1
|
Toner JP, Pirtea P. Luteinizing hormone's critical role in ovarian stimulation. Fertil Steril 2025; 123:31-40. [PMID: 39522745 DOI: 10.1016/j.fertnstert.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
|
2
|
Mannaerts B, Yding Andersen C, Howles CM. Does HCG and LH supplementation during ovarian stimulation improve clinical outcome? A evaluation of 30 years of clinical research. Reprod Biomed Online 2024; 50:104782. [PMID: 40294495 DOI: 10.1016/j.rbmo.2024.104782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 04/30/2025]
Abstract
This review evaluates the effect of HCG and LH supplementation during ovarian stimulation. Controlled trials were divided into four groups comparing the treatment effect of recombinant FSH (r-FSH) with urinary FSH (u-FSH), with human menopausal gonadotrophin (HMG), with r-FSH + recombinant LH (r-LH) and with rFSH + recombinant HCG (r-HCG). First r-FSH seemed to be more potent than u-FSH in downregulated women, which translated into more follicles and oocytes. In line, numerous trials comparing HMG (containing u-FSH + u-HCG) with r-FSH demonstrated that HMG recruited fewer follicles, thus providing fewer oocytes but resulted in slightly higher pregnancy rates after a first fresh embryo transfer. The latter may be explained by the higher potency of r-FSH resulting in more and higher premature progesterone rises. Prospective trials addressing r-FSH + r-LH compared with r-FSH could not demonstrate any difference in pregnancy rates in normal or poor responders. A placebo-controlled trial of r-HCG added to a fixed daily r-FSH dose revealed that r-HCG inhibited the growth of intermediate follicles, resulting in fewer oocytes, fewer embryos and lower pregnancy rates. In conclusion, the beneficial effect of HCG and LH supplementation on clinical outcome has never been definitely proven for any of the combined gonadotrophin products.
Collapse
Affiliation(s)
| | - Claus Yding Andersen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Fertility Clinic, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Colin M Howles
- ARIES Consulting Sàrl, Geneva, Switzerland; Deanery of Biomedical Sciences, University of Edinburgh, UK
| |
Collapse
|
3
|
Zheng M, Poulsen LC, Wang NF, Mamsen LS, Johannsen ML, Styrishave B, Grøndahl ML, Løssl K, Englund ALM, Skouby SO, Andersen CY. Progesterone and 17-hydroxy-progesterone concentrations in follicular fluid and serum reflect their production in granulosa and theca cells. Reprod Biomed Online 2024; 49:103853. [PMID: 38865783 DOI: 10.1016/j.rbmo.2024.103853] [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: 11/02/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 06/14/2024]
Abstract
RESEARCH QUESTION How is the production of progesterone (P4) and 17-hydroxy-P4 (17-OH-P4) regulated between theca cells and granulosa cells during the follicular phase, during ovulation and after transformation into a corpus luteum? DESIGN Three cohorts were examined: (i) 31 women undergoing natural and stimulated cycles, with serum hormone measurements taken every 3 days; (ii) 50 women undergoing ovarian stimulation, with hormone concentrations in serum and follicular fluid assessed at five time points during final follicle maturation; and (iii) 12 women undergoing fertility preservation, with hormone concentrations evaluated via the follicular fluid of small antral follicles. RESULTS In the early follicular phase, theca cells primarily synthesized 17-OH-P4 while granulosa cells produced limited P4, maintaining the P4:17-OH-P4 ratio <1. As follicles reached follicle selection at a diameter of approximately 10 mm, P4 synthesis in granulosa cells was up-regulated, but P4 was mainly accumulated in follicular fluid. During final maturation, enhanced activity of the enzyme HSD3B2 in granulosa cells enhanced P4 production, with the P4:17-OH-P4 ratio increasing to >1. The concentration of 17-OH-P4 in the luteal phase was similar to that in the follicular phase, but P4 production increased in the luteal phase, yielding a P4:17-OH-P4 ratio significantly >1. CONCLUSIONS The P4:17-OH-P4 ratio reflects the activity of granulosa cells and theca cells during the follicular phase and following luteinization in the corpus luteum. Managing the function of granulosa cells is key for reducing the concentration of P4 during ovarian stimulation, but the concerted action of FSH and LH on granulosa cells during the second half of the follicular phase makes this complex.
Collapse
Affiliation(s)
- M Zheng
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.
| | - L C Poulsen
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - N F Wang
- The Fertility Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - L S Mamsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - M L Johannsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark; Toxicology and Drug Metabolism Group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - B Styrishave
- Toxicology and Drug Metabolism Group, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M L Grøndahl
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - K Løssl
- The Fertility Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - A L M Englund
- The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - S O Skouby
- The Fertility Clinic, Herlev Hospital, Herlev, Denmark
| | - C Y Andersen
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark; The Fertility Clinic, Herlev Hospital, Herlev, Denmark.
| |
Collapse
|
4
|
Sa SW, Qiao XY, Zhang JC, Ma QH. Successful Transvaginal Oocyte Retrieval After Laparoscopic Management of Tubal Pregnancy. J Minim Invasive Gynecol 2024; 31:553-554. [PMID: 38705379 DOI: 10.1016/j.jmig.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Affiliation(s)
- Sha-Wei Sa
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, and Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China (all authors)
| | - Xiao-Yong Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, and Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China (all authors)
| | - Jin-Cheng Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, and Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China (all authors)
| | - Qian-Hong Ma
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, and Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China (all authors).
| |
Collapse
|
5
|
Doroftei B, Ilie OD, Anton N, Marcu OA, Scripcariu IS, Ilea C. A Narrative Review Discussing the Efficiency of Personalized Dosing Algorithm of Follitropin Delta for Ovarian Stimulation and the Reproductive and Clinical Outcomes. Diagnostics (Basel) 2023; 13:177. [PMID: 36672987 PMCID: PMC9858569 DOI: 10.3390/diagnostics13020177] [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: 11/11/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Follitropin delta is the third recombinant human follicle-stimulating hormone (r-hFSH) expressed in a host cell line of human fetal retinal origin that currently emphasizes that the actual tendency of administration is a personalized dosing algorithm based on the anti-Müllerian hormone (AMH) and body mass index (BMI) for ovarian stimulation. Methods: In this context, we aimed, in the present manuscript, to gather all available data published between 2018-2022 regarding the co-administration and administration of follitropin delta and the clinical outcomes reported following an in vitro fertilization (IVF). Results: Follitropin delta is non-inferior in contrast to its previously launched agents for ovarian stimulation, enhancing a similar-to-superior response reflected by both the reproductive and pregnancy outcomes in parallel with a low risk of ovarian hyperstimulation syndrome (OHSS), being well tolerated. The body weight and AMH level are factors that may influence the outcome in a patient. Despite controversy and results that refute these arguments on several occasions, follitropin delta exceeds the benefits of conventional dosing with either follitropin alfa or follitropin beta. Thus, all post hoc, derived analyses and subsets of patients that participated in subsequent studies support this statement. Conclusions: Despite the relatively limited spectrum of data in the current literature, most authors brought potent proof, supporting the subsequent use of this drug depending on the patient's profile and overcoming ethnic-related limitations. Although others contradict these observations, this topic and drug possess substantial potential, which is why additional studies are mandatory to fill the existing gaps in our knowledge and expand these experiences at a larger scale supported by the obtained reproductive and clinical outcomes that clearly indicate an overcoming of all limitations.
Collapse
Affiliation(s)
- Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, no 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street, no 3C, 700032 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Nicoleta Anton
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Olivia-Andreea Marcu
- Department of Preclinics, Faculty of Medicine and Pharmacy, University of Oradea, December 1 Market Street, no 10, 410068 Oradea, Romania
| | - Ioana-Sadyie Scripcariu
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Ciprian Ilea
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, no 34, 700038 Iasi, Romania
| |
Collapse
|
6
|
Lin H, Huang X, Zhao Y, Wang Y, Wang S, Hong F, Pan M, Liu L. Low-dose human chorionic gonadotropin supplementation initiated at the onset of ovarian stimulation can improve oocyte quality without impairing endometrial receptivity: Case series. Medicine (Baltimore) 2022; 101:e32175. [PMID: 36482630 PMCID: PMC9726314 DOI: 10.1097/md.0000000000032175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Whether continuous low-dose human chorionic gonadotropin (hCG) supplementation during controlled ovarian hyperstimulation (COH) can improve oocyte and embryo quality is still controversial in clinical practice. PATIENT CONCERNS We report the first case series of inadvertent COH in luteal-phase stimulation in the presence of endogenous or exogenous low-dose hCG. DIAGNOSES Patients were diagnosed with infertility. OUTCOMES The first two cases had inadvertent COH during preexisting pregnancy, and one of which produced more high-quality embryos (5 vs 1) in the presence of low hCG. Both cases had a live birth. The third case had 7 repeated failures of IVF, during which a total of 55 oocytes were obtained, but only 3 developed into transferable embryo. However, supplementation of 330 IU hCG per day from the onset of COH resulted in the recovery of one high-quality embryo and subsequent delivery of a healthy baby following fresh embryo transfer in eighth attemption. LESSONS In conclude, supplementation with low-dose hCG from the onset of ovarian stimulation can improve oocyte quality without impairing endometrial receptivity.
Collapse
Affiliation(s)
- Huizhen Lin
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaona Huang
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yue Zhao
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yangyang Wang
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Shasha Wang
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Fang Hong
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Mei Pan
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Liu Liu
- Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- * Correspondence: Liu Liu, Center of Reproductive Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# Qing chun East Road, Hangzhou 310016, PR China (e-mail: )
| |
Collapse
|
7
|
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
|
8
|
Coughlan C, Vitorino R, Melado L, Digma S, Sibal J, Patel R, Lawrenz B, Fatemi H. Evolution of serum progesterone levels in the very early luteal phase of stimulated IVF/ICSI cycles post hCG trigger: a proof of concept study. J Assist Reprod Genet 2022; 39:1095-1104. [PMID: 35391631 PMCID: PMC9107532 DOI: 10.1007/s10815-022-02474-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/19/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Studies have suggested that controlled ovarian hyperstimulation adversely affects endometrial receptivity due to advanced endometrial maturation. This adverse effect is mainly attributed to supraphysiological levels of both estrogen and progesterone identified in stimulated cycles. There is a paucity of published data investigating the very early luteal steroid profile following hCG trigger. AIM OF THE STUDY This prospective, observational study was undertaken to determine the increase in serum progesterone levels after human chorionic gonadotrophin (hCG) trigger in stimulated IVF/ICSI cycles. MATERIALS AND METHODS This proof-of-concept study included 11 patients requiring ovarian stimulation for IVF/ICSI and who planned to avail of pre-implantation genetic screening with embryo vitrification of their biopsied embryos at blastocyst stage. For each study participant, five additional blood samples were drawn at the following specific times in the stimulation cycle, on the morning (10.00-12.00) of the assigned day to induce final oocyte maturation with hCG trigger, immediately prior to administration of hCG for final oocyte maturation, 1 h, 2 h, and 36 h post hCG trigger. A prediction model, the Gompertz curve, was used to determine serum progesterone levels at intervals between the 2 h post hCG trigger sample and the day of oocyte retrieval. RESULTS Statistically significant increases in serum progesterone levels were identified following hCG administration as early as 1 h following trigger (P4 0.57 ng/ml, p < 0.05), 2 h following trigger (P4 0.88 ng/ml, p < 0.001) and on the day of oocyte retrieval (P4 9.68 ng/ml, p < 0.001). According to our prediction model, the Gompertz curve, the projected serum progesterone level at 4 h post trigger would have achieved a level of 1.45 ng/ml, 8 h post trigger of 3.04 ng/ml, and 12 h post trigger of 4.8 ng/ml. The very early and significant increases in serum progesterone following hCG trigger are clearly demonstrated in this study. CONCLUSION The endometrium is undoubtedly exposed to rapidly increasing serum progesterone levels post hCG trigger that would not be identified until much later in natural menstrual cycles. TRIAL REGISTRATION NUMBER This study is registered with clinicaltrials.gov under the identifier NCT04417569.
Collapse
Affiliation(s)
- Carol Coughlan
- ART Fertility Clinic Dubai, DMCC-F109B, Red Diamond Building, Al Sarayat Street, Dubai, UAE. .,ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE.
| | - R Vitorino
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| | - L Melado
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| | - S Digma
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| | - J Sibal
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| | - R Patel
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| | - B Lawrenz
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE.,Women's University Hospital Tuebingen, Calwers tr., 72076, Tuebingen, Germany
| | - H Fatemi
- ART Fertility Clinic Abu Dhabi, Royal Marina Village, Villa B22-23, Abu Dhabi, UAE
| |
Collapse
|
9
|
Fernández Sánchez M, Višnová H, Larsson P, Yding Andersen C, Filicori M, Blockeel C, Pinborg A, Khalaf Y, Mannaerts B. OUP accepted manuscript. Hum Reprod 2022; 37:1161-1174. [PMID: 35451013 PMCID: PMC9156848 DOI: 10.1093/humrep/deac061] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/03/2022] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION SUMMARY ANSWER WHAT IS KNOWN ALREADY STUDY DESIGN, SIZE, DURATION PARTICIPANTS/MATERIALS, SETTING, METHODS MAIN RESULTS AND THE ROLE OF CHANCE LIMITATIONS, REASONS FOR CAUTION WIDER IMPLICATIONS OF THE FINDINGS STUDY FUNDING/COMPETING INTEREST(S) TRIAL REGISTRATION NUMBER TRIAL REGISTRATION DATE DATE OF FIRST PATIENT’S ENROLMENT
Collapse
Affiliation(s)
- Manuel Fernández Sánchez
- IVI-RMA Seville, Seville, Spain
- Departament of Surgery, Universidad de Sevilla, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
- Fundacion IVI, Instituto Investigación Sanitaria La Fe, Valencia, Spain
| | | | - Per Larsson
- Global Biometrics, Ferring Pharmaceuticals, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital and Faculty of Health Science, Copenhagen University, Copenhagen, Denmark
| | | | - Christophe Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Anja Pinborg
- Fertility Clinic, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yacoub Khalaf
- Assisted Conception Unit, Guy’s Hospital, London, UK
| | - Bernadette Mannaerts
- Correspondence address. Reproductive Medicine & Maternal Health, Ferring Pharmaceuticals, Amager Strandvej 405, 2770 Kastrup, Denmark. E-mail:
| | | |
Collapse
|
10
|
The Use of Androgen Priming in Women with Reduced Ovarian Reserve Undergoing Assisted Reproductive Technology. Semin Reprod Med 2021; 39:207-219. [PMID: 34500477 DOI: 10.1055/s-0041-1735646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Androgen priming with either dehydroepiandrosterone (DHEA) or testosterone has been suggested as an adjunct to improve in vitro fertilization (IVF) outcomes in women with diminished ovarian reserve (DOR). Numerous studies have investigated the effects of both DHEA and testosterone on IVF outcome. The results were inconsistent, and the quality of most studies is substandard. Meta-analyses have consistently reported that DHEA does appear to significantly improve IVF outcome in women with predicted or proven poor ovarian response (POR), but these have included some normal responders and/or nonrandomized studies. Our meta-analyses including randomized controlled trials (RCTs) incorporating only women with DOR or POR suggest that DHEA confers no benefit. While meta-analyses of RCTs on the use of testosterone in women with DOR or POR showed an improved IVF outcome, most studies included are of low quality with high risk of bias. When analysis of data from studies of only low-risk bias was performed, such a benefit with testosterone was not observed. Although recruitment may well be a challenge, a large, well-designed RCT is, however, still warranted to investigate whether or not androgen priming with either DHEA or testosterone should be recommended as an adjuvant treatment for women with DOR or POR undergoing IVF.
Collapse
|
11
|
Siristatidis C, Stavros S, Dafopoulos K, Sergentanis T, Domali E, Drakakis P, Loutradis D. A Randomized Controlled Trial on the Efficacy and Safety of Low-Dose hCG in a Short Protocol with GnRH Agonist and Ovarian Stimulation with Recombinant FSH (rFSH) During the Follicular Phase in Infertile Women Undergoing ART. Reprod Sci 2021; 29:497-505. [PMID: 34254280 PMCID: PMC8275065 DOI: 10.1007/s43032-021-00683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/25/2021] [Indexed: 11/25/2022]
Abstract
Τhis study aims to investigate whether the addition of low-dose hCG throughout stimulation in infertile women undergoing IVF improves IVF outcome parameters. This is a prospective, multicenter, randomized, double-blind, placebo-controlled, Phase IIIb clinical study, conducted in three university IVF units. We studied whether the addition of 100 IU hCG/day to a short GnRH agonist IVF protocol from the onset of the follicular phase (group 1, n=40) or placebo (group 2, n=41) had any impact on the number of high-quality transferred embryos at day 2 and clinical pregnancy rates. The comparison encompassed descriptive statistics, and univariate and multivariate analyses. Concerning the primary outcomes, we found no differences in both the number of high-quality embryos (≥2) at day 3 [21/40 (52.5%) vs. 14/41 (34.2%), p=0.095] and clinical pregnancy rates [10/40 (25%) vs. 10/41 (24.4%), p=0.949], respectively. Similarly, there were no differences concerning the secondary outcomes preset for this trial. According to the results of the multivariate logistic regression analysis, no significant associations were noted for primary outcomes (clinical pregnancy: adjusted OR=0.89, 95% CI: 0.29–2.75; (≥2 excellent quality embryos at day 3: adjusted OR=0.54, 95% CI: 0.21–1.42, with group 1 set as reference category); similarly, no differences were noted with respect to secondary outcomes, except from the increased odds of ≥2 poor-quality embryos at day 3 occurring in group 2 (adjusted OR= 11.69, 95%CI: 1.29–106.19). The addition of low-dose hCG to a short GnRH agonist protocol for IVF does not improve the number of top-quality embryos and clinical pregnancy rates.
Collapse
Affiliation(s)
- Charalampos Siristatidis
- Assisted Reproduction Unit, Third Department of Obstetrics and Gynecology, "Attikon" Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini Str., 12642, Chaidari, Athens, Greece. .,Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, "Aretaieion" University Hospital, Medical School, National and Kapodistrian University of Athens, 76 Vas. Sofias Av, 11528, Athens, Greece.
| | - Sofoklis Stavros
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, "Alexandra" Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Av. and Lourou str., 11528, Athens, Greece
| | - Konstantinos Dafopoulos
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, University Hospital, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110, Larissa, Greece
| | - Theodoros Sergentanis
- Department of Clinical Therapeutics, "Alexandra" Hospital, Medical School, National and Kapodistrian University of Athens , 80 Vas. Sofias Av., 11528, Athens, Greece
| | - Ekaterini Domali
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, "Alexandra" Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Av. and Lourou str., 11528, Athens, Greece
| | - Peter Drakakis
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, "Alexandra" Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Av. and Lourou str., 11528, Athens, Greece
| | - Dimitrios Loutradis
- Assisted Reproduction Unit, First Department of Obstetrics and Gynecology, "Alexandra" Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Av. and Lourou str., 11528, Athens, Greece
| |
Collapse
|
12
|
Nataraja S, Yu H, Guner J, Palmer S. Discovery and Preclinical Development of Orally Active Small Molecules that Exhibit Highly Selective Follicle Stimulating Hormone Receptor Agonism. Front Pharmacol 2021; 11:602593. [PMID: 33519465 PMCID: PMC7845544 DOI: 10.3389/fphar.2020.602593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
An orally active follicle stimulating hormone receptor allosteric agonist would provide a preferred treatment for over 16 million infertile women of reproductive age in low complexity methods (ovulation induction-intrauterine insemination) or in high complexity methods (controlled ovarian stimulation-in vitro fertilization). We present two oral follicle stimulating hormone receptor allosteric agonist compounds that have the desired pharmacology, drug metabolism, pharmacokinetics, and safety profile for clinical use. These molecules provide a single agent suitable for ovulation induction-intrauterine insemination or controlled ovarian stimulation-in vitro fertilization that is more convenient for patients and achieves similar preclinical efficacy as rec-hFSH. TOP5668, TOP5300 were evaluated in vitro in Chinese hamster ovary cells transfected with individual glycoprotein receptors measuring cAMP (FSHR, LH/CGR, thyroid stimulating hormone receptor). TOP5668 was found to have solely follicle stimulating hormone receptor allosteric agonist activity while TOP5300 was found to have mixed follicle stimulating hormone receptor allosteric agonist and LHR-AA activity. Both compounds stimulated concentration-dependent increases in estradiol production from cultured rat granulosa cells in the presence or absence of low dose rec-hFSH, while only TOP5300 stimulated testosterone production from rat primary Leydig cells. In pooled human granulosa cells obtained from patients undergoing controlled ovarian stimulation-in vitro fertilization, TOP5300 stimulated 7-fold greater maximal estradiol response than rec-hFSH and TOP5668 was 10-fold more potent than TOP5300. Both TOP5300 and TOP5668 stimulated follicular development in immature rat to the same efficacy as recombinant follicle stimulating hormone. In mice treated with TOP5300, in the presence of low dose of follicle stimulating hormone, there were no differences in oocyte number, fertilization rate, and hatched blastocyst rate in mice with TOP5300 and low dose follicle stimulating hormone vs. reference proteins pregnant mare serum gonadotropin or high dose rec-hFSH. ADME/PK and safety profiles were favorable. In addition, there was no appreciable activity on thyroid hormones by TOP5300 in 14-days toxicological study in rat or dog. The selected lead compound, TOP5300 stimulated a more robust increase in estradiol production from granulosa-lutein cells from women with polycystic ovarian syndrome patient compared to rec-hFSH. Conclusions: Two novel oral FSHR allosteric agonist, TOP5668 and TOP5300, were found to mimic the biological activity of rec hFSH in preclinical studies. Both compounds led to folliculogenesis and superovulation in rat and mice. Specifically, TOP5300 led to a similar number of ovulated oocytes that fertilized and developed into hatched blastocysts in mice when compared to rec-hFSH. The safety profile demonstrated lack of toxicity.
Collapse
Affiliation(s)
| | | | - Joie Guner
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Stephen Palmer
- TocopheRx, Inc., Groton, MA, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
13
|
Lawrenz B, Coughlan C, Melado L, Digma S, Sibal J, Jean A, Fatemi HM. Step-Down of FSH- Dosage During Ovarian Stimulation - Basic Lessons to Be Learnt From a Randomized Controlled Trial. Front Endocrinol (Lausanne) 2021; 12:661707. [PMID: 33927696 PMCID: PMC8078176 DOI: 10.3389/fendo.2021.661707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/22/2021] [Indexed: 11/24/2022] Open
Abstract
A rise in serum progesterone in the late follicular phase is a well described adverse effect of ovarian stimulation for IVF/ICSI. Previous data suggest, that enhanced gonadotropin stimulation causes progesterone elevation and the incidence of premature progesterone elevation can be reduced by declining gonadotropin dosages. This randomized controlled trial (RCT) aimed to achieve a significant reduction of the progesterone level on the day of final oocyte maturation by a daily reduction of 12.5 IU rec-FSH from a follicle size of 14 mm in a GnRH-antagonist protocol. A total of 127 patients had been recruited (Control group (CG): 62 patients; Study group (SG): 65 patients). Due to drop out, data from 108 patients (CG: 55 patients; SG: 53 patients) were included into the analysis. Patients' basic parameters, gonadotropin (Gn)-starting dose, total Gn-stimulation dosage, the number of retrieved and mature oocytes as well as in the hormonal parameters on the day of trigger (DoT) were not statistically significantly different. However, through stepwise Gn-reduction of 12.5 IU/day in the SG, there was a statistically highly significant difference in the Gn-stimulation dosage on the day of trigger (p < 0.0001) and statistically significant associations for the DoT-P4-levels with the DoT-FSH-levels for both groups (CG: p = 0.001; SG: p = 0.0045). The herein described significant associations between DoT-P4-levels and DoT-FSH-levels confirm the theory that enhanced FSH stimulation is the primary source of progesterone elevation on the day of final oocyte maturation in stimulated IVF/ICSI cycles. Given the pathophysiologic mechanism of progesterone elevation during ovarian stimulation, the use of an increased FSH step-down dosage should be studied in future RCTs, despite the fact that a step-down approach of daily 12.5 IU rec-FSH did not achieve a significantly reduced progesterone level on the DoT. Clinical Trial Registration: clinicaltrials.gov, identifier NCT03356964.
Collapse
Affiliation(s)
- Barbara Lawrenz
- In-Vitro-Fertilisation (IVF) Department, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
- Obstetrical Department, Women’s University Hospital Tuebingen, Tuebingen, Germany
- *Correspondence: Barbara Lawrenz,
| | - Carol Coughlan
- In-Vitro-Fertilisation (IVF) Department, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - Laura Melado
- In-Vitro-Fertilisation (IVF) Department, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - Shieryl Digma
- In-Vitro-Fertilisation (IVF) Department, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - Junard Sibal
- Clinical Laboratory, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - Alliza Jean
- Clinical Laboratory, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - Human M. Fatemi
- In-Vitro-Fertilisation (IVF) Department, ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| |
Collapse
|
14
|
Zhang C, Wu F, Wu Z, Sun B, Chen C, Qian W. Early Follicular Phase Human Chorionic Gonadotropin Addition May Improve the Outcomes of In Vitro Fertilization/Intracytoplasmic Sperm Injection in Patients With "Unpredictable" Poor Response to Gonadotropin-Releasing Hormone Antagonist Protocol. Front Endocrinol (Lausanne) 2021; 12:739773. [PMID: 34707571 PMCID: PMC8544820 DOI: 10.3389/fendo.2021.739773] [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: 07/11/2021] [Accepted: 09/03/2021] [Indexed: 12/02/2022] Open
Abstract
PURPOSE To compare the effects of early and mid-late follicular phase administration of 150 IU of human chorionic gonadotropin (hCG) on gonadotropin-releasing hormone (GnRH) antagonist protocol in "unpredictable" poor ovarian response (POR) women undergoing in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment. METHODS A retrospective single-center cohort study was conducted on 67 patients with "unpredictable" POR in their first IVF/ICSI cycle receiving GnRH antagonist protocol. Patients were treated with a second IVF/ICSI cycle using the same GnRH antagonist protocol with the same starting dose of recombinant follicle-stimulating hormone (rFSH) as the first cycle; a daily dose of 150 IU of hCG was administrated on either stimulation day 1 (Group A, n = 35) or day 6 (Group B, n = 32). The number of oocytes retrieved, number of usable embryos, serum level of estradiol (E2) on day of hCG trigger, and clinical pregnant outcomes were studied. RESULTS The addition of 150 IU of hCG on either the first day or sixth day of stimulation increases the serum level of E2, luteinizing hormone (LH), and hCG on the day of hCG trigger. Only the use of 150 IU of hCG on the first stimulation day improved the number of oocytes retrieved, mature of oocytes, and usable embryos, but not the addition of hCG on stimulation day 6. Implantation rate, clinical pregnancy rate, and ongoing pregnancy rate showed an increasing trend in patients receiving 150 IU of hCG in the early phase compared with mid-late phase, even thought there was no statistically significant difference. CONCLUSIONS Our study demonstrated that adding 150 IU of hCG in subsequent GnRH antagonist cycle in "unpredictable" poor responders is associated with the improvement of response to stimulation. Furthermore, early follicular phase addition of 150 IU of hCG significantly increased the number of oocytes retrieved and usable embryos than did the mid-late addition of the same dose.
Collapse
|
15
|
Friis Wang N, Skouby SO, Humaidan P, Andersen CY. Response to ovulation trigger is correlated to late follicular phase progesterone levels: A hypothesis explaining reduced reproductive outcomes caused by increased late follicular progesterone rise. Hum Reprod 2020; 34:942-948. [PMID: 30927415 DOI: 10.1093/humrep/dez023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/23/2019] [Accepted: 02/11/2019] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there an association between progesterone (P4) levels on the day of hCG or GnRH trigger and on the day of oocyte retrieval in IVF/ICSI cycles? SUMMARY ANSWER A significant positive correlation between P4 levels on the day of trigger and the day of oocyte retrieval is seen; HCG trigger induces a steeper P4 increase than GnRHa trigger. WHAT IS KNOWN ALREADY FSH induces LH receptor (LHR) expression on granulosa cells, and LHR produces progesterone when exposed to LH-like activity. FSH per se also to some extent induces P4 secretion. Late follicular phase progesterone rise has been associated with reduced reproductive outcomes. STUDY DESIGN, SIZE, DURATION This study is based on data from a previously published RCT conducted from 2009 to 2011. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 384 participants were enrolled; 199 received 5000 IU hCG and 185 received buserelin 0.5 mg for triggering ovulation. P4 was measured on the day of ovulation induction and on the day of oocyte retrieval. FSH consumption and number of retrieved follicles were recorded. MAIN RESULTS AND THE ROLE OF CHANCE A significant linear relationship between P4 on the day of ovulation induction and oocyte retrieval was seen in the hCG trigger group (P < 0.00001) as well as in the GnRHa trigger group (P < 0.00001). The P4 ratio (the increase in P4 between ovulation induction and oocyte retrieval) was significantly higher in the group of patients with <5 follicles compared to those with 5-15 and >15 follicles (P < 0.0001). The FSH consumption per follicle was significantly higher in the group of patients with <5 follicles compared to those with 5-15 and >15 follicles (P < 0.0001). LIMITATIONS, REASONS FOR CAUTION Although the study demonstrates a significant correlation between P4 levels before and after ovulation trigger, it does not demonstrate a causal relation to the number of LHRs present on granulosa cells. WIDER IMPLICATIONS OF THE FINDINGS The findings of this study support the proposed hypothesis that follicles exposed to high levels of FSH during ovarian stimulation will respond with an inappropriately high LHR expression. This in turn causes a high P4 output in response to the trigger. This study further expands our understanding of the underlying mechanisms affecting reproductive outcomes in relation to ovarian stimulation. STUDY FUNDING/COMPETING INTEREST(S) The authors received no specific funding for this work and disclose no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- N Friis Wang
- Laboratory of Reproductive Biology, Section 5712, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medicine, University of Copenhagen, Rigshospitalet, Blegdamsvej 9, Copenhagen N, Denmark
| | - S O Skouby
- Reproductive Medicine Unit, Herlev-Gentofte Hospital, Herlev Ringvej 75, Herlev, Denmark
| | - P Humaidan
- Fertility Clinic, Skive Regional Hospital, Resenvej 25, Skive, Denmark.,Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, Aarhus N, Denmark
| | - C Y Andersen
- Laboratory of Reproductive Biology, Section 5712, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medicine, University of Copenhagen, Rigshospitalet, Blegdamsvej 9, Copenhagen N, Denmark
| |
Collapse
|
16
|
Løssl K, Freiesleben NLC, Wissing ML, Birch Petersen K, Holt MD, Mamsen LS, Anderson RA, Andersen CY. Biological and Clinical Rationale for Androgen Priming in Ovarian Stimulation. Front Endocrinol (Lausanne) 2020; 11:627. [PMID: 33013703 PMCID: PMC7498541 DOI: 10.3389/fendo.2020.00627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/31/2020] [Indexed: 12/24/2022] Open
Abstract
Androgen receptors are expressed by all stages of growing follicles, and follicular fluid androgen levels are positively correlated to granulosa cell androgen receptor and follicle-stimulating hormone (FSH) receptor expression. Thus, androgens may promote follicular growth, accumulation and/or responsiveness to gonadotropins. This is explored therapeutically in the concept of androgen priming, to improve the ovarian response to stimulation in assisted reproduction. Androgen effects may be achieved in two different ways, either directly by providing exogenous androgen or by providing luteinizing hormone (LH) activity [i.e., LH or human chorionic gonadotropin (hCG)] to stimulate local ovarian production of androgen. The androgen concentrations in follicular fluid by far exceed the levels in female circulation and it has recently been shown that there was no correlation between serum testosterone levels and follicular fluid androgen levels. There is some evidence that administration of exogenous dehydroepiandrosterone or testosterone increases live birth rates, but an optimal protocol has not been established and such adjuvant treatment should be considered experimental. Furthermore, studies exploring long-term administration of LH activity, achieving LH levels comparable to those seen in women with polycystic ovary syndrome, are awaited. The aim of the present review is to discuss critically the most suitable approach for androgen priming from a biological and clinical standpoint, and to evaluate current approaches and results obtained in clinical trials.
Collapse
Affiliation(s)
- Kristine Løssl
- The Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Claus Yding Andersen
| |
Collapse
|
17
|
Theofanakis C, Athanasiou V, Liokari E, Stavrou S, Sakellariou M, Athanassiou AI, Athanassiou A, Drakakis P, Loutradis D. The impact of HCG in IVF Treatment: Does it depend on age or on protocol? J Gynecol Obstet Hum Reprod 2019; 48:341-345. [PMID: 30794953 DOI: 10.1016/j.jogoh.2019.02.012] [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: 11/13/2018] [Revised: 01/27/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE to evaluate the effect of the addition of low dose human chorionic gonadotropin (hCG) to human menopausal gonadotropin (HMG) throughout the early follicular phase in controlled ovarian stimulation (COS) conducted with two difference regimens. Gonadotropin-releasing hormone (GnRH) antagonist and short GnRH-agonist protocol were applied in two in vitro fertilization (IVF) clinics. METHODS Clinical study conducted during the period 2014-2016 in two IVF clinics in a cohort of 240 women. In the first group 1 (124 women), a GnRH antagonist protocol with HMG and addition of low dose (100IU/day) h CG was applied. The other group 2 consisted of 116 women who underwent a short GnRH- agonist protocol with HMG and addition of low dose (100IU/day) h CG. RESULTS Multiple logistic regression analysis was performed. The group 2 found to be associated with greater number of follicles and oocytes. The pregnancy rates were 12.1% and 26.7% in group 1 and group 2, respectively (p=0.004). For patients over 40 years, the number of follicles and oocytes retrieved were significant higher in group 2.The pregnancy rate in group 2 was higher than in group 1 (21, 6% vs 5%, p=0.017). CONCLUSIONS Advanced age women are likely to achievepregnancy using the GnRH Short than GnRH antagonist, when HMG/hCG is used, while HMG-hCG gonadotropins have the same potentialas Recombinant follicle stimulating hormone (rFSH)-hCG used in GnRH short protocol.
Collapse
Affiliation(s)
- Charalampos Theofanakis
- IVF Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece; Fertility Institute, Athens, Greece.
| | - Vasilios Athanasiou
- IVF Athens Center, Athens, Greece; OB/GYN, Reproductive Endocrinology & Infertility Co-founder & Scientific Director, IVF Athens Center, Greece.
| | | | - Sofoklis Stavrou
- IVF Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece; Fertility Institute, Athens, Greece.
| | | | | | | | - Petros Drakakis
- IVF Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece; Fertility Institute, Athens, Greece.
| | - Dimitris Loutradis
- IVF Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece; Fertility Institute, Athens, Greece.
| |
Collapse
|
18
|
Zhu X, Fu Y. Randomized, Controlled Pilot Study of Low-Dose Human Chorionic Gonadotropin Administration Beginning From the Early Follicular Phase for Women With Polycystic Ovarian Syndrome Undergoing Ovarian Stimulation Using the Progesterone Protocol. Front Endocrinol (Lausanne) 2019; 10:875. [PMID: 31920984 PMCID: PMC6923733 DOI: 10.3389/fendo.2019.00875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/29/2019] [Indexed: 11/13/2022] Open
Abstract
Purpose: To illustrate whether low-dose human chorionic gonadotropin (hCG) administration during the early follicular phase could reduce the number of large preovulatory follicles in women with polycystic ovarian syndrome (PCOS) undergoing ovarian stimulation using the progesterone protocol. Methods: We performed a randomized, controlled pilot trial at a university-affiliated tertiary hospital. A total of 40 infertile women undergoing their first in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) treatment with the freeze-all strategy were included. Human menopausal gonadotropin (hMG) and progesterone soft capsule 100 mg/d were added simultaneously beginning from menstrual cycle day 3 for all participants. Low-dose hCG (200 IU) was injected every 3 days in the study group from the first day of ovarian stimulation until trigger. The primary outcome was the number of large preovulatory follicles. Secondary outcomes included the incidence of ovarian hyperstimulation syndrome (OHSS); the number of oocytes retrieved, mature oocytes, and good-quality embryos; and clinical results after frozen-thawed embryo transfer (FET) cycles. Results: The study group had slightly more large preovulatory follicles than the control group (17.75 ± 10 vs. 13.2 ± 5.34; P > 0.05). None of the participants experienced severe OHSS. There were no statistically significant differences in the number of oocytes retrieved (15.9 ± 8.46 vs. 15.75 ± 6.96), mature oocytes (13.55 ± 6.56 vs. 13.4 ± 6.34), and good-quality embryos (5.5 ± 3.41 vs. 4.9 ± 2.99) between the two groups (P > 0.05). Clinical pregnancy rates (65.52 vs. 41.94%; P = 0.067) and live birth rates (48.28 vs. 35.48%; P = 0.315) per transfer following FET of the study group were higher than those of the control group, but without statistical significance. Conclusions: Administration of low-dose hCG from the early follicular phase for PCOS patients undergoing ovarian stimulation with progesterone protocol may lead to slightly more early preovulatory follicles and marginally, but not significantly, higher clinical pregnancy rates. A continuous trial should be performed to explore the effects of supplementation with different doses of hCG from the start of ovarian stimulation in PCOS patients using the progesterone protocol. Clinical Trial Registration: Chictr.org.cn, identifier: ChiCTR-IOR-15007165.
Collapse
|
19
|
de Ziegler D, Andersen CY, Stanczyk FZ, Ayoubi JM. Endocrine mechanisms and assay issues in premature progesterone elevation in assisted reproductive technology. Fertil Steril 2018; 109:571-576. [PMID: 29653702 DOI: 10.1016/j.fertnstert.2018.02.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 11/27/2022]
Abstract
Progesterone elevation occurring in the late phases of controlled ovarian stimulation (COS) has been reported for over 25 years. Yet doubts remain regarding the mechanisms at play in this phenomenon and its net consequences on assisted reproductive technology outcome, which is known to occur in poor and good assisted reproductive technology responders. The pathophysiology of end-COS progesterone elevation encountered in gonadotropin-suppressed cycles is different from that prevailing at the time of, and just after, ovulation. The different divergence in practical consequences of end-COS progesterone elevation led to review the progesterone assays developed for measuring progesterone in the luteal phase of the menstrual cycle, but commonly used for measuring all forms of progesterone elevation.
Collapse
Affiliation(s)
- Dominique de Ziegler
- Department of Obstetrics Gynecology and Reproductive Medicine, Hopital Foch-Faculté de Médecine Paris Ouest (UVSQ), Suresnes, France.
| | - Clauslaus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Frank Z Stanczyk
- University of Southern California Keck School of Medicine, Los Angeles, California
| | - Jean Marc Ayoubi
- Department of Obstetrics Gynecology and Reproductive Medicine, Hopital Foch-Faculté de Médecine Paris Ouest (UVSQ), Suresnes, France
| |
Collapse
|
20
|
Esteves SC, Khastgir G, Shah J, Murdia K, Gupta SM, Rao DG, Dash S, Ingale K, Patil M, Moideen K, Thakor P, Dewda P. Association Between Progesterone Elevation on the Day of Human Chronic Gonadotropin Trigger and Pregnancy Outcomes After Fresh Embryo Transfer in In Vitro Fertilization/Intracytoplasmic Sperm Injection Cycles. Front Endocrinol (Lausanne) 2018; 9:201. [PMID: 29755412 PMCID: PMC5932157 DOI: 10.3389/fendo.2018.00201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/10/2018] [Indexed: 11/14/2022] Open
Abstract
Progesterone elevation (PE) during the late follicular phase of controlled ovarian stimulation in fresh embryo transfer in vitro fertilization (IVF)/intracytoplasmic sperm injection cycles has been claimed to be associated with decreased pregnancy rates. However, the evidence is not unequivocal, and clinicians still have questions about the clinical validity of measuring P levels during the follicular phase of stimulated cycles. We reviewed the existing literature aimed at answering four relevant clinical questions, namely (i) Is gonadotropin type associated with PE during the follicular phase of stimulated cycles? (ii) Is PE on the day of human chorionic gonadotropin (hCG) associated with negative fresh embryo transfer IVF/intracytoplasmic sperm injection (ICSI) cycles outcomes in all patient subgroups? (iii) Which P thresholds are best to identify patients at risk of implantation failure due to PE in a fresh embryo transfer? and (iv) Should a freeze all policy be adopted in all the cycles with PE on the day of hCG? The existing evidence indicates that late follicular phase progesterone rise in gonadotropin releasing analog cycles is mainly caused by the supraphysiological stimulation of granulosa cells with exogenous follicle-stimulating hormone. Yet, the type of gonadotropin used for stimulation seems to play no significant role on progesterone levels at the end of stimulation. Furthermore, PE is not a universal phenomenon with evidence indicating that its detrimental consequences on pregnancy outcomes do not affect all patient populations equally. Patients with high ovarian response to control ovarian stimulation are more prone to exhibit PE at the late follicular phase. However, in studies showing an overall detrimental effect of PE on pregnancy rates, the adverse effect of PE on endometrial receptivity seems to be offset, at least in part, by the availability of good quality embryo for transfer in women with a high ovarian response. Given the limitations of the currently available assays to measure progesterone at low ranges, caution should be applied to adopt specific cutoff values above which the effect of progesterone rise could be considered detrimental and to recommend "freeze-all" based solely on pre-defined cutoff points.
Collapse
Affiliation(s)
- Sandro C. Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil
- Division of Urology, Department of Surgery, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Jatin Shah
- Mumbai Fertility Center – Kamala Polyclinic & Nursing Home, Mumbai, India
| | | | | | - Durga G. Rao
- Oasis Centre for Reproductive Medicine, Hyderabad, India
| | - Soumyaroop Dash
- Srishti Assisted Fertility & Advanced Laparoscopy Center, Srishti Hospital, Moolakulam, India
| | - Kundan Ingale
- Nirmiti Clinic, Centre for Assisted Reproduction, Chinchwad, India
| | | | | | | | - Pavitra Dewda
- Merck Limited, Mumbai, India
- *Correspondence: Pavitra Dewda,
| |
Collapse
|
21
|
Volovsky M, Healey M, MacLachlan V, Vollenhoven BJ. Should intrauterine human chorionic gonadotropin infusions ever be used prior to embryo transfer? J Assist Reprod Genet 2017; 35:273-278. [PMID: 28948440 DOI: 10.1007/s10815-017-1049-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/13/2017] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The aim of this study was to explore the factors that influence the outcome of intrauterine human chorionic gonadotropin (hCG) infusion at the time of embryo transfer (ET), in particular, the effect of hCG infusions on fresh and frozen embryo transfers (FETs) and whether prior recurrent implantation failure (RIF) impacts upon outcomes. METHOD This was a case-control study based on a standardized database from a multi-site in vitro fertilization clinic. The analysis contains 458 cases and 749 matched controls, with an intervention group of those given intrauterine hCG prior to ET and a control group of patients receiving no hCG infusion. Outcomes were defined as clinical pregnancy and live birth rates. Two analyses were performed. The first separated FETs (cases n = 224, controls n = 325) and fresh ETs (cases n = 234, controls n = 424), with outcomes calculated in each group. The second analysis divided patients into those with RIF (cases n = 149, controls n = 200) and those without (cases n = 309, controls n = 549). RESULTS Results in fresh ETs demonstrated a 5.8% reduction (adjusted odds ratio (AOR) = 0.60, p = 0.041) in clinical pregnancy rates with the use of intrauterine hCG. In those without defined RIF, clinical pregnancy rates were reduced by 8.1% (AOR = 0.61, p = 0.023) and live birth rates by 7.2% (AOR = 0.56, p = 0.32) with intrauterine hCG use. There were no significant differences in outcomes in FETs and in the RIF cohort. CONCLUSION Intrauterine hCG at the time of ET not only seems to have no benefit, but rather a negative effect in fresh ETs and those without RIF.
Collapse
Affiliation(s)
- Michelle Volovsky
- Royal Melbourne Hospital, 300 Grattan St, Parkville, VIC, 3050, Australia. .,Monash University, Wellington Rd & Blackburn Rd, Clayton, VIC, 3800, Australia.
| | - Martin Healey
- Royal Women's Hospital, 20 Flemington Rd, Parkville, VIC, 3052, Australia.,University of Melbourne, Parkville, VIC, 3010, Australia.,Monash IVF, Level 1, 21-31 Goodwood St, Richmond, VIC, 3121, Australia
| | - Vivien MacLachlan
- Monash IVF, Level 1, 21-31 Goodwood St, Richmond, VIC, 3121, Australia
| | - Beverley J Vollenhoven
- Monash University, Wellington Rd & Blackburn Rd, Clayton, VIC, 3800, Australia.,Monash IVF, Level 1, 21-31 Goodwood St, Richmond, VIC, 3121, Australia.,Monash Health, 246 Clayton Road, Clayton, VIC, 3168, Australia
| |
Collapse
|
22
|
Alsbjerg B, Elbaek HO, Laursen RJ, Povlsen BB, Haahr T, Yding Andersen C, Humaidan P. Bio-equivalent doses of recombinant HCG and recombinant LH during ovarian stimulation result in similar oestradiol output: a randomized controlled study. Reprod Biomed Online 2017; 35:232-238. [PMID: 28532660 DOI: 10.1016/j.rbmo.2017.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 10/19/2022]
Abstract
In nature, HCG is secreted by the implanting embryo from peri-implantation and onwards. In contrast, LH is mandatory for steroidogenesis and follicular development during the follicular phase, working in synergy with FSH. Moreover, LH is mandatory for the function of the corpus luteum. Although LH and HCG bind to the same receptor, significant molecular, structural and functional differences exist, inducing differences in bioactivity. This randomized controlled study compared the effect of recombinant FSH stimulation combined with daily either micro-dose recombinant HCG or recombinant LH supplementation in a 1:1 bioactivity ratio from day 1 of stimulation in a long gonadotrophin releasing hormone agonist down regulation protocol. A total of 100 patients from a public clinic completed the study. The primary end-point was the oestradiol level on the day of ovulation trigger and the median oestradiol level in the HCG supplemented group was 8662 pmol/l versus 9203 pmol/l in the recombinant LH supplemented group; therefore, no significant difference was found. Moreover, no differences were observed in the number of oocytes retrieved or in the live birth rate. We conclude that recombinant HCG and recombinant LH are equally effective in boosting oestradiol synthesis during ovarian stimulation when used in a 1:1 bioactivity ratio.
Collapse
Affiliation(s)
- Birgit Alsbjerg
- The Fertility Clinic Skive Region Hospital, Resenvej 25, 7870 Skive, Denmark; Faculty of Health, Aarhus Universitet, Nordre Ringgade 1, 8000 Aarhus C, Denmark
| | - Helle Olesen Elbaek
- The Fertility Clinic Skive Region Hospital, Resenvej 25, 7870 Skive, Denmark
| | | | - Betina Boel Povlsen
- The Fertility Clinic Skive Region Hospital, Resenvej 25, 7870 Skive, Denmark
| | - Thor Haahr
- The Fertility Clinic Skive Region Hospital, Resenvej 25, 7870 Skive, Denmark; Faculty of Health, Aarhus Universitet, Nordre Ringgade 1, 8000 Aarhus C, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen East, Denmark
| | - Peter Humaidan
- The Fertility Clinic Skive Region Hospital, Resenvej 25, 7870 Skive, Denmark; Faculty of Health, Aarhus Universitet, Nordre Ringgade 1, 8000 Aarhus C, Denmark.
| |
Collapse
|
23
|
Outcomes and Recommendations of an Indian Expert Panel for Improved Practice in Controlled Ovarian Stimulation for Assisted Reproductive Technology. Int J Reprod Med 2017; 2017:9451235. [PMID: 28246628 PMCID: PMC5299198 DOI: 10.1155/2017/9451235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/08/2016] [Accepted: 12/21/2016] [Indexed: 11/17/2022] Open
Abstract
Purpose. To improve success of in vitro fertilization (IVF), assisted reproductive technology (ART) experts addressed four questions. What is optimum oocytes number leading to highest live birth rate (LBR)? Are cohort size and embryo quality correlated? Does gonadotropin type affect oocyte yield? Should “freeze-all” policy be adopted in cycles with progesterone >1.5 ng/mL on day of human chorionic gonadotropin (hCG) administration? Methods. Electronic database search included ten studies on which panel gave opinions for improving current practice in controlled ovarian stimulation for ART. Results. Strong association existed between retrieved oocytes number (RON) and LBRs. RON impacted likelihood of ovarian hyperstimulation syndrome (OHSS). Embryo euploidy decreased with age, not with cohort size. Progesterone > 1.5 ng/dL did not impair cycle outcomes in patients with high cohorts and showed disparate results on day of hCG administration. Conclusions. Ovarian stimulation should be designed to retrieve 10–15 oocytes/treatment. Accurate dosage, gonadotropin type, should be selected as per prediction markers of ovarian response. Gonadotropin-releasing hormone (GnRH) antagonist based protocols are advised to avoid OHSS. Cumulative pregnancy rate was most relevant pregnancy endpoint in ART. Cycles with serum progesterone ≥1.5 ng/dL on day of hCG administration should not adopt “freeze-all” policy. Further research is needed due to lack of data availability on progesterone threshold or index.
Collapse
|
24
|
Andersen CY, Fischer R, Giorgione V, Kelsey TW. Micro-dose hCG as luteal phase support without exogenous progesterone administration: mathematical modelling of the hCG concentration in circulation and initial clinical experience. J Assist Reprod Genet 2016; 33:1311-1318. [PMID: 27448021 DOI: 10.1007/s10815-016-0764-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 06/22/2016] [Indexed: 12/23/2022] Open
Abstract
For the last two decades, exogenous progesterone administration has been used as luteal phase support (LPS) in connection with controlled ovarian stimulation combined with use of the human chorionic gonadotropin (hCG) trigger for the final maturation of follicles. The introduction of the GnRHa trigger to induce ovulation showed that exogenous progesterone administration without hCG supplementation was insufficient to obtain satisfactory pregnancy rates. This has prompted development of alternative strategies for LPS. Augmenting the local endogenous production of progesterone by the multiple corpora lutea has been one focus with emphasis on one hand to avoid development of ovarian hyper-stimulation syndrome and, on the other hand, to provide adequate levels of progesterone to sustain implantation. The present study evaluates the use of micro-dose hCG for LPS support and examines the potential advances and disadvantages. Based on the pharmacokinetic characteristics of hCG, the mathematical modelling of the concentration profiles of hCG during the luteal phase has been evaluated in connection with several different approaches for hCG administration as LPS. It is suggested that the currently employed LPS provided in connection with the GnRHa trigger (i.e. 1.500 IU) is too strong, and that daily micro-dose hCG administration is likely to provide an optimised LPS with the current available drugs. Initial clinical results with the micro-dose hCG approach are presented.
Collapse
Affiliation(s)
- C Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Laboratory of Reproductive Biology, Section 5712, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, University of Copenhagen, Blegdamsvej 9, Rigshospitalet, 2100, Copenhagen, Denmark.
| | - R Fischer
- MVZ Fertility Center Hamburg GmbH, Hamburg, Germany
| | - V Giorgione
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas W Kelsey
- School of Computer Science, University of St Andrews, St Andrews, Fife, UK
| |
Collapse
|
25
|
Borup R, Thuesen LL, Andersen CY, Nyboe-Andersen A, Ziebe S, Winther O, Grøndahl ML. Competence Classification of Cumulus and Granulosa Cell Transcriptome in Embryos Matched by Morphology and Female Age. PLoS One 2016; 11:e0153562. [PMID: 27128483 PMCID: PMC4851390 DOI: 10.1371/journal.pone.0153562] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/31/2016] [Indexed: 12/23/2022] Open
Abstract
Objective By focussing on differences in the mural granulosa cell (MGC) and cumulus cell (CC) transcriptomes from follicles resulting in competent (live birth) and non-competent (no pregnancy) oocytes the study aims on defining a competence classifier expression profile in the two cellular compartments. Design: A case-control study. Setting: University based facilities for clinical services and research. Patients: MGC and CC samples from 60 women undergoing IVF treatment following the long GnRH-agonist protocol were collected. Samples from 16 oocytes where live birth was achieved and 16 age- and embryo morphology matched incompetent oocytes were included in the study. Methods MGC and CC were isolated immediately after oocyte retrieval. From the 16 competent and non-competent follicles, mRNA was extracted and expression profile generated on the Human Gene 1.0 ST Affymetrix array. Live birth prediction analysis using machine learning algorithms (support vector machines) with performance estimation by leave-one-out cross validation and independent validation on an external data set. Results We defined a signature of 30 genes expressed in CC predictive of live birth. This live birth prediction model had an accuracy of 81%, a sensitivity of 0.83, a specificity of 0.80, a positive predictive value of 0.77, and a negative predictive value of 0.86. Receiver operating characteristic analysis found an area under the curve of 0.86, significantly greater than random chance. When applied on 3 external data sets with the end-point outcome measure of blastocyst formation, the signature resulted in 62%, 75% and 88% accuracy, respectively. The genes in the classifier are primarily connected to apoptosis and involvement in formation of extracellular matrix. We were not able to define a robust MGC classifier signature that could classify live birth with accuracy above random chance level. Conclusion We have developed a cumulus cell classifier, which showed a promising performance on external data. This suggests that the gene signature at least partly include genes that relates to competence in the developing blastocyst.
Collapse
Affiliation(s)
- Rehannah Borup
- Center for Genomic Medicine, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
- * E-mail:
| | - Lea Langhoff Thuesen
- Fertility Clinic, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Anders Nyboe-Andersen
- Fertility Clinic, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Søren Ziebe
- Fertility Clinic, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Ole Winther
- Bioinformatics Center, Department of Biology and Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Marie Louise Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev Hospital, Copenhagen, Denmark
| |
Collapse
|
26
|
Major drawbacks and additional benefits of agonist trigger—not ovarian hyperstimulation syndrome related. Fertil Steril 2015; 103:874-8. [DOI: 10.1016/j.fertnstert.2015.01.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 11/18/2022]
|
27
|
Polyzos NP, Corona R, Van De Vijver A, Blockeel C, Drakopoulos P, Vloeberghs V, De Vos M, Camus M, Humaidan P, Tournaye H. Corifollitropin alfa followed by hpHMG in GnRH agonist protocols. Two prospective feasibility studies in poor ovarian responders. Gynecol Endocrinol 2015; 31:885-90. [PMID: 26172925 DOI: 10.3109/09513590.2015.1065481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In two prospective uncontrolled feasibility trials, we examined the effect of corifollitropin alfa (CFA) followed by highly purified human menopausal gonadotrophin (hpHMG) in a short flare-up gonadotropin-releasing hormone (GnRH) agonist and a long GnRH agonist protocol for women with poor ovarian response. Overall, 45 patients were treated with short flare-up and 47 patients with the long agonist protocol. All patients received a single dose of 150 μg CFA, followed by 300 IU hpHMG 7 days later, triggering with 10 000 IU hCG, CSI and day 3 embryo transfer. Ongoing pregnancy rates (OPRs) did not differ between the short 15.6% and the long 17% agonist protocol (p = 0.85). Among patients treated with the short flare-up protocol, OPRs were 20% for younger patients (<40 years old) and 12% in older women (≥40 years old), p = 0.68. Similarly, in patients treated with the long agonist protocol younger women had an OPR of 26.7% versus 12.5% in older women, p = 0.23. Among patients treated with the short flare-up, live births rate were 15% and 4.3% for younger (<40 years old) and older patients (≥40 years old), respectively, p = 0.32. Similarly, in patients treated with the long agonist protocol, live births rate were 25% and 12.9% for younger (<40 years old) and older patients (≥40 years old), respectively, p = 0.41. None of the patients reported any serious adverse event related to treatment. According to our results, CFA followed by hpHMG in a short flare-up or long GnRH agonist protocol appears to be a feasible option for poor ovarian responders. Large phase III trials are mandatory prior to introduction in clinical practice.
Collapse
Affiliation(s)
- Nikolaos P Polyzos
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Roberta Corona
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Arne Van De Vijver
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Christophe Blockeel
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Panagiotis Drakopoulos
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Veerle Vloeberghs
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Michel De Vos
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Michel Camus
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| | - Peter Humaidan
- b The Fertility Clinic, Skive Regional Hospital , Skive , Denmark , and
- c Faculty of Health , Aarhus University , Aarhus , Denmark
| | - Herman Tournaye
- a Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels , Belgium
| |
Collapse
|
28
|
Choi J, Smitz J. Luteinizing hormone and human chorionic gonadotropin: a review of their varied clinical applications in assisted reproductive technology. Expert Rev Endocrinol Metab 2015; 10:87-100. [PMID: 30289044 DOI: 10.1586/17446651.2015.969711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are often viewed as interchangeable from a functional standpoint because they are highly homologous members of the same glycoprotein hormone family that share a common α-subunit and receptor. However, technological advances yielding highly purified and recombinant gonadotropin preparations have revealed that LH and hCG fulfill different roles, both endogenously and when administered exogenously. These differences are becoming more apparent as the individual hormones are incorporated into the treatment of infertility - a therapeutic area that is continually advancing with the introduction of new agents and emerging clinical trial data. This review examines the unique attributes of LH and hCG that drive their distinctive applications in the treatment of female infertility.
Collapse
Affiliation(s)
- Janet Choi
- a 1 The Center for Women's Reproductive Care at Columbia University, 1790 Broadway, 2nd Floor, New York, NY 10019, USA
| | - Johan Smitz
- b 2 Laboratory Hormonology and Tumormarkers, UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
29
|
Andersen CY, Ezcurra D. Human steroidogenesis: implications for controlled ovarian stimulation with exogenous gonadotropins. Reprod Biol Endocrinol 2014; 12:128. [PMID: 25543693 PMCID: PMC4396073 DOI: 10.1186/1477-7827-12-128] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/12/2014] [Indexed: 11/10/2022] Open
Abstract
In the menstrual cycle, the mid-cycle surge of gonadotropins (both luteinising hormone [LH] and follicle-stimulating hormone [FSH]) signals the initiation of the periovulatory interval, during which the follicle augments progesterone production and begins to luteinise, ultimately leading to the rupture of the follicle wall and the release of an oocyte. The administration of gonadotropins in controlled ovarian stimulation (COS) leads to supraphysiological steroid concentrations of a very different profile compared with those seen during natural cycles. It has been suggested that these high steroid concentrations cause alterations in endometrial development, affecting oocyte viability in assisted reproductive technology. Furthermore, it has been proposed that elevated progesterone levels have a negative effect on the reproductive outcome of COS. This may arise from an asynchrony between embryo stage and endometrium status at the window of implantation. The regulation of progesterone production by the developing follicles during COS is a complicated interplay of hormonal systems involving the theca and granulosa cells, and the effect of the actions of both LH and FSH. The present paper reviews current knowledge of the regulation of progesterone in the human ovary during the follicular phase and highlights areas where knowledge remains limited. In this review, we provide in-depth information outlining the regulation and function of gonadotropins in the complicated area of steroidogenesis. Based on current evidence, it is not clear whether the high levels of progesterone produced during COS have detrimental effects on fertility.
Collapse
Affiliation(s)
- Claus Y Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark.
| | - Diego Ezcurra
- Global Medical Affairs, EMD/Merck Serono, One Technology Place, Rockland, MA, 02370, USA.
| |
Collapse
|
30
|
Ezcurra D, Humaidan P. A review of luteinising hormone and human chorionic gonadotropin when used in assisted reproductive technology. Reprod Biol Endocrinol 2014; 12:95. [PMID: 25280580 PMCID: PMC4287577 DOI: 10.1186/1477-7827-12-95] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/20/2014] [Indexed: 12/02/2022] Open
Abstract
Gonadotropins extracted from the urine of post-menopausal women have traditionally been used to stimulate folliculogenesis in the treatment of infertility and in assisted reproductive technology (ART). Products, such as human menopausal gonadotropin (hMG), consist not only of a mixture of the hormones, follicle-stimulating hormone (FSH), luteinising hormone (LH) and human chorionic gonadotropin (hCG), but also other biologically active contaminants, such as growth factors, binding proteins and prion proteins. The actual amount of molecular LH in hMG preparations varies considerably due to the purification process, thus hCG, mimicking LH action, is added to standardise the product. However, unlike LH, hCG plays a different role during the natural human menstrual cycle. It is secreted by the embryo and placenta, and its main role is to support implantation and pregnancy. More recently, recombinant gonadotropins (r-hFSH and r-hLH) have become available for ART therapies. Recombinant LH contains only LH molecules. In the field of reproduction there has been controversy in recent years over whether r-hLH or hCG should be used for ART. This review examines the existing evidence for molecular and functional differences between LH and hCG and assesses the clinical implications of hCG-supplemented urinary therapy compared with recombinant therapies used for ART.
Collapse
Affiliation(s)
- Diego Ezcurra
- EMD/Merck Serono, One Technology Place, Rockland, MA 02370 USA
| | - Peter Humaidan
- Skive Regional Hospital and Faculty of Health, Aarhus University and Odense University, Resenvej 25, Skive, 7800 Denmark
| |
Collapse
|
31
|
Yding Andersen C, Vilbour Andersen K. Improving the luteal phase after ovarian stimulation: reviewing new options. Reprod Biomed Online 2014; 28:552-9. [DOI: 10.1016/j.rbmo.2014.01.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 11/30/2022]
|
32
|
Thuesen LL, Andersen AN, Loft A, Smitz J. Intrafollicular endocrine milieu after addition of hCG to recombinant FSH during controlled ovarian stimulation for in vitro fertilization. J Clin Endocrinol Metab 2014; 99:517-26. [PMID: 24297796 DOI: 10.1210/jc.2013-1528] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT The role of human chorionic gonadotropin (hCG) supplementation on the intrafollicular steroid milieu has been studied. OBJECTIVE The objective of the study was to assess the impact on steroid levels in follicular fluids (FFs) after different doses of hCG supplementation to recombinant FSH for controlled ovarian stimulation. SETTING This was a prospective randomized dose-response study conducted at Copenhagen University Hospital, Rigshospitalet, Denmark. PATIENTS From 62 in vitro fertilization patients, 334 FFs were selected for analyses. INTERVENTIONS Patients were treated using a GnRH agonist protocol with recombinant FSH 150 IU/d and randomized from stimulation day 1 to supplementation with hCG: D0, 0 IU/d; D50, 50 IU/d; D100, 100 IU/d; and D150, 150 IU/d. MAIN OUTCOME MEASURE Intrafollicular hormone concentrations in relation to treatment groups, follicular sizes, and embryo quality were measured. RESULTS In large follicles, hCG supplementation induced a nearly 3-fold increase of estradiol (nanomoles per liter) [D0: 1496; D50: 3138; D100: 4338; D150: 4009 (P < .001)], a significant 3-fold increase of androstenedione, and a 5-fold increase of T (nanomoles per liter) [D0: 15; D50: 38; D100: 72; D150: 56 (P < .001)]. The estradiol to T ratio decreased significantly, with the lowest ratio in D100 and the highest in D0. Large follicles giving rise to good-quality embryos had significantly higher estradiol and progesterone levels and estradiol to T, estradiol to androstenedione, and progesterone to estradiol ratios, compared with small follicles, leading to poor-quality embryos. CONCLUSIONS Increasing doses of hCG supplementation markedly stimulated the intrafollicular concentration of both estradiol and androgens, with a shift toward a more androgenic milieu. In large follicles with oocytes giving rise to good-quality embryos, the FFs were significantly more estrogenic than in small follicles with oocytes developing into poor quality embryos.
Collapse
Affiliation(s)
- L L Thuesen
- The Fertility Clinic (L.L.T., A.N.A., A.L.), Section 4071, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark; and Follicle Biology Laboratory (J.S.), Vrije Universiteit Brussel, UZ Brussel, 1090 Brussels, Belgium
| | | | | | | |
Collapse
|
33
|
Griesinger G, Mannaerts B, Andersen CY, Witjes H, Kolibianakis EM, Gordon K. Progesterone elevation does not compromise pregnancy rates in high responders: a pooled analysis of in vitro fertilization patients treated with recombinant follicle-stimulating hormone/gonadotropin-releasing hormone antagonist in six trials. Fertil Steril 2013; 100:1622-8.e1-3. [PMID: 24083873 DOI: 10.1016/j.fertnstert.2013.08.045] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/02/2013] [Accepted: 08/27/2013] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To compare the impact of elevated P during the late follicular phase on the chance of pregnancy in low, normal, and high responders. DESIGN Retrospective combined analysis from six clinical trials. SETTING IVF centers. PATIENT(S) Women up to 39 years of age with a regular menstrual cycle and an indication for ovarian stimulation before IVF/intracytoplasmic sperm injection. INTERVENTION(S) Ovarian stimulation with recombinant (r) FSH in a GnRH antagonist protocol. MAIN OUTCOME MEASURE(S) Ongoing pregnancy rates (OPRs) assessed with the use of univariate and multivariate analyses according to serum P levels ≤ 1.5 ng/mL versus >1.5 ng/mL on the day of hCG administration and compared among low (1-5 oocytes), normal (6-18 oocytes), and high (>18 oocytes) responders. RESULT(S) A total of 157/1,866 women (8.4%; 95% confidence interval [CI] 7.2%-9.8%) had elevated P. Incidence of elevated P increased from 4.5% in low responders to 19.0% in high responders. Overall, OPRs were significantly lower in women with elevated P. Per started cycle, the >1.5 to ≤ 1.5 ng/mL adjusted odds ratio was 0.55 (95% CI 0.37-0.81). OPRs were not impaired in high responders with P elevation and were significantly higher compared with normal responders with P elevation. CONCLUSION(S) The incidence of elevated P increases with ovarian response, and elevated P at a threshold of 1.5 ng/mL is independently associated with a decreased chance of pregnancy in low to normal responders, but not in high responders, when using an rFSH/GnRH antagonist protocol.
Collapse
Affiliation(s)
- Georg Griesinger
- Department of Reproductive Medicine and Gynecologic Endocrinology, University Clinic of Schleswig-Holstein, Luebeck, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
Thuesen LL, Smitz J, Loft A, Nyboe Andersen A. Endocrine effects of hCG supplementation to recombinant FSH throughout controlled ovarian stimulation for IVF: a dose-response study. Clin Endocrinol (Oxf) 2013; 79:708-15. [PMID: 23448396 DOI: 10.1111/cen.12186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 02/13/2013] [Accepted: 02/19/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To analyse the endocrine response in relation to the Δ-4 and Δ-5 pathways of ovarian steroidogenesis after different doses of human chorionic gonadotrophin (hCG) supplementation to recombinant FSH from Day 1 of controlled ovarian stimulation for IVF. DESIGN A randomized dose-response pilot study. PATIENTS A total of 62 IVF patients aged 25-37 years with regular cycles and FSH <12 IU/l were treated with a fixed dose of rFSH 150 IU/day and randomized to four hCG dose groups: Dose 0: 0 IU/day, Dose 50: 50 IU/day, Dose 100: 100 IU/day and Dose 150: 150 IU/day. RESULTS A significant hCG dose-dependent incremental increase was found for progesterone (49-160%), 17-OH-progesterone (223-614%), androstenedione (91-340%) and testosterone (95-338%) from Dose 0 to Dose 150, respectively. Dehydroepiandrosterone (DHEA) showed minor changes during stimulation and no differences between the groups. The highest oestradiol concentrations were observed in Dose 100 and Dose 150. Sex hormone-binding globulin (SHBG) increased similarly in all groups at the end of stimulation. No difference was observed for anti-müllerian hormone (AMH) concentration between the groups, but a 50% decline from the start to the end of the stimulation was found. CONCLUSION Supplementation with hCG resulted in a clear dose-related response for androgens, progesterone and 17-OH-progesterone. Oestradiol concentration reached maximum levels with an hCG dose of 100 IU/day, suggesting saturation of aromatase function. No difference between the groups was observed for DHEA, supporting that the stimulatory effects of hCG doses on androgens and oestrogen production were mainly induced via the Δ-5 pathway. SHBG, being a biomarker of oestrogen/androgen balance, was not changed by increasing hCG.
Collapse
Affiliation(s)
- L L Thuesen
- The Fertility Clinic, Section 4071, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | |
Collapse
|
35
|
Goeckenjan M, Rösner S, Toth B, Strowitzki T, Germeyer A. Successful controlled ovarian stimulation despite elevated hCG levels after first-trimester abortion in the context of fertility preservation. Gynecol Endocrinol 2013; 29:960-2. [PMID: 23952105 DOI: 10.3109/09513590.2013.824961] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Fertility preservation prior to gonadotoxic chemotherapy by cryopreservation of the ovarian tissue and controlled ovarian stimulation can be effective immediately after induced abortion in the first trimenon. In a reproductive endocrinology and infertility unit of a tertiary care university-based medical centre (University Hospital of Heidelberg) a 37-year-old women with breast cancer was counseled for fertility preservation. Cryopreservation of ovarian tissue, followed by ovarian stimulation for planned intracytoplasmatic sperm injection (ICSI), transvaginal oocyte aspiration and cryopreservation of fertilized eggs was performed in spite of persistently elevated human chorionic gonadotropin (hCG)-levels after induced abortion. Twenty-four fertilized oocytes with a fertilization rate of 92% were cryopreserved. Ovarian stimulation and oocyte cryopreservation can be successfully performed with good results immediately after miscarriage, despite persistent high hCG-levels.
Collapse
Affiliation(s)
- M Goeckenjan
- Department of Gynaecological Endocrinology and Reproductive Medicine, University Hospital of Heidelberg , Germany and
| | | | | | | | | |
Collapse
|
36
|
|
37
|
König TE, van der Houwen LEE, Overbeek A, Hendriks ML, Beutler-Beemsterboer SN, Kuchenbecker WKH, Renckens CNM, Bernardus RE, Schats R, Homburg R, Hompes PGA, Lambalk CB. Recombinant LH supplementation to a standard GnRH antagonist protocol in women of 35 years or older undergoing IVF/ICSI: a randomized controlled multicentre study. Hum Reprod 2013; 28:2804-12. [PMID: 23838159 DOI: 10.1093/humrep/det266] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Does the addition of exogenous LH to an IVF/ICSI stimulation protocol with recombinant FSH (r-FSH) and a GnRH antagonist improve the ovarian response and pregnancy rates in women of 35 years and older? SUMMARY ANSWER Supplementation of LH during the second half of the follicular phase has no effect on pregnancy rates, implantation rates or on ovarian response in women of 35 years and older undergoing GnRH antagonist IVF/ICSI cycles. WHAT IS KNOWN ALREADY In IVF/ICSI stimulation protocols GnRH agonists or antagonists are administered to prevent a premature pituitary LH surge, which can have a detrimental effect on the IVF/ICSI procedure. In effect, GnRH analogues cause the levels of both gonadotrophins to drop. In order to allow follicle growth FSH is administered exogenously, whereas LH is usually not supplemented. Although GnRH analogues prevent LH surges, there is evidence that, particularly in older women, administration of GnRH analogues may cause endogenous LH levels to decrease excessively. Several studies have been performed to investigate whether the addition of recombinant LH (r-LH) to r-FSH improves cycle outcome. Only a few studies have analysed this issue in the GnRH antagonist protocol and the results of these trials obtained in older women (>35 years old) are conflicting. STUDY DESIGN, SIZE, DURATION A multicentre RCT was performed between 2004 and 2010 in 253 couples who were undergoing IVF or ICSI. Women were 35 years or older and received ovarian stimulation in a protocol with r-FSH (Gonal-F 225 IU/day) starting from cycle day 3 and GnRH antagonist (Cetrotide 0.25 mg/day) from stimulation day 6. Randomization took place on stimulation day 6 to receive both r-FSH and r-LH (Luveris 150 IU/day) or continue with FSH alone. Randomization for r-LH supplementation was performed centrally by serially numbered, opaque, sealed envelopes, stratified by centre. PARTICIPANTS/MATERIALS, SETTING, METHODS Of 253 subjects randomized, 125 received both r-FSH and r-LH and 128 received r-FSH only. Patients were recruited from the Division of Reproductive Medicine of the Obstetrics and Gynaecology department of four hospitals in the Netherlands. MAIN RESULTS AND THE ROLE OF CHANCE There were no demographic or clinical differences between the groups. The intention-to-treat analysis revealed that of those receiving both r-FSH and r-LH, 35 (28.0%) had a clinical pregnancy, compared with 38 (29.7%) receiving only r-FSH (mean difference -1.5%; 95% confidence interval (CI) -9.4 to 12.7, P = 0.9). Ongoing pregnancy rates were 25 (20%) versus 28 (21.9%) (mean difference -1.9%; 95% CI -8.2 to 11.9, P = 0.9) and implantation rates 18.8 versus 20.7% (mean difference -1.9%; 95% CI -8.0 to 11.7, P = 0.6) in the 'r-FSH and r-LH' and 'r-FSH only' groups respectively. LIMITATIONS, REASONS FOR CAUTION A limitation of our study is its early closure. This was done because the interim analysis after randomization of 250 patients indicated no benefit in any aspect of the experiment. WIDER IMPLICATIONS OF THE FINDINGS Given previous data, including a Cochrane review, and our own results the evidence indicates that LH supplementation has no benefit on ongoing pregnancy rates in women of 35 years or older. STUDY FUNDING/COMPETING INTEREST(S) Merck Serono Netherlands, an affiliate of Merck Serono SA- Geneva, an affiliate of Merck KGaA, Darmstadt, Germany has donated the r-LH (Luveris(®)). No conflict of interest to declare. TRIAL REGISTRATION NUMBER The trial was registered in the Dutch trial register (ISRCTN10841210).
Collapse
Affiliation(s)
- T E König
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, VU University Medical Center (VUmc), PO Box 7057, Amsterdam 1007 MB, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Martins WP, Vieira ADD, Figueiredo JBP, Nastri CO. FSH replaced by low-dose hCG in the late follicular phase versus continued FSH for assisted reproductive techniques. Cochrane Database Syst Rev 2013; 2013:CD010042. [PMID: 23543584 PMCID: PMC11558307 DOI: 10.1002/14651858.cd010042.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND During controlled ovarian hyperstimulation (COH) follicle-stimulating hormone (FSH) is frequently used for several days to achieve follicular development. FSH is a relatively expensive drug, substantially contributing to the total expenses of assisted reproductive techniques (ART). When follicles achieve a diameter greater than 10 mm they start expressing luteinising hormone (LH) receptors. At this point, FSH might be replaced by low-dose human chorionic gonadotropin (hCG), which is less expensive. In addition to cost reduction, replacing FSH by low-dose hCG has a theoretical potential to reduce the incidence of ovarian hyperstimulation syndrome (OHSS). OBJECTIVES To evaluate the effectiveness and safety of using low-dose hCG to replace FSH during the late follicular phase in women undergoing COH for assisted reproduction, compared to the use of a conventional COH protocol. SEARCH METHODS We searched for randomised controlled trials (RCT) in electronic databases (Menstrual Disorders and Subfertility Group Specialized Register, CENTRAL, MEDLINE, EMBASE, PsycINFO, CINAHL, LILACS), trials registers (ClinicalTrials.gov, Current Controlled Trials, World Health Organization International Clinical Trials Registry Platform), conference abstracts (ISI Web of knowledge), and grey literature (OpenGrey); additionally we handsearched the reference list of included studies and similar reviews. The last electronic search was performed in February 2013.. SELECTION CRITERIA Only true RCTs comparing the replacement of FSH by low-dose hCG during late follicular phase of COH were considered eligible; quasi or pseudo-randomised trials were not included. Cross-over trials would be included only if data regarding the first treatment of each participant were available; trials that included the same participant more than once would be included only if each participant was always allocated to the same intervention and follow-up periods were the same in both/all arms, or if data regarding the first treatment of each participant were available. We excluded trials that sustained FSH after starting low-dose hCG and those that started FSH and low-dose hCG at the same time. DATA COLLECTION AND ANALYSIS Study eligibility, data extraction, and assessment of the risk of bias were performed independently by two review authors, and disagreements were solved by consulting a third review author. We corresponded with study investigators in order to solve any query, as required. The overall quality of the evidence was assessed in a GRADE summary of findings table. MAIN RESULTS The search retrieved 1585 records; from those five studies were eligible, including 351 women (intervention = 166; control = 185). All studies were judged to be at high risk of bias. All reported per-woman rather than per-cycle data.When use of low-dose hCG to replace FSH was compared with conventional COH for the outcome of live birth, confidence intervals were very wide and findings were compatible with appreciable benefit, no effect or appreciable harm for the intervention (RR 1.56, 95% CI 0.75 to 3.25, 2 studies, 130 women, I² = 0%, very-low-quality evidence). This suggests that for women with a 14% chance of achieving live birth using conventional COH, the chance of achieving live birth using low-dose hCG would be between 10% and 45%.Similarly confidence intervals were very wide for the outcome of OHSS and findings were compatible with benefit, no effect or harm for the intervention (OR 0.30, 95% CI 0.06 to 1.59, 5 studies, 351 women, I² = 59%, very-low-quality evidence). This suggests that for women with a 3% risk of OHSS using conventional COH, the risk using low-dose hCG would be between 0% and 4%.The confidence intervals were wide for the outcome of ongoing pregnancy and findings were compatible with benefit or no effect for the intervention (RR 1.14, 95% CI 0.81 to 1.60, 3 studies, 252 women, I² = 0%, low-quality evidence). This suggests that for women with a 32% chance of achieving ongoing pregnancy using conventional COH, the chance using low-dose hCG would be between 27% and 53%.The confidence intervals were wide for the outcome of clinical pregnancy and findings were compatible with benefit or no effect for the intervention (RR 1.19, 95% CI 0.92 to 1.55, 5 studies, 351 women, I² = 0%, low-quality evidence). This suggests that for women with a 35% chance of achieving clinical pregnancy using conventional COH, the chance using low-dose hCG would be between 32% and 54%.The confidence intervals were very wide for the outcome of miscarriage and findings were compatible with benefit, no effect or harm for the intervention (RR 1.08, 95% CI 0.50 to 2.31, 3 studies, 127 pregnant women, I² = 0%, very-low-quality evidence). This suggests that for pregnant women with a 16% risk of miscarriage using conventional COH, the risk using low-dose hCG would be between 8% and 36%.The findings for the outcome of FSH consumption were compatible with benefit for the intervention (MD -639 IU, 95% CI -893 to -385, 5 studies, 333 women, I² = 88%, moderate-quality evidence).The findings for the outcome of number of oocytes retrieved were compatible with no effect for the intervention (MD -0.12 oocytes, 95% CI -1.0 to 0.8 oocytes, 5 studies, 351 women, I² = 0%, moderate-quality evidence). AUTHORS' CONCLUSIONS We are very uncertain of the effect on live birth, OHSS and miscarriage of using low-dose hCG to replace FSH during the late follicular phase of COH in women undergoing ART, compared to the use of conventional COH. The current evidence suggests that this intervention does not reduce the chance of ongoing and clinical pregnancy; and that it is likely to result in an equivalent number of oocytes retrieved expending less FSH. More studies are needed to strengthen the evidence regarding the effect of this intervention on important reproductive outcomes.
Collapse
Affiliation(s)
- Wellington P Martins
- University of Sao PauloDepartment of Obstetrics and Gynecology, Medical School of Ribeirao PretoHospital das Clinicas da FMRP‐USP, 8 andarCampus Universitario da USPRibeirao PretoSao PauloBrazil14048‐900
| | - Andrea DD Vieira
- University of Sao PauloDepartment of Obstetrics and Gynecology, Medical School of Ribeirao PretoHospital das Clinicas da FMRP‐USP, 8 andarCampus Universitario da USPRibeirao PretoSao PauloBrazil14048‐900
| | - Jaqueline BP Figueiredo
- University of Sao PauloDepartment of Obstetrics and Gynecology, Medical School of Ribeirao PretoHospital das Clinicas da FMRP‐USP, 8 andarCampus Universitario da USPRibeirao PretoSao PauloBrazil14048‐900
| | - Carolina O Nastri
- University of Sao PauloDepartment of Obstetrics and Gynecology, Medical School of Ribeirao PretoHospital das Clinicas da FMRP‐USP, 8 andarCampus Universitario da USPRibeirao PretoSao PauloBrazil14048‐900
| | | |
Collapse
|