a
challenge in everyday practice, frustrating to both the patient and the
fertility expert. Poor ovarian responders (PORs) embody 9–24% of patients
undergoing ovarian stimulation, meaning that up to one in four patients conceals
a poor reproductive prognosis. The last decade has witnessed the attempts of the
medical community to standardize diagnosis of POR with the developing of the
Bologna Criteria and the subsequent evolution of the low prognosis patient
elaborated in the POSEIDON classification. The aim of this article is to
summarize all evidence concerning etiology and management of poor ovarian
response, including the most recent advances and future prospects in this
regard.
Collapse
Affiliation(s)
- Panagiotis Drakopoulos
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium. Department of Obstetrics and Gynecology, Medical School, University of Crete, Heraklion, Greece
| | - Erlisa Bardhi
- Center for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Liese Boudry
- Center for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alberto Vaiarelli
- G.EN.E.R.A., Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Antonis Makrigiannakis
- Department of Obstetrics and Gynecology, Medical School, University of Crete, Heraklion, Greece
| | - Sandro C Esteves
- ANDROFERT-Andrology and Human Reproduction Clinic, Brazil, São Paulo
| | - Herman Tournaye
- Center for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christophe Blockeel
- Center for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
29
|
Yan J, Tian Y, Gao X, Cui L, Ning Y, Cao Y, Chen Y, Peng F, You L, Liu F, Zhao H. A genome-wide association study identifies FSHR rs2300441 associated with follicle-stimulating hormone levels. Clin Genet 2020; 97:869-877. [PMID: 32185793 DOI: 10.1111/cge.13741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022]
Abstract
Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) play critical roles in female reproduction, while the underlying genetic basis is poorly understood. Genome-wide association studies (GWASs) of FSH and LH levels were conducted in 2590 Chinese females including 1882 polycystic ovary syndrome (PCOS) cases and 708 controls. GWAS for FSH level identified multiple variants at FSHR showing genome-wide significance with the top variant (rs2300441) located in the intron of FSHR. The A allele of rs2300441 led to a reduced level of FSH in the PCOS group (β = -.43, P = 6.70 × 10-14 ) as well as in the control group (β = -.35, P = 6.52 × 10-4 ). In the combined sample, this association was enhanced after adjusting for the PCOS status (before: β = -.38, P = 1.77 × 10-13 ; after: β = -.42, P = 3.33 × 10-16 ), suggesting the genetic effect is independent of the PCOS status. The rs2300441 explained sevenfold higher proportion of the FSH variance than the total variance explained by the two previously reported FSHR missense variants (rs2300441 R2 = 1.40% vs rs6166 R2 = 0.17%, rs6165 R2 = 0.03%). GWAS for LH did not identify any genome-wide significant associations. In conclusion, we identified genome-wide significant association between variants in FSHR and circulating FSH first, with the top associated variant rs2300441 might be a primary contributor at the population level.
Collapse
Affiliation(s)
- Jinting Yan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ye Tian
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China.,Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xingjian Gao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Linlin Cui
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China
| | - Yunna Ning
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China
| | - Yongzhi Cao
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China
| | - Yan Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Fuduan Peng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Li You
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China
| | - Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, China
| |
Collapse
|
30
|
Paschalidou C, Anagnostou E, Mavrogianni D, Raouasnte R, Klimis N, Drakakis P, Loutradis D. The effects of follicle-stimulating hormone receptor (FSHR) -29 and Ser680Asn polymorphisms in IVF/ICSI. Horm Mol Biol Clin Investig 2020; 41:hmbci-2019-0058. [PMID: 32114522 DOI: 10.1515/hmbci-2019-0058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/15/2020] [Indexed: 11/15/2022]
Abstract
Background The aim of this study was to analyze two different polymorphisms, Ser680Asn and -29 (G>A) promoter polymorphism, of the follicle-stimulating hormone receptor (FSHR) gene, individually but also in combination, in a sample of Greek women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI). Materials and methods One hundred and forty-one women undergoing IVF or ICSI and 94 controls were genotyped by real-time polymerase chain reaction (RT-PCR) for the two FSHR polymorphisms. The association of the alleles with the clinical, biochemical and other parameters concerning the controlled ovarian stimulation (COS) protocol and outcome was investigated, as well as the pregnancy rate. Results The study of each polymorphism individually revealed a positive correlation of the SerSer genotype (Ser680Asn polymorphism) with higher luteinizing hormone (LH) levels on the third day of the menstrual cycle. On the other hand, the A allele for the -29 (G>A) promoter polymorphism correlated with the increased number and quality of cumulus-oocyte complexes (COCs). No differences were detected when the different genotypes of the two polymorphisms were combined - the population study was grouped according to the number of polymorphic alleles they carried (0-4 alleles). Women who presented all polymorphic alleles, AsnAsn/AA, exhibited the lowest LH levels (2.62 ± 0.68 mIU/L), but were rarely detected (n = 2, 1.4% of the studied population). Conclusions The data from this study reflect that the investigation of the combination of polymorphisms, such as FSHR -29 and Ser680Asn, could offer a valuable tool in order to evaluate and anticipate the outcome of the ovulation induction protocols, especially in the group of patients with failed attempts.
Collapse
Affiliation(s)
- Chrysa Paschalidou
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Elli Anagnostou
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Despoina Mavrogianni
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Rami Raouasnte
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Nikiforos Klimis
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Peter Drakakis
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Loutradis
- 1st Department of Obstetrics and Gynecology, Division of Human Reproduction, IVF Unit, Alexandra Hospital, Medical School of National Kapodistrian University of Athens, 62 Sirinon Str, Athens 17562, P. Faliro, Greece, Phone: 0030 210 9833576
| |
Collapse
|
31
|
Lunding SA, Pors SE, Kristensen SG, Bøtkjær JA, Ramløse M, Jeppesen JV, Flachs EM, Pinborg A, Macklon KT, Pedersen AT, Andersen CY, Andersen AN. Ovarian cortical follicle density in infertile women with low anti-Müllerian hormone. J Assist Reprod Genet 2019; 37:109-117. [PMID: 31758514 DOI: 10.1007/s10815-019-01633-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/08/2019] [Indexed: 10/25/2022] Open
Abstract
PURPOSE To evaluate the association between anti-Müllerian hormone (AMH) and follicle density in infertile women with diminished ovarian reserve (DOR) versus women with normal ovarian reserve? METHODS Case-control study comparing follicle densities in ovarian cortex from 20 infertile women with DOR (AMH ≤ 5 pmol/L) and 100 controls with presumed normal ovarian reserve. RESULTS For all women > 25 years, the follicle densities correlated positively with AMH levels. For each single picomole per liter increase in AMH the follicle density increased by 6% (95% CI 3.3-8.5%) when adjusted for age. This was similar for women with DOR and controls. The follicle density was 1.8 follicles/mm3 cortical tissue in women with DOR versus 7.0 in age-paired controls (p = 0.04). The women with DOR had a median AMH of 1.8 pmol/L versus 14.4 pmol/L in the age-paired control group (p < 0.001). The ratio of AMH/follicle density was 1:1 (1.8/1.8) in women with DOR and 2:1 (14.4/7.0) in the age-paired controls. Analyses for gonadotropin receptor polymorphisms could not explain the characteristics of women with DOR. The proportion of secondary follicles was higher in women with DOR compared with controls (4.6% versus 1.4%, p = 0.0003). Pooling all patients, the follicle density decreased significantly by 7.7% for every year added (p < 0.0001). The women with DOR had lower follicle densities than the controls, but the slopes were equal in the two cohorts. CONCLUSIONS Follicle density and AMH concentrations correlate also when AMH is low. However, AMH is only a reliable marker for the true ovarian reserve when age is included in the estimation and women with DOR may have more follicles than their AMH levels imply.
Collapse
Affiliation(s)
- Stine Aagaard Lunding
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Maja Ramløse
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Janni Vikkelsø Jeppesen
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Esben Meulengracht Flachs
- Department of Occupational and Environmental Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Anja Pinborg
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- The Fertility Clinic, Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre Hospital, Hvidovre, Denmark
| | - Kirsten Tryde Macklon
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Anette Tønnes Pedersen
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Gynecology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anders Nyboe Andersen
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
32
|
Kochar IS, Jain R, Ramachandran S. Apparent mineralocorticoid excess: A case of hypertension in a child with delayed diagnosis leading to stroke. ACTA ACUST UNITED AC 2019. [DOI: 10.17352/ijcem.000041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
33
|
Nenonen HA, Lindgren IA, Prahl AS, Trzybulska D, Kharraziha I, Hultén M, Giwercman YL, Henic E. The N680S variant in the follicle-stimulating hormone receptor gene identifies hyperresponders to controlled ovarian stimulation. Pharmacogenet Genomics 2019; 29:114-120. [PMID: 30829738 PMCID: PMC6587209 DOI: 10.1097/fpc.0000000000000374] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To study if the follicle-stimulating hormone receptor (FSHR) variant asparagine/serine in amino acid 680 (N680S) can predict hypersensitivity to gonadotropins in women undergoing assisted reproduction. PATIENTS AND METHODS In this retrospective study, 586 women undergoing their first in-vitro fertilisation treatment were enroled, and their FSHR N680S genetic variant was analysed. The main outcome measures were number of retrieved oocytes and any grade of ovarian hyperstimulation syndrome (OHSS). Experimental studies were performed on FSHR variants transfected into eukaryotic cells treated with 1-90 IU recombinant follicle-stimulating hormone. The receptors' ability to induce a second messenger 3',5'-cyclic AMP was measured. RESULTS The proportion of women who developed OHSS was 6% (n=36). None of the women who developed this condition had the homozygous serine variant. The N680S polymorphism in the FSHR was associated with the condition, Ptrend (genotype)=0.004 and Pallelic (alleles)=0.04. Mean oocyte number was 11±6 in women without OHSS and 16±8 in women who developed OHSS (P=0.001), despite exposure to lower total hormonal dose in the latter group. The odds ratio for developing OHSS in carriers of the asparagine allele was 1.7 (95% confidence interval: 1.025-2.839, P=0.04). A higher receptor activity in cells expressing asparagine compared with the serine was also evident at all concentrations of recombinant follicle-stimulating hormone used (P<0.05 for all). CONCLUSION This study confirms previous findings regarding higher hormonal sensitivity in carriers of asparagine in the N680S position. These women are at higher risk for OHSS during in-vitro fertilisation. Genetic testing could identify those at highest risk to develop this adverse effect.
Collapse
Affiliation(s)
- Hannah A. Nenonen
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Ida A. Lindgren
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Alexandra S. Prahl
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Dorota Trzybulska
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Isabella Kharraziha
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Mathilda Hultén
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Yvonne L. Giwercman
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
| | - Emir Henic
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Lund
- Department of Translational Medicine, Reproductive Medicine Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
34
|
Ghaderian SMH, Akbarzadeh R, Mohajerani F, Khodaii Z, Salehpour S. The implication of single-nucleotide polymorphisms in ovarian hyperstimulation syndrome. Mol Reprod Dev 2019; 86:964-971. [PMID: 31115963 DOI: 10.1002/mrd.23171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 01/05/2023]
Abstract
Ovarian hyperstimulation syndrome (OHSS) is an undesirable complication in the course of ovarian stimulation. This kind of stimulation is aimed at acquiring a sufficient number of high-quality oocytes in in vitro fertilization (IVF). Whereas the predisposition to OHSS could be impacted by genetic polymorphisms in susceptible genes, the present study has been jointly conducted with an Iranian cohort to scrutinize its relevant implication. Genomic DNA was extracted from blood samples of patients with a normal ovarian response (NOR) or with OHSS. Samples were analyzed to detect polymorphisms MTHFR rs1801131, MTHFR rs1801133, AMHR2 rs2002555, LHCGR rs2293275, PGR rs10895068, and SERPINE1 rs1799889. Variations of MTHFR, AMHR2, LHCGR, and PGR genes were significantly associated with the developing OHSS. After correction for multiple analysis, this difference was not evident for PGR genotypes. The polymorphic alleles of MTHFR (rs1801131 C-allele and rs1801133 T-allele), AMHR2 (rs2002555 G-allele), and LHCGR (rs2293275 G-allele) were significantly more prevalent among patients with OHSS compared to those in the NOR group. In contrast, the minor allele of PGR single-nucleotide polymorphism (SNP) (rs10895068, A-allele) was more prominent among patients with a NOR than those with OHSS. No significant difference was observed in genotypes or alleles of SERPINE1 rs1799889. The observations indicated that the minor alleles of MTHFR, AMHR2, and LHCGR genes could be considered an independent risk factor in susceptibility to OHSS. Nevertheless, polymorphic allele in the PGR rs10895068 SNP contributes to preventing OHSS occurrence. Therefore, it can be argued that these genes have a significant impact on OHSS.
Collapse
Affiliation(s)
| | - Reza Akbarzadeh
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Fatemeh Mohajerani
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Zohreh Khodaii
- Dietary Supplements and Probiotics Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Saghar Salehpour
- Preventative Gynecology Research Center (PGRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Abrahami N, Izhaki I, Younis JS. Do young women with unexplained infertility show manifestations of decreased ovarian reserve? J Assist Reprod Genet 2019; 36:1143-1152. [PMID: 31115740 DOI: 10.1007/s10815-019-01467-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To investigate whether unexplained infertility at a young age demonstrates manifestations of decreased ovarian reserve. METHODS A total of 100 women were divided into two equally sized groups. The study group comprised women aged ≤ 37 years diagnosed with unexplained infertility, and the control group included age-matched women with either mechanical factor or severe male factor infertility. RESULTS Both groups were comparable in their basic characteristics. Overall, women with unexplained infertility presented with inferior ovarian reserve results set against women of the control group. The number of ≥ 14-mm follicles on the day of hCG administration was significantly lower in the study compared with the control group (7.0 ± 4.5 vs. 10.4 ± 4.1 follicles, respectively, P < 0.001). Likewise, basal serum FSH was higher in the study compared with the control group (8.4 ± 5.5 vs. 6.4 ± 1.7 IU/L, respectively, P = 0.015), while antral follicle count was lower (10.9 ± 6.6 vs. 16.2 ± 6.6 follicles, respectively, P < 0.001). Furthermore, women with unexplained infertility required a higher total dose of FSH for ovarian stimulation (2,923 ± 1,701 vs. 2,196 ± 941 IU/L, respectively, P = 0.010), but exhibited a lower number of retrieved oocytes (9.3 ± 6.3 vs. 15.6 ± 7.9 oocytes, respectively, P < 0.001), alongside a lower number of achieved embryos (5.3 ± 4.0 vs. 8.0 ± 4.7 embryos, respectively, P = 0.002). Interestingly, the cumulative clinical pregnancy rate was not significantly different between the two groups (44% vs. 58%, respectively, P = 0.163). CONCLUSIONS Young women ≤ 37 years of age with unexplained infertility have clear manifestations of sub-optimal ovarian reserve set against controls. Our findings suggest that unexplained infertility at a young age may be a risk factor for developing poor ovarian response, specifically as a quantitative, rather than a qualitative, risk factor.
Collapse
Affiliation(s)
- Noa Abrahami
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel.,Reproductive Medicine Unit, Department of Obstetrics & Gynecology, Baruch-Padeh Medical Center, Poriya, Israel
| | - Ido Izhaki
- Department of Evolutionary and Environmental Biology, University of Haifa, Haifa, Israel
| | - Johnny S Younis
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel. .,Reproductive Medicine Unit, Department of Obstetrics & Gynecology, Baruch-Padeh Medical Center, Poriya, Israel.
| |
Collapse
|
36
|
Errázuriz J, Romito A, Drakopoulos P, Frederix B, Racca A, De Munck N, Tournaye H, De Vos M, Blockeel C. Cumulative Live Birth Rates Following Stimulation With Corifollitropin Alfa Compared With hp-hMG in a GnRH Antagonist Protocol in Poor Ovarian Responders. Front Endocrinol (Lausanne) 2019; 10:175. [PMID: 30967840 PMCID: PMC6439614 DOI: 10.3389/fendo.2019.00175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Bologna criteria poor ovarian responders have a very low prognosis. Although, it has been proposed that LH supplementation could be beneficial in women with previous hypo-response to FSH. There are no studies comparing the cumulative live birth rates (LBRs) between corifollitropin alfa (CFA) and highly purified human menopausal gonadotrophin (hp-hMG). Objective: To compare cumulative LBRs in Bologna poor ovarian responders undergoing ovarian stimulation with CFA followed by hp-hMG vs. hp-hMG alone in a GnRH antagonist protocol. Design: This is a retrospective cohort study. We included in total 917 poor responders fulfilling the Bologna criteria for poor ovarian response (POR) at a university-affiliated tertiary center from January 2011 until March 2017. Patients were administered either fixed daily doses of 300-450 IU of hp-hMG (group A) or a single dose of 150 μg of CFA followed by daily injections of ≥300 IU of hp-hMG from Day 8 of stimulation until the day of ovulation trigger (group B), in a fixed GnRH antagonist protocol. Results: LBRs after fresh embryo transfer (ET) were similar in group A 71/510 (14%) and B 42/407 (10%). Cumulative LBR per cycle was significantly higher in group A (16.9%) compared to group B (11.8%); (P = 0.03). However, logistic regression analysis showed no association between the type of gonadotropin administered and cumulative LBR. Only age was significantly associated with cumulative LBR (OR = 0.93, P = 0.007). Conclusion: Cumulative LBRs are similar in Bologna poor responders stimulated with CFA followed by hp-hMG compared to hp-hMG monotreatment in an antagonist protocol.
Collapse
Affiliation(s)
- Joaquín Errázuriz
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Departamento de Ginecología y Obstetricia, Clínica Alemana, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile
| | - Alessia Romito
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Ginecological-Obstetrical and Urological Sciences, Sapienza University, Rome, Italy
| | - Panagiotis Drakopoulos
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Billie Frederix
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Surgical and Clinical Science, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Analissa Racca
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Academic Unit of Obstetrics and Gynecology, Azienda Ospedaliera Universitaria San Martino (IRCCS), University of Genoa, Genova, Italy
| | - Neelke De Munck
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Obstetrics and Gynecology, University of Zagreb-School of Medicine, Zagreb, Croatia
| | - Herman Tournaye
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Michel De Vos
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christophe Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Obstetrics and Gynecology, University of Zagreb-School of Medicine, Zagreb, Croatia
| |
Collapse
|
37
|
Alviggi C, Conforti A, Santi D, Esteves SC, Andersen CY, Humaidan P, Chiodini P, De Placido G, Simoni M. Clinical relevance of genetic variants of gonadotrophins and their receptors in controlled ovarian stimulation: a systematic review and meta-analysis. Hum Reprod Update 2019; 24:599-614. [PMID: 29924306 DOI: 10.1093/humupd/dmy019] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/12/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Genotype has been implicated in the outcome of ovarian stimulation. The analysis of patient-specific genotypes might lead to an individualized pharmacogenomic approach to controlled ovarian stimulation (COS). However, the validity of such an approach remains to be established. OBJECTIVE AND RATIONALE To define the impact of specific genotype profiles of follicle-stimulating hormone, luteinizing hormone and their receptors (FSHR, LHR and LHCGR) on ovarian stimulation outcome. Specifically, our aim was to identify polymorphisms that could be useful in clinical practice, and those that need further clinical investigation. SEARCH METHODS A systematic review followed by a meta-analysis was performed according to the Cochrane Collaboration and Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines without time restriction. We searched the PubMed/MEDLINE, Cochrane Library, SCOPUS and EMBASE databases to identify all relevant studies published before January 2017. Only clinical trials published as full-text articles in peer-reviewed journals were included. The primary outcome was the number of oocytes retrieved. OUTCOMES Fifty-seven studies were assessed for eligibility, 33 of which were included in the qualitative and quantitative analyses. Data were independently extracted using quality indicators. COS outcomes related to seven polymorphisms (FSHR [rs6165], FSHR [rs6166], FSHR [rs1394205], LHB [rs1800447], LHB [rs1056917], LHCGR [rs2293275] and LHCGR [rs13405728]) were evaluated. More oocytes were retrieved from FSHR (rs6165) AA homozygotes (five studies, 677 patients, weighted mean difference [WMD]: 1.85, 95% CI: 0.85-2.85, P < 0.001; I2 = 0%) than from GG homozygotes and AG heterozygotes (four studies, 630 patients, WMD: 1.62, 95% CI: 0.28-2.95, P = 0.020; I2 = 56%). Moreover, stimulation duration was shorter in FSHR (rs6165) AA homozygotes than in AG carriers (three studies, 588 patients, WMD -0.48, 95% CI: -0.87 to -0.10, P = 0.010, I2 = 44%). A higher number of oocytes (21 studies, 2632 patients WMD: 0.84, 95% CI: 0.19 to 1.49, P = 0.01, I2 = 76%) and metaphase II oocytes (five studies, 608 patients, WMD: 1.03, 95% CI: 0.01-2.05, P = 0.050, I2 = 0%) was observed in AA than in GG homozygote carriers. FSH consumption was significantly lower in FSHR (rs1394205) GG homozygotes (three studies, 411 patients, WMD: -1294.61 IU, 95% CI: -593.08 to -1996.14 IU, P = 0.0003, I2 = 99%) and AG heterozygotes (three studies, 367 patients, WMD: -1014.36 IU, 95% CI: -364.11 to -1664.61 IU, P = 0.002, I2 = 99%) than in AA homozygotes. WIDER IMPLICATIONS These results support the clinical relevance of specific genotype profiles on reproductive outcome. Further studies are required to determine their application in a pharmacogenomic approach to ovarian stimulation.
Collapse
Affiliation(s)
- Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy.,Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, and Azienda Ospedaliera-Universitaria di Modena, Italy
| | - Sandro C Esteves
- Androfert, Andrology and Human Reproduction Clinic, and Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Peter Humaidan
- Fertility Clinic, Skive Regional Hospital, Skive, Denmark, and Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, and Azienda Ospedaliera-Universitaria di Modena, Italy
| |
Collapse
|
38
|
Conforti A, Esteves SC, Di Rella F, Strina I, De Rosa P, Fiorenza A, Zullo F, De Placido G, Alviggi C. The role of recombinant LH in women with hypo-response to controlled ovarian stimulation: a systematic review and meta-analysis. Reprod Biol Endocrinol 2019; 17:18. [PMID: 30728019 PMCID: PMC6366097 DOI: 10.1186/s12958-019-0460-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/28/2019] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To study the role of recombinant human LH supplementation in women with hypo-response to ovarian stimulation. METHODS We performed a systematic review and meta-analysis of prospective clinical trials in which recombinant FSH monotherapy protocols were compared with LH-supplemented protocols in hypo-responders. A search was conducted of the Scopus, MEDLINE databases without time or language restrictions. Primary outcome was clinical pregnancy rate. RESULTS Significantly higher clinical pregnancy rates (odds ratio: 2.03, P = 0.003), implantation rates (odds ratio: 2.62, P = 0.004) and number of oocytes retrieved (weight mean differences: 1.98, P = 0.03) were observed in hypo-responders supplemented with recombinant LH versus hypo-responders who underwent FSH monotherapy. No differences in terms of mature oocytes or miscarriage rates were found between the two groups. CONCLUSION In conclusion, our analysis confirms that women with a hypo-response to exogenous gonadotropins might benefit from LH supplementation. However, more trials are required before a definitive conclusion can be drawn.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy.
| | - Sandro C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil
| | - Francesca Di Rella
- Department of Senology, Medical Oncology, National Cancer Institute, IRCCS "Fondazione G.Pascale", Naples, Italy
| | - Ida Strina
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Pasquale De Rosa
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Alessia Fiorenza
- Unit of Obstetrics and Gynaecology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Fulvio Zullo
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) Consiglio Nazionale delle Ricerche, Naples, Italy
| |
Collapse
|
39
|
Conforti A, Esteves SC, Cimadomo D, Vaiarelli A, Di Rella F, Ubaldi FM, Zullo F, De Placido G, Alviggi C. Management of Women With an Unexpected Low Ovarian Response to Gonadotropin. Front Endocrinol (Lausanne) 2019; 10:387. [PMID: 31316461 PMCID: PMC6610322 DOI: 10.3389/fendo.2019.00387] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/29/2019] [Indexed: 12/04/2022] Open
Abstract
POSEIDON groups 1 and 2 patients respond poorly (<4 oocytes retrieved) or sub-optimally (4-9 oocytes retrieved) to gonadotropin stimulation despite the presence of adequate ovarian parameters, which negatively affect their cumulative chances of delivering a baby using Assisted Reproductive Technology. A polygenic trait involving gonadotropins and/or their receptors seems to be the primary pathophysiology mechanism explaining this phenomenon. The clinical management is mainly focused on maximizing oocyte yield as to increase the likelihood of having at least one euploid embryo for transfer. Indices such as FORT (follicle output rate) and FOI (follicle-to-oocyte index) may be used to determine if the ovarian reserve was properly explored during a previous ovarian stimulation. Testing for the presence of common polymorphisms affecting gonadotropins and/or their receptors can also be considered to identify patients at risk of hypo-response. An individualized estimation of the minimum number of oocytes needed to obtain at least one euploid embryo can assist counseling and treatment planning. Among currently existing pharmacological interventions, use of recombinant FSH in preference over urinary gonadotropin preparations, FSH dosage increase, and use of rLH supplementation may be considered -alone or combined- for optimally managing POSEIDON's groups 1 and 2 patients. However, given the recent introduction of the POSEIDON criteria, there is still a lack of studies examining the role of interventions specifically to patients classified as groups 1 and 2, thus making it an area for open research.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- *Correspondence: Alessandro Conforti
| | - Sandro C. Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil
- Department of Surgery, University of Campinas, Campinas, Brazil
- Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Danilo Cimadomo
- GENERA, Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Alberto Vaiarelli
- GENERA, Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Francesca Di Rella
- Medical Oncology, Department of Senology, National Cancer Institute, IRCCS Fondazione G. Pascale, Naples, Italy
| | | | - Fulvio Zullo
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
40
|
Chambers AE, Fairbairn C, Gaudoin M, Mills W, Woo I, Pandian R, Stanczyk FZ, Chung K, Banerjee S. Soluble LH-HCG receptor and oestradiol as predictors of pregnancy and live birth in IVF. Reprod Biomed Online 2018; 38:159-168. [PMID: 30598377 DOI: 10.1016/j.rbmo.2018.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/23/2018] [Accepted: 11/01/2018] [Indexed: 01/03/2023]
Abstract
RESEARCH QUESTION Circulating soluble LH-HCG receptor (sLHCGR) is a first-trimester marker for screening pregnancy pathologies and predicts premature or multiple births before fertility treatment. Oestradiol per oocyte at ovulation induction predicts IVF treatment outcomes. We asked whether sLHCGR levels are stable during fertility treatment and whether, alone or with oestradiol, they could improve prediction of fertility treatment outcomes. DESIGN Serum sLHCGR, anti-Müllerian hormone [AMH] and oestradiol were measured in patients undergoing IVF. Antral follicle count before ovarian stimulation and oocyte yield were used to establish sLHCGR- oocyte ratio (SOR), sLHCGR- antral follicle ratio (SAR), oestradiol at trigger per oocyte (oestradiol-oocyte ratio [EOR]) and oestradiol at trigger per antral follicle (oestradiol-antral follicle ratio [EAR]). RESULTS The relatively stable sLHCGR was negatively related to AMH when oocyte yield was high. The sLHCGR levels were proportional (r = 0.49) to oestradiol at early cycle (day-3). Pregnancy and live birth were highest at low sLHCGR (≤1.0 pmol/ml) and SOR (≤ 0.1 pmol/ml/oocyte). A total of 86-89% of live births in IVF treatment were within the cut-off parameters of SAR and SOR (0.5 pmol/ml) and EAR and EOR (380 pg/ml). For failed pregnancy, age, SOR and EOR together had positive and negative predictive values of 0.841 and 0.703, respectively. CONCLUSIONS sLHCGR levels are negatively related to AMH when oocyte yield is high. High early cycle sLHCGR is associated with elevated day-3 oestradiol. Low sLHCGR and SOR are indicators of increased clinical pregnancy and live birth rates. Patient age and SOR, combined with EOR, might improve prediction of IVF treatment outcomes.
Collapse
Affiliation(s)
| | - Craig Fairbairn
- GCRM Glasgow Centre For Reproductive Medicine, 21 Fifty Pitches Way, Cardonald Business Park, Glasgow, G51 4FD, UK
| | - Marco Gaudoin
- GCRM Glasgow Centre For Reproductive Medicine, 21 Fifty Pitches Way, Cardonald Business Park, Glasgow, G51 4FD, UK
| | - Walter Mills
- Origin Biomarkers, Biocity Scotland, B'oness Road, Newhouse, Lanarkshire ML1 5UH, UK
| | - Irene Woo
- Division of Reproductive Endocrinology and Infertility, University of Southern California, Keck School of Medicine, 1127 Wilshire Blvd., Los Angeles California, CA 90017, USA
| | - Raj Pandian
- Pan Laboratories, 15375 Barranca Parkway, Irvine California, USA
| | - Frank Z Stanczyk
- Division of Reproductive Endocrinology and Infertility, University of Southern California, Keck School of Medicine, 1127 Wilshire Blvd., Los Angeles California, CA 90017, USA
| | - Karine Chung
- Division of Reproductive Endocrinology and Infertility, University of Southern California, Keck School of Medicine, 1127 Wilshire Blvd., Los Angeles California, CA 90017, USA
| | - Subhasis Banerjee
- Origin Biomarkers, Biocity Scotland, B'oness Road, Newhouse, Lanarkshire ML1 5UH, UK
| |
Collapse
|
41
|
Wang D, Du X, Li Y, Li Q. A polymorphism in the transcriptional regulatory region strongly influences ovine FSHR mRNA decay. Reprod Domest Anim 2018; 54:83-90. [PMID: 30153358 DOI: 10.1111/rda.13316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
Follicle-stimulating hormone receptor (FSHR) is an important G protein-coupled receptor, which is required for steroidogenesis, follicular development and female infertility. Here, we report a novel polymorphism in the 3'-UTR that strongly influences ovine FSHR mRNA decay. The partial 3'-UTR sequence of Hu sheep FSHR gene was isolated and characterized, and a polymorphism (c.2327A>G) was identified. Luciferase assay and qRT-PCR showed that c.2327A>G polymorphism in the 3'-UTR exerts a strong regulatory role in FSHR transcription. This regulatory role is achieved by affecting FSHR mRNA decay. Furthermore, the c.2327A>G mutation in the 3'-UTR influences ARE (AU-rich element, a cis-acting element promoting mRNA decay)-mediated mRNA decay of Hu sheep FSHR gene. Together, our study identified a novel polymorphism and elucidated a new mechanism underlying transcriptional regulation of FSHR in mammals.
Collapse
Affiliation(s)
- Dedi Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yinxia Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
42
|
Lindgren I, Nenonen H, Henic E, Bungum L, Prahl A, Bungum M, Leijonhufvud I, Huhtaniemi I, Yding Andersen C, Lundberg Giwercman Y. Gonadotropin receptor variants are linked to cumulative live birth rate after in vitro fertilization. J Assist Reprod Genet 2018; 36:29-38. [PMID: 30232643 PMCID: PMC6338601 DOI: 10.1007/s10815-018-1318-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 01/06/2023] Open
Abstract
Purpose The objective was to investigate if the gonadotropin receptor variants N680S (N: asparagine, S: serine, rs6166) in the follicle-stimulating hormone receptor (FSHR) and N312S (rs2293275) in the luteinizing hormone/human chorionic gonadotropin receptor (LHCGR) predicted cumulative live birth rate after in vitro fertilization (IVF). Methods A total of 665 women were consecutively enrolled for IVF during the period 2007–2016. Inclusion criteria were < 40 years of age, body mass index < 30 kg/m2, non-smoking, regular menstruation cycle of 21–35 days, and bilateral ovaries. A blood sample was drawn for endocrine hormonal analysis and for DNA extraction with subsequent genotyping of the FSHR N680S and LHCGR N312S polymorphisms. Statistical analyses were done on all completed IVF cycles. Results Women homozygous for S in both receptors combined (4S) had significantly higher live birth rate compared to those with other receptor variants when combining the first three IVF cycles (OR = 2.00, 95% CI [1.02, 3.92], p = 0.043). Cumulatively higher chance of live birth rate, during all IVF cycles, was also evident (HR = 1.89, 95% CI [1.00, 3.57], p = 0.049). Conclusions Gonadotropin receptor variants are promising candidates for the prediction of the possibility to have a baby to take home after IVF treatment.
Collapse
Affiliation(s)
- I. Lindgren
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - H. Nenonen
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - E. Henic
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - L. Bungum
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - A. Prahl
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - M. Bungum
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - I. Leijonhufvud
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - I. Huhtaniemi
- Hammersmith Campus, Institute of Reproductive and Developmental Biology, Imperial College London, London, SW7 2AZ UK
| | - C. Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Y. Lundberg Giwercman
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| |
Collapse
|
43
|
Roque M, Bianco B, Christofolini DM, Barchi Cordts E, Vilarino F, Carvalho W, Valle M, Sampaio M, Geber S, Esteves SC, Parente Barbosa C. Pharmacogenetic algorithm for individualized controlled ovarian stimulation in assisted reproductive technology cycles. Panminerva Med 2018; 61:76-81. [PMID: 29916218 DOI: 10.23736/s0031-0808.18.03496-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Controlled ovarian stimulation (COS) is crucial for optimizing in-vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) success. Multiple factors influence the ovarian response to COS, making predictions about oocyte yields not so straightforward. As a result, the ovarian response may be poor or suboptimal, or even excessive, all of which have negative consequences for the affected patient. There is a group of patients that present with a suboptimal response to COS despite normal biomarkers of ovarian reserve, such as AFC and AMH. These patients have a lower number of retrieved oocytes than what was expected based on their ovarian reserve, thus showing the inadequacy of using only the traditional ovarian reserve biomarkers to predict the ovarian response. Suboptimal response to COS might be related to ovarian sensitivity to exogenous gonadotropins modulated by genetic factors. The understanding of the gene polymorphisms related to reproductive function can help to improve the clinical management of this patient population and to explain some of the individual patient variability in response to COS. The development of a pharmacogenetic approach concerning COS in the context of assisted reproduction seems attractive as it might help to understand the relationship between genetic variants and ovarian response to exogenous gonadotropins. The patient's genetic profile could be used to select the most appropriate gonadotropin type, predict the optimal dosage for each drug, develop a cost-effective treatment plan, maximize the success rates, and lastly, decrease the time-to-pregnancy.
Collapse
Affiliation(s)
- Matheus Roque
- Center for Reproductive Medicine ORIGEN, Rio de Janeiro, Brazil - .,Universidade Federal de Minas Gerais, Belo Horizonte, Brazil -
| | - Bianca Bianco
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| | - Denise M Christofolini
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| | - Emerson Barchi Cordts
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| | - Fabia Vilarino
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| | - Waldemar Carvalho
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| | - Marcello Valle
- Center for Reproductive Medicine ORIGEN, Rio de Janeiro, Brazil
| | - Marcos Sampaio
- Center for Reproductive Medicine ORIGEN, Rio de Janeiro, Brazil
| | - Selmo Geber
- Center for Reproductive Medicine ORIGEN, Rio de Janeiro, Brazil.,Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sandro C Esteves
- Division of Urology, Department of Surgery, Andrology and Human Reproduction Clinic ANDROFERT, University of Campinas (UNICAMP), Campinas, Brazil
| | - Caio Parente Barbosa
- Department of Collective Health, Faculty of Medicine of ABC, Human Reproduction and Genetic Center, Santo André, Brazil
| |
Collapse
|
44
|
Younis JS, Laufer N. Recombinant luteinizing hormone supplementation to recombinant follicle stimulating hormone therapy in gonadotropin releasing hormone analogue cycles: what is the evidence? Curr Med Res Opin 2018; 34:881-886. [PMID: 29292650 DOI: 10.1080/03007995.2017.1417827] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To look into current evidence exploring the added value of rLH supplementation to rFSH in GnRH analogues cycles, to identify groups of women that still have no evidence for adjuvant rLH therapy and to discuss ways that may advance research on this topic. METHODS Eight systematic reviews and meta-analyses exploring the benefit for pregnancy achievement of rLH supplementation, excluding other LH activity preparations, to GnRH analogues cycles in the ART setting were thoroughly evaluated. RESULTS Evidence exists to show that rLH supplementation seems to have added value for pregnancy achievement in women with poor ovarian response and in women ≥35 years of age employing the GnRH agonist protocol, while the evidence is still debatable when the GnRH antagonist is administered. In the general infertile population, rLH supplementation does not have added value in the GnRH-antagonist cycles while the evidence is still controversial when the GnRH agonist is employed. Whether rLH supplementation may have a benefit in some young, normo-gonadotropic women, who may develop LH deficiency following GnRH analogues, is still to be shown. The main task remains how to identify subgroups of women that may benefit from rLH supplementation. CONCLUSION An accurate definition of the LH threshold in GnRH analogue treated cycles may contribute to the discussion of which subgroups of women may benefit from adjuvant rLH therapy.
Collapse
Affiliation(s)
- Johnny S Younis
- a Reproductive Medicine, Department of Obstetrics and Gynecology , Poriya Medical Center , Tiberias , Israel
- b Faculty of Medicine in Galilee , Bar Ilan University , Tiberias , Israel
| | - Neri Laufer
- c Reproductive Medicine, Department of Obstetrics and Gynecology , Hadassah-Hebrew University Medical Center , Ein-Kerem , Jerusalem , Israel
- d Hadassah Medical School , Hebrew University , Jerusalem , Israel
| |
Collapse
|
45
|
Soave I, Marci R. Exogenous luteinizing hormone supplementation in controlled ovarian stimulation: why, when and to whom? Curr Med Res Opin 2018; 34:939-941. [PMID: 29415578 DOI: 10.1080/03007995.2018.1438380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ilaria Soave
- a Department of Medical and Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology , University of Rome "Sapienza" , Rome , Italy
| | - Roberto Marci
- b Department of Morphology, Surgery and Experimental Medicine , University of Ferrara , Ferrara , Italy
| |
Collapse
|
46
|
Kalinderi K, Asimakopoulos B, Nikolettos N, Manolopoulos VG. Pharmacogenomics in IVF: A New Era in the Concept of Personalized Medicine. Reprod Sci 2018; 26:1313-1325. [PMID: 29587614 DOI: 10.1177/1933719118765970] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pharmacogenomics is a promising approach in the field of individualized medicine in in vitro fertilization (IVF) treatment that aims to develop optimized pharmacotherapy depending on the genetic background of each infertile woman, thus to ensure maximum effectiveness of the medication used, with minimal side effects. The unique genetic information of each infertile woman, in combination with already known, as well as new predictors of ovarian response and the progress of pharmacoepigenomics, is anticipated to greatly benefit the process of controlled ovarian stimulation. This review analyses current data on IVF pharmacogenomics, a new approach that is gradually moving to the frontline of modern IVF treatment.
Collapse
Affiliation(s)
- Kallirhoe Kalinderi
- Department of General Biology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,3rd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Byron Asimakopoulos
- Laboratory of Physiology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Nikos Nikolettos
- Laboratory of Reproductive Physiology-In Vitro Fertilization, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
47
|
G A R, Cheemakurthi R, Prathigudupu K, Balabomma KL, Kalagara M, Thota S, Kota M. Role of Lh polymorphisms and r-hLh supplementation in GnRh agonist treated ART cycles: A cross sectional study. Eur J Obstet Gynecol Reprod Biol 2018; 222:119-125. [PMID: 29408742 DOI: 10.1016/j.ejogrb.2018.01.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/23/2017] [Accepted: 01/19/2018] [Indexed: 11/29/2022]
Abstract
STUDY OBJECTIVES To investigate the effect of N312S polymorphism in the LHCGR gene as a predictive pharmacogenetic marker on clinical and embryological parameters and determining the need of r-hLH supplementation combine with r-hFSH in patients undergoing ART treatment. STUDY DESIGN In a cross-sectional study, a retrospective analysis of women (n = 553), who underwent controlled ovarian stimulation treatment protocol was conducted during the years 2012-2014. R-hFSH (Gonal-F, Merck Serono) was administered to all patients undergoing ART cycle after initiating long luteal gonadotrophin-releasing hormone (GnRH) agonist down-regulation. R-hLH was supplemented based on P.C. Wong criteria. N312S genotype was determined using sequencing methodology. The mean r-hFSH, r-hLH doses, total number of oocytes, cleavage rates of embryos and clinical pregnancy were recorded. The association between the r-hLH supplementation and LHCGR N312S polymorphism and clinical pregnancy rates was determined using regression analysis by SPSS. RESULTS 19.7% of women were homozygous for A allele encoding asparagine (N/N), 45.7% were heterozygous (N/S) and 34.6% were homozygous (S/S) for G allele encoding serine. Women heterozygous (N/S) or homozygous (S/S) for serine showed a higher requirement for r-hLH (OR, 95% p-trend = <0.0001) compared to those homozygous for asparagine (N/N). Homozygous G allele was also associated with higher daily and total r-hLH dose per treatment cycle p-trend = <0.0001. Though, the total no of oocytes (14.87 ± 4.95 vs 12.98 ± 5.39 and 13.58 ± 5.45), Gr-I quality embryos (2.61 ± 1.81 vs 2.18 ± 1.96 and 1.98 ± 2.05) were significantly higher in women homozygous for A allele compared to women with heterozygous and homozygous for G allele, clinical pregnancy rates were significantly more in women with for G allele after excluding patients with PCOS and endometriosis conditions (P < 0.04). CONCLUSION The present findings reveal that women heterozygous and homozygous for G allele required higher doses of r-hLH supplementation and these women were shown to have higher clinical pregnancy rates.
Collapse
Affiliation(s)
- Ramaraju G A
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Ravikrishna Cheemakurthi
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Kavitha Prathigudupu
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Kavitha Lakshmi Balabomma
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Madan Kalagara
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Sivanarayana Thota
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| | - Muralikrishna Kota
- Center for Assisted Reproduction, Krishna IVF Clinic, Maharanipeta, Visakhapatnam 530002, Andhra Pradesh, India.
| |
Collapse
|
48
|
Robeva R, Andonova S, Tomova A, Kumanov P, Savov A. LHCG receptor polymorphisms in PCOS patients. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2017.1423246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Ralitsa Robeva
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Silvia Andonova
- Medical Faculty, National Genetic Laboratory, USHATOG “Maichin Dom”, Medical University of Sofia, Sofia, Bulgaria
| | - Analia Tomova
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Philip Kumanov
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Alexey Savov
- Medical Faculty, National Genetic Laboratory, USHATOG “Maichin Dom”, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
49
|
Genetics of gonadotropins and their receptors as markers of ovarian reserve and response in controlled ovarian stimulation. Best Pract Res Clin Obstet Gynaecol 2017; 44:15-25. [PMID: 28506471 DOI: 10.1016/j.bpobgyn.2017.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 02/03/2017] [Accepted: 04/01/2017] [Indexed: 01/11/2023]
Abstract
Several controlled ovarian stimulation (COS) protocols have been developed to increase the yield of mature oocytes retrieved in assisted reproductive techniques (ARTs). The ovarian reserve (OR) influences the COS response, and it represents the main parameter that helps clinicians in refining clinical treatments in the perspective of a "personalized" ART. This approach is even more needed in particular conditions such as poor OR or polycystic ovary syndrome. Follicle-stimulating hormone, luteinizing hormone, and human chorionic gonadotropin are currently used in COS at different combinations and with different efficacies, even if the best approach definition is controversial. Differences in individual-specific ovarian response to gonadotropin stimulation can be due to alterations of genes encoding for hormones or their receptors. In particular, FSHB c.-211G>T, FSHR p.Asn680Ser, and c.-29G>A SNP allelic combinations may be used as OR and COS response markers. The purpose of this review is to highlight the evidence-based relevance of mutations and polymorphisms in gonadotropins and their receptor genes as predictive markers of OR and COS response to achieve fine-tuned therapeutic regimens.
Collapse
|
50
|
Bang AK, Nordkap L, Almstrup K, Priskorn L, Petersen JH, Rajpert-De Meyts E, Andersson AM, Juul A, Jørgensen N. Dynamic GnRH and hCG testing: establishment of new diagnostic reference levels. Eur J Endocrinol 2017; 176:379-391. [PMID: 28077499 DOI: 10.1530/eje-16-0912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/05/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Gonadotropin-releasing hormone (GnRH) and human chorionic gonadotropin (hCG) stimulation tests may be used to evaluate the pituitary and testicular capacity. Our aim was to evaluate changes in follicular-stimulating hormone (FSH), luteinizing hormone (LH) and testosterone after GnRH and hCG stimulation in healthy men and assess the impact of six single nucleotide polymorphisms on the responses. DESIGN GnRH and hCG stimulation tests were performed on 77 healthy men, 18-40 years (reference group) at a specialized andrology referral center at a university hospital. The potential influence of the tests was illustrated by results from 45 patients suspected of disordered hypothalamic-pituitary-gonadal axis. METHODS Baseline, stimulated, relative and absolute changes in serum FSH and LH were determined by ultrasensitive TRIFMA, and testosterone was determined by LC-MS/MS. RESULTS For the reference group, LH and FSH increased almost 400% and 40% during GnRH testing, stimulated levels varied from 4.4 to 58.8 U/L and 0.2 to 11.8 U/L and FSH decreased in nine men. Testosterone increased approximately 110% (range: 18.7-67.6 nmol/L) during hCG testing. None of the polymorphisms had any major impact on the test results. Results from GnRH and hCG tests in patients compared with the reference group showed that the stimulated level and absolute increase in LH showed superior identification of patients compared with the relative increase, and the absolute change in testosterone was superior in identifying men with Leydig cell insufficiency, compared with the relative increase. CONCLUSIONS We provide novel reference ranges for GnRH and hCG test in healthy men, which allows future diagnostic evaluation of hypothalamic-pituitary-gonadal disorders in men.
Collapse
Affiliation(s)
- A Kirstine Bang
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Loa Nordkap
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Kristian Almstrup
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Lærke Priskorn
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Jørgen Holm Petersen
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
- Department of BiostatisticsUniversity of Copenhagen, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Anna-Maria Andersson
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Anders Juul
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Niels Jørgensen
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| |
Collapse
|