1
|
Jabarpour M, Pouri S, Aleyasin A, Shabani Nashtaei M, Najafian A. Comparison of progesterone protocol versus gonadotropin-releasing hormone antagonist protocol in terms of preventing premature LH surge and assisted reproductive technology outcome in infertile women: a randomized controlled trial. Arch Gynecol Obstet 2024; 309:1999-2008. [PMID: 38421423 DOI: 10.1007/s00404-024-07387-4] [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: 09/12/2023] [Accepted: 01/12/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Progesterone can be used instead of GnRH agonists and antagonists in order to avert a premature LH surge during controlled ovarian stimulation (COS) protocol. Nonetheless, there is limited knowledge regarding its utilization. Thus, this study compared the effects of progesterone and GnRH antagonists (GnRH-ant) on premature LH surges and assisted reproductive technology (ART) results in infertile women undergoing ART. MATERIALS AND METHODS In this clinical trial, the progesterone protocol (study group) and GnRH-ant protocol (control group) were tested in 300 infertile individuals undergoing IVF/ICSI. The main outcome was the number of oocytes retrieved. The secondary outcomes included premature LH rise/surge, the quantity of follicles measuring ≥ 10 and 14 mm, oocyte maturity and fertilization rate, the number of viable embryos, high-quality embryo rate and pregnancy outcomes. RESULTS The study group exhibited a statistically significant increase in the number of retrieved oocytes, follicles measuring 14 mm or greater, and viable embryos compared to the control group (P < 0.05). The study group also increased oocyte maturity, chemical pregnancy rate, and clinical pregnancy rate (P < 0.05). Both groups had similar mean serum LH, progesterone, and E2 levels on trigger day. The control group had more premature LH rise than the study group, although this difference was not statistically significant. CONCLUSION In conclusion, it can be stated that the progesterone protocol and the GnRH-ant protocol exhibit similar rates of sudden premature LH surge in infertile patients. However, it is important to note that the two regiments differ in their outcomes in ART. TRIAL REGISTRATION This study was retrospectively registered in the Iranian website ( www.irct.ir ) for clinical trials registration ( http://www.irct.ir : IRCT-ID: IRCT20201029049183N, 2020-11-27).
Collapse
Affiliation(s)
- Masoome Jabarpour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Pouri
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Jalal-e-Al-e-Ahmad Hwy, Tehran, 1411713135, Iran
| | - Ashraf Aleyasin
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Jalal-e-Al-e-Ahmad Hwy, Tehran, 1411713135, Iran
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Najafian
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Jalal-e-Al-e-Ahmad Hwy, Tehran, 1411713135, Iran.
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Martínez-Moya M, Guerrero J, Girela JL, Pitas A, Bernabeu A, Bernabeu R, Castillo JC. Micronized natural progesterone (Seidigestan ®) vs GnRH antagonists for preventing the LH surge during controlled ovarian stimulation (PRO_NAT study): study protocol of a randomized clinical trial. Front Endocrinol (Lausanne) 2024; 15:1350154. [PMID: 38577571 PMCID: PMC10991791 DOI: 10.3389/fendo.2024.1350154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/16/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Progesterone-primed cycles effectively suppress the pituitary LH surge during ovarian stimulation in oocyte donors and in the infertile population. Particularly in oocyte donors, the use of synthetic progesterone (progestins) has been explored in prospective clinical trials, showing mixed results. This trial was designed to determine whether the use of micronized natural progesterone is as effective as the GnRH-antagonist protocol in terms of the number of mature oocytes (MII) retrieved in oocyte donation cycles as a primary outcome, and it also aims to explore the corresponding results in recipients as a secondary outcome. Methods We propose a prospective, open-label, non-inferiority clinical trial to compare a novel approach for oocyte donors with a control group, which follows the standard ovarian stimulation protocol used in our institution. A total of 150 donors (75 in each group) will be recruited and randomized using a computer algorithm. After obtaining informed consent, participants will be randomly assigned to one of two ovarian stimulation protocols: either the standard GnRH antagonist or the oral micronized natural progesterone protocol. Both groups will receive recombinant gonadotropins tailored to their antral follicle count and prior donation experiences, if any. The primary outcome is the number of mature metaphase II (MII) oocytes. Secondary measures include treatment duration, pregnancy outcomes in recipients, as well as the economic cost per MII oocyte obtained in each treatment regimen. Analyses for the primary outcome will be conducted in both the intention-to-treat (ITT) and per-protocol (PP) populations. Each donor can participate only once during the recruitment period. The estimated duration of the study is six months for the primary outcome and 15 months for the secondary outcomes. Discussion The outcomes of this trial have the potential to inform evidence-based adjustments in the management of ovarian stimulation protocols for oocyte donors. Clinical trial registration ClinicalTrials.gov, identifier, NCT05954962.
Collapse
Affiliation(s)
| | | | | | - A. Pitas
- Bernabeu Institute, Alicante, Spain
| | | | | | | |
Collapse
|
3
|
Vaiarelli A, Cimadomo D, Ruffa A, Rania E, Pittana E, Gallo C, Fiorenza A, Alviggi E, Alfano S, Carmelo R, Trabucco E, Alviggi C, Rosaria Campitiello M, Rienzi L, Maria Ubaldi F, Venturella R. Oocyte competence is comparable between progestin primed ovarian stimulation with Norethisterone acetate (NETA-PPOS) and GnRH-antagonist protocols: A matched case-control study in PGT-A cycles. Eur J Obstet Gynecol Reprod Biol 2024; 294:4-10. [PMID: 38171151 DOI: 10.1016/j.ejogrb.2023.12.035] [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: 08/31/2023] [Revised: 12/05/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE To outline oocyte competence after progestin primed ovarian stimulation with Norethisterone acetate (NETA-PPOS) compared to conventional GnRH-antagonist protocol. STUDY DESIGN Retrospective matched case-control study involving advanced-maternal-age women undergoing ICSI with PGT-A. 89 NETA-PPOS were matched with 178 control patients based on maternal age and ovarian reserve biomarkers. Both groups underwent recombinant-FSH OS with GnRH-agonist ovulation trigger and collected ≥1 MII. In the study group, NETA (10 mg/day) was administered orally starting from day2 of the menstrual cycle. Euploid blastocyst rate per cohort of metaphase-II oocytes (EBR per MII) was the primary outcome. All other embryological and clinical outcomes were reported. Gestational age, birthweight and length were also assessed. RESULTS The EBR per MII was comparable among PPOS and control (13.9 % ± 19.3 % versus 13.3 % ± 17.9 %; the sample size allowed to exclude up to a 10 % difference). Blastocysts morphology and developmental rate were similar. No difference was reported for all clinical outcomes among the 61 and 107 vitrified-warmed euploid single blastocyst transfers respectively conducted. The cumulative live birth delivery rate per concluded cycles was also comparable (24.7 % versus 21.9 %). Neonatal outcomes were analogous. CONCLUSIONS Oocyte competence after NETA-PPOS and standard OS is comparable. This evidence is reassuring and, because of its lower cost and possibly higher patients' compliance, supports PPOS administration whenever the patients are indicated to freeze-all (e.g., fertility preservation, PGT-A, oocyte donation). More data are required about follicle recruitment, oocyte yield, gestational and perinatal outcomes. Randomized-controlled-trials are advisable to confirm our evidence.
Collapse
Affiliation(s)
- Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy.
| | - Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Alessandro Ruffa
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy; Physiopathology of Reproduction and IVF Unit, Obstetrics and Gynecology 1U, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Erika Rania
- Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi Magna Graecia di Catanzaro, Catanzaro, Italy
| | - Erika Pittana
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy; Università degli Studi della Campania Luigi Vanvitelli, Caserta, Italy
| | - Cinzia Gallo
- Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi Magna Graecia di Catanzaro, Catanzaro, Italy
| | - Alessia Fiorenza
- Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi Magna Graecia di Catanzaro, Catanzaro, Italy
| | - Erminia Alviggi
- IVIRMA Global Research Alliance, GENERA, Clinica Ruesch, Naples, Italy
| | - Simona Alfano
- IVIRMA Global Research Alliance, GENERA, Clinica Ruesch, Naples, Italy
| | - Ramona Carmelo
- IVIRMA Global Research Alliance, GENERA, Clinica Ruesch, Naples, Italy
| | | | - Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, Salerno, Italy
| | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy; Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | | | - Roberta Venturella
- Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi Magna Graecia di Catanzaro, Catanzaro, Italy
| |
Collapse
|
4
|
Glujovsky D, Pesce R, Miguens M, Sueldo C, Ciapponi A. Progestogens for prevention of luteinising hormone (LH) surge in women undergoing controlled ovarian hyperstimulation as part of an assisted reproductive technology (ART) cycle. Cochrane Database Syst Rev 2023; 11:CD013827. [PMID: 38032057 PMCID: PMC10687848 DOI: 10.1002/14651858.cd013827.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
BACKGROUND Currently, gonadotrophin releasing hormone (GnRH) analogues are used to prevent premature ovulation in ART cycles. However, their costs remain high, the route of administration is invasive and has some adverse effects. Oral progestogens could be cheaper and effective to prevent a premature LH surge. OBJECTIVES To evaluate the effectiveness and safety of using progestogens to avoid spontaneous ovulation in women undergoing controlled ovarian hyperstimulation (COH). SEARCH METHODS We searched the Cochrane Gynaecology and Fertility Group trials register, CENTRAL, MEDLINE, Embase and PsycINFO in Dec 2021. We contacted study authors and experts to identify additional studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) that included progestogens for ovulation inhibition in women undergoing controlled ovarian hyperstimulation (COH). DATA COLLECTION AND ANALYSIS We used standard methodological procedures recommended by Cochrane, including the risk of bias (RoB) assessment. The primary review outcomes were live birth rate (LBR) and oocyte pick-up cancellation rate (OPCR). Secondary outcomes were clinical pregnancy rate (CPR), cumulative pregnancy, miscarriage rate (MR), multiple pregnancies, LH surge, total and MII oocytes, days of stimulation, dose of gonadotropins, and moderate/severe ovarian hyperstimulation syndrome (OHSS) rate. The primary analyses were restricted to studies at overall low and some concerns RoB, and sensitivity analysis included all studies. We used the GRADE approach to assess the certainty of evidence. MAIN RESULTS We included 14 RCTs (2643 subfertile women undergoing ART, 47 women used oocyte freezing for fertility preservation and 534 oocyte donors). Progestogens versus GnRH antagonists We are very uncertain of the effect of medroxyprogesterone acetate (MPA) 10 mg compared with cetrorelix on the LBR in poor responders (odds ratio (OR) 1.25, 95% confidence interval (CI) 0.73 to 2.13, one RCT, N = 340, very-low-certainty evidence), suggesting that if the chance of live birth following GnRH antagonists is assumed to be 18%, the chance following MPA would be 14% to 32%. There may be little or no difference in OPCR between progestogens and GnRH antagonists, but due to wide Cs (CIs), we are uncertain (OR 0.92, 95%CI 0.42 to 2.01, 3 RCTs, N = 648, I² = 0%, low-certainty evidence), changing the chance of OPCR from 4% with progestogens to 2% to 8%. Given the imprecision found, no conclusions can be retrieved on CPR and MR. Low-quality evidence suggested that using micronised progesterone in normo-responders may increase by 2 to 6 the MII oocytes in comparison to GnRH antagonists. There may be little or no differences in gonadotropin doses. Progestogens versus GnRH agonists Results were uncertain for all outcomes comparing progestogens with GnRH agonists. One progestogen versus another progestogen The analyses comparing one progestogen versus another progestogen for LBR did not meet our criteria for primary analyses. The OPCR was probably lower in the MPA 10 mg in comparison to MPA 4 mg (OR 2.27, 95%CI 0.90 to 5.74, one RCT, N = 300, moderate-certainty evidence), and MPA 4 mg may be lower than micronised progesterone 100 mg, but due to wide CI, we are uncertain of the effect (OR 0.81, 95%CI 0.43 to 1.53, one RCT, N = 300, low-certainty evidence), changing the chance of OPCR from 5% with MPA 4 mg to 5% to22%, and from 17% with micronised progesterone 100 mg to 8% to 24%. When comparing dydrogesterone 20 mg to MPA, the OPCR is probably lower in the dydrogesterone group in comparison to MPA 10 mg (OR 1.49, 95%CI 0.80 to 2.80, one RCT, N = 520, moderate-certainty evidence), and it may be lower in dydrogesterone group in comparison to MPA 4 mg but due to wide confidence interval, we are uncertain of the effect (OR 1.19, 95%CI 0.61 to 2.34, one RCT, N = 300, low-certainty evidence), changing the chance of OPCR from 7% with dydrogesterone 20 to 6-17%, and in MPA 4 mg from 12% to 8% to 24%. When comparing dydrogesterone 20 mg to micronised progesterone 100 mg, the OPCR is probably lower in the dydrogesterone group (OR 1.54, 95%CI 0.94 to 2.52, two RCTs, N=550, I² = 0%, moderate-certainty evidence), changing OPCR from 11% with dydrogesterone to 10% to 24%. We are very uncertain of the effect in normo-responders of micronised progesterone 100 mg compared with micronised progesterone 200 mg on the OPCR (OR 0.35, 95%CI 0.09 to 1.37, one RCT, N = 150, very-low-certainty evidence). There is probably little or no difference in CPR and MR between MPA 10 mg and dydrogesterone 20 mg. There may be little or no differences in MII oocytes and gonadotropins doses. No cases of moderate/severe OHSS were reported in most of the groups in any of the comparisons. AUTHORS' CONCLUSIONS Little or no differences in LBR may exist when comparing MPA 4 mg with GnRH agonists in normo-responders. OPCR may be slightly increased in the MPA 4 mg group, but MPA 4 mg reduces the doses of gonadotropins in comparison to GnRH agonists. Little or no differences in OPCR may exist between progestogens and GnRH antagonists in normo-responders and donors. However, micronised progesterone could improve by 2 to 6 MII oocytes. When comparing one progestogen to another, dydrogesterone suggested slightly lower OPCR than MPA and micronised progesterone, and MPA suggested slightly lower OPCR than the micronised progesterone 100 mg. Finally, MPA 10 mg suggests a lower OPCR than MPA 4 mg. There is uncertainty regarding the rest of the outcomes due to imprecision and no solid conclusions can be drawn.
Collapse
Affiliation(s)
- Demián Glujovsky
- Reproductive Medicine, CEGYR (Centro de Estudios en Genética y Reproducción), Buenos Aires, Argentina
| | - Romina Pesce
- Reproductive Medicine, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Miguens
- Reproductive Medicine, CEGYR (Centro de Estudios en Genética y Reproducción), Buenos Aires, Argentina
| | - Carlos Sueldo
- Reproductive Medicine, CEGYR (Centro de Estudios en Ginecologia y Reproducción), Buenos Aires, Argentina
| | - Agustín Ciapponi
- Argentine Cochrane Centre, Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
| |
Collapse
|
5
|
Pech M, Steinbach C, Kocour M, Prokopová I, Šandová M, Bořík A, Lutz I, Kocour Kroupová H. Effects of mifepristone, a model compound with anti-progestogenic activity, on the development of African clawed frog (Xenopus laevis). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 263:106694. [PMID: 37716317 DOI: 10.1016/j.aquatox.2023.106694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/23/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
The objective of this study was to assess the effects of a model substance with anti-progestogenic activity on development of African clawed frog (Xenopus laevis) from tadpole to juvenile stage. Mifepristone, a synthetic progesterone receptor-blocking steroid hormone used in medicine as an abortifacient, was chosen as a model compound with anti-progestogenic activity. In the experiment, African clawed frog tadpoles were exposed to mifepristone at three concentrations (2, 21, and 215 ng L-1). A control group was exposed to dimethyl sulfoxide (DMSO; 0.001 %). The experiment started when tadpoles reached stages 47-48 according to Nieuwkoop and Faber (NF; 1994) and continued until stage NF 66, when metamorphosis was complete. Exposure to mifepristone had no significant effect on the rate of tadpole development, occurrence of morphological anomalies, weight, body length, or sex ratio. Mortality was within an acceptable range of 0-3.6 % throughout the test and did not differ among the groups. Histopathological examination of the gonads and thyroid gland revealed no significant changes. Therefore, we can conclude that mifepristone had no negative effect on development of the African clawed frog up to juvenile stage. Nevertheless, at the highest tested mifepristone concentration (215 ng L-1), gene expression analysis revealed up-regulation of mRNA expression of nuclear progesterone receptor (npr), membrane progesterone receptor (mpr), estrogen receptor beta (esrβ), and luteinizing hormone (lh) in the brain-pituitary complex of exposed frogs at stage NF 66. Higher mRNA expression of npr was also found in frogs exposed to 22 ng L-1 mifepristone compared to the solvent control. These findings confirmed the anti-progestogenic activity of mifepristone in frogs because the up-regulation of progesterone receptors occurs if progesterone availability in the body is reduced. All the observed changes in combination may have negative consequences for reproduction and reproductive behavior later in life.
Collapse
Affiliation(s)
- Michal Pech
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic.
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Martin Kocour
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Ilona Prokopová
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Marie Šandová
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Adam Bořík
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Ilka Lutz
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Müggelseedamm 310, Berlin 12587, Federal Republic of Germany
| | - Hana Kocour Kroupová
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| |
Collapse
|
6
|
Sun X, Chen X, Zhao J, Ma C, Yan C, Liswaniso S, Xu R, Qin N. Transcriptome comparative analysis of ovarian follicles reveals the key genes and signaling pathways implicated in hen egg production. BMC Genomics 2021; 22:899. [PMID: 34911438 PMCID: PMC8672471 DOI: 10.1186/s12864-021-08213-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 11/26/2021] [Indexed: 01/19/2023] Open
Abstract
Background Ovarian follicle development plays an important role in determination of poultry egg production. The follicles at the various developmental stages possess their own distinct molecular genetic characteristics and have different biological roles in chicken ovary development and function. In the each stage, several genes of follicle-specific expression and biological pathways are involved in the vary-sized follicular development and physiological events. Identification of the pivotal genes and signaling pathways that control the follicular development is helpful for understanding their exact regulatory functions and molecular mechanisms underlying egg-laying traits of laying hens. Results The comparative mRNA transcriptomic analysis of ovarian follicles at three key developmental stages including slow growing white follicles (GWF), small yellow follicles (SYF) of recruitment into the hierarchy, and differentiated large yellow follicles (LYF), was accomplished in the layers with lower and higher egg production. Totally, 137, 447, and 229 of up-regulated differentially expressed genes (DEGs), and 99, 97, and 157 of down-regulated DEGs in the GWF, SYF and LYF follicles, including VIPR1, VIPR2, ADRB2, and HSD17B1 were identified, respectively. Moreover, NDUFAB1 and GABRA1 genes, two most promising candidates potentially associated with egg-laying performance were screened out from the 13 co-expressed DEGs in the GWF, SYF and LYF samples. We further investigated the biological effects of NDUFAB1 and GABRA1 on ovarian follicular development and found that NDUFAB1 promotes follicle development by stimulating granulosa cell (GC) proliferation and decreasing cell apoptosis, increases the expression of CCND1 and BCL-2 but attenuates the expression of caspase-3, and facilitates steroidogenesis by enhancing the expression of STAR and CYP11A1. In contrast, GABRA1 inhibits GC proliferation and stimulates cell apoptosis, decreases the expression of CCND1, BCL-2, STAR, and CYP11A1 but elevates the expression of caspase-3. Furthermore, the three crucial signaling pathways such as PPAR signaling pathway, cAMP signaling pathway and neuroactive ligand-receptor interaction were significantly enriched, which may play essential roles in ovarian follicle growth, differentiation, follicle selection, and maturation. Conclusions The current study provided new molecular data for insight into the regulatory mechanism underlying ovarian follicle development associated with egg production in chicken. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08213-w.
Collapse
Affiliation(s)
- Xue Sun
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiaoxia Chen
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jinghua Zhao
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Chang Ma
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Chunchi Yan
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Simushi Liswaniso
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Rifu Xu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China. .,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Ning Qin
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China. .,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
7
|
Guan S, Feng Y, Huang Y, Huang J. Progestin-Primed Ovarian Stimulation Protocol for Patients in Assisted Reproductive Technology: A Meta-Analysis of Randomized Controlled Trials. Front Endocrinol (Lausanne) 2021; 12:702558. [PMID: 34531825 PMCID: PMC8438422 DOI: 10.3389/fendo.2021.702558] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives Progestin-primed ovarian stimulation (PPOS) is a new ovarian stimulation protocol that can block the luteinizing hormone (LH) surge through progesterone instead of traditional down regulating or gonadotropin-releasing hormone (GnRH) antagonist, and in order to achieve multi-follicle recruitment. This paper aims to investigate the effectiveness of PPOS and its suitability for infertile patients with different ovarian reserve functions. Methods We searched published randomized controlled trials (RCTs) about PPOS on Cochrane Library, PubMed, Embase, and Web of Science. The search period spanned from January 1, 2015 to November 16, 2020. The data were extracted, and the meta-analysis was performed on ovarian stimulation as well as embryological and clinical outcomes. The outcomes were pooled by a random effects model, and the risk of heterogeneity was evaluated. Subgroup analysis was performed for different ovarian reserve patients. Results The clinical pregnancy rates and live birth or ongoing pregnancy rates with the PPOS protocol were not different from those with the control group. In the diminished ovarian reserve (DOR) subgroup, the PPOS protocol had a lower rate of premature LH surge [RR = 0.03, 95% CI = 0.01 to 0.13, p < 0.001]. The PPOS protocol had a lower rate of ovarian hyperstimulation syndrome (OHSS) [RR = 0.52, 95% CI = 0.36 to 0.76, p < 0.001, I2 = 0.00%]. The secondary outcomes showed that the number of oocytes retrieved, MII oocytes, and viable embryos was higher than that of the control protocol in DOR patients [(MD = 0.33, 95% CI = 0.30 to 0.36, p < 0.001), (MD = 0.30, 95% CI = 0.27 to 0.33, p < 0.001), (MD = 0.21, 95% CI = 0.18 to 0.24, p < 0.001)] and normal ovarian reserve (NOR) patients [(MD = 1.41, 95% CI = 0.03 to 2.78, p < 0.001), (MD = 1.19, 95% CI = 0.04 to 2.35, p < 0.001), (MD = 1.01, 95% CI = 0.21 to 1.81, p = 0.01)]. Conclusion The findings suggest that PPOS is an effective ovarian stimulation protocol and is beneficial for patients with different ovarian reserve functions, which needs to be validated in more RCTs with larger samples.
Collapse
Affiliation(s)
- Shaogen Guan
- Reproductive Medicine Center, The First People’s Hospital of Foshan, Foshan, China
| | - Yuezhi Feng
- Reproductive Medicine Center, The First People’s Hospital of Foshan, Foshan, China
| | - Yonghan Huang
- Reproductive Medicine Center, The First People’s Hospital of Foshan, Foshan, China
| | - Jia Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
8
|
Lu BJ, Lin CJ, Lin BZ, Huang L, Chien LT, Chen CH. ART outcomes following ovarian stimulation in the luteal phase:a systematic review and meta-analysis. J Assist Reprod Genet 2021; 38:1927-1938. [PMID: 34036454 PMCID: PMC8417163 DOI: 10.1007/s10815-021-02237-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE This study aimed to evaluate the impact of luteal phase ovarian stimulation (LPS) on the outcomes of assisted reproductive technology (ART) for infertile couples and patients desiring non-urgent egg cryopreservation. METHODS We included all studies reported patients who received LPS and that used follicular phase ovarian stimulation (FPS) as a comparison group until January 2021. Prior meta-analysis regarding the outcomes of LPS in double stimulation and fertility preservation have already been published, so these studies were excluded. Risk of Bias in Non-randomized Studies of Interventions was used to assess the study quality. The study was registered in the International Prospective Register of Systematic Reviews database (CRD42020183946). RESULTS Twelve studies with a total of 4433 patients were included. The regimen employed can be categorized into two groups, but there is currently no evidence to support one over the other. After we excluded the largest study, the clinical pregnancy rate and live birth rate were similar after FPS and LPS. There were significantly more stimulation days and total gonadotropins used in the LPS group. After subgroup analysis, we found that poor responders received significantly more cumulus oocyte complexes (+0.64) in the LPS group. CONCLUSION Current evidence indicates that patients in the LPS group could achieve pregnancy outcomes non-inferior to those in the FPS group. Because of current debate over freeze-all policy and the limited data about live birth rate, the universal use of LPS is considered controversial. In the future, more well-designed studies are necessary to investigate the indications for LPS and its cost-effectiveness.
Collapse
Affiliation(s)
- Buo-Jia Lu
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, No.252, Wu Hsing Street, Taipei, 110 Taiwan
| | - Chien-Ju Lin
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, No. 690, Section 2, Guangfu Road, East District, Hsinchu, 30071 Taiwan
| | - Bou-Zenn Lin
- Department of Gastroenterology, Ren-Ai Branch, Taipei City Hospital, No. 10, Sec. 4, Ren-Ai Rd., Da’an Dist, Taipei, 106 Taiwan
| | - Li Huang
- Department of Family Medicine, Taipei Medical University Hospital, No.252, Wu Hsing Street, Taipei, 110 Taiwan
| | - Li-Ting Chien
- Taipei Medical University Library, No.250, Wu Hsing Street, Taipei, 110 Taiwan
| | - Chi-Huang Chen
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, No.252, Wu Hsing Street, Taipei, 110 Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, No.250, Wu Hsing Street, Taipei, Taiwan
| |
Collapse
|
9
|
Li X, Zhang X, Cai J, Pang J, Sun P, Chen J, Wang M, Liang X. Granulosa cells apoptosis and follicular fluid hormones: comparison of progestin-primed ovarian stimulation versus GnRH antagonist protocols. Gynecol Endocrinol 2021; 37:609-613. [PMID: 33092415 DOI: 10.1080/09513590.2020.1822802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To explore the effect of progestin-primed ovarian stimulation protocol (PPOS) on mural granulosa cells (GCs) apoptosis and hormonal profiles in follicular fluid (FF) and efficacy over GnRH antagonist (GnRH-A) protocols. METHODS We performed a prospective cohort study from June through August 2017 at a tertiary teaching hospital. 63 Patients meeting our criteria were recruited in this prospective study voluntarily and stratified to PPOS or GnRH-A group randomly. Mural GCs and FF were collected during oocyte retrieval. Apoptosis of GCs was assessed using the Annexin V-affinity assay by flow cytometry and hormonal profiles in FF were measured using electrochemiluminescence. RESULTS A total of 63 women were assessed for eligibility, with 25 cases in PPOS group and 38 in GnRH-A group. Difference of early stage apoptosis rate, late stage apoptosis rate, and total apoptosis rate did not reach statistical significance between groups. Meanwhile, concentrations of hormones in FF were comparable in two groups. No statistically significant differences were observed in number of oocytes retrieved, mature oocyte rate, fertilization rate, and top-quality embryos rate. No patients experienced premature LH surge in both groups during the study. CONCLUSION Compared to GnRH antagonist protocol, PPOS had comparable laboratory outcomes, GCs apoptosis rate and hormonal profiles in FF. PPOS is an effective and safe alternative option to provide controlled ovarian hyperstimulation (COH).
Collapse
Affiliation(s)
- Xiaolan Li
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Xiaoli Zhang
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Jiawei Cai
- Gastrointestinal Surgical Department, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Jiahui Pang
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Peng Sun
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Jianhui Chen
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Meng Wang
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Xiaoyan Liang
- Reproductive Medicine Research Center, Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| |
Collapse
|
10
|
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: 1.3] [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
|
11
|
Glujovsky D, Pesce R, Miguens M, Sueldo C, Ciapponi A. Progestogens for prevention of luteinising hormone (LH) surge in women undergoing controlled ovarian hyperstimulation as part of an assisted reproductive technology (ART) cycle. Hippokratia 2020. [DOI: 10.1002/14651858.cd013827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Demián Glujovsky
- Reproductive Medicine; CEGYR (Centro de Estudios en Genética y Reproducción); Buenos Aires Argentina
| | - Romina Pesce
- Reproductive Medicine; Hospital Italiano de Buenos Aires; Buenos Aires Argentina
| | - Mariana Miguens
- Reproductive Medicine; CEGYR (Centro de Estudios en Genética y Reproducción); Buenos Aires Argentina
| | - Carlos Sueldo
- Reproductive Medicine; CEGYR (Centro de Estudios en Ginecologia y Reproducción); Buenos Aires Argentina
| | - Agustín Ciapponi
- Argentine Cochrane Centre; Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET); Buenos Aires Argentina
| |
Collapse
|
12
|
A novel GnRH-antagonist protocol by switching to medroxyprogesterone when patients being at risk of ovarian hyperstimulation syndrome during ovarian stimulation. J Formos Med Assoc 2020; 119:1642-1649. [DOI: 10.1016/j.jfma.2019.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/19/2019] [Accepted: 12/16/2019] [Indexed: 01/24/2023] Open
|
13
|
Ata B, Capuzzo M, Turkgeldi E, Yildiz S, La Marca A. Progestins for pituitary suppression during ovarian stimulation for ART: a comprehensive and systematic review including meta-analyses. Hum Reprod Update 2020; 27:48-66. [PMID: 33016316 DOI: 10.1093/humupd/dmaa040] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/02/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Progestins are capable of suppressing endogenous LH secretion from the pituitary. Progestins can be used orally and are less expensive than GnRH analogues. However, early endometrial exposure to progestin precludes a fresh embryo transfer (ET), but the advent of vitrification and increasing number of oocyte cryopreservation cycles allow more opportunities for using progestins for pituitary suppression. OBJECTIVE AND RATIONALE This review summarizes: the mechanism of pituitary suppression by progestins; the effectiveness of progestins when compared with GnRH analogues and with each other; the effect of progestins on oocyte and embryo developmental potential and euploidy status; and the cost-effectiveness aspects of progestin primed stimulation. Future research priorities are also identified. SEARCH METHODS The Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE via PubMed, the Web of Science and Scopus were screened with a combination of keywords related to ART, progesterone, GnRH analogue and ovarian stimulation, in various combinations. The search period was from the date of inception of each database until 1 April 2020. Only full text papers published in English were included. OUTCOMES Overall, the duration of stimulation, gonadotrophin consumption and oocyte yield were similar with progestins and GnRH analogues. However, sensitivity analyses suggested that progestins were associated with significantly lower gonadotrophin consumption than the long GnRH agonist protocol (mean difference (MD) = -648, 95% CI = -746 to -550 IU) and significantly higher gonadotrophin consumption than the short GnRH agonist protocol (MD = 433, 95% CI = 311 to 555 IU). Overall, live birth, ongoing and clinical pregnancy rates per ET were similar with progestins and GnRH analogues. However, when progestins were compared with GnRH agonists, sensitivity analyses including women with polycystic ovary syndrome (risk ratio (RR) = 1.27, 95% CI = 1.06 to 1.53) and short GnRH agonist protocols (RR = 1.14, 95% CI = 1.02 to 1.28) showed significantly higher clinical pregnancy rates with progestins. However, the quality of evidence is low. Studies comparing medroxyprogesterone acetate, dydrogesterone and micronized progesterone suggested similar ovarian response and pregnancy outcomes. The euploidy status of embryos from progestin primed cycles was similar to that of embryos from conventional stimulation cycles. Available information is reassuring regarding obstetric and neonatal outcomes with the use of progestins. Despite the lower cost of progestins than GnRH analogues, the mandatory cryopreservation of all embryos followed by a deferred transfer may increase cost per live birth with progestins as compared to an ART cycle culminating in a fresh ET. WIDER IMPLICATIONS Progestins can present an effective option for women who do not contemplate a fresh ET, e.g. fertility preservation, anticipated hyper responders, preimplantation genetic testing, oocyte donors, double stimulation cycles.
Collapse
Affiliation(s)
- Baris Ata
- Department of Obstetrics and Gynecology, Koç University School of Medicine, Istanbul, Turkish Republic
| | - Martina Capuzzo
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| | - Engin Turkgeldi
- Department of Obstetrics and Gynecology, Koç University School of Medicine, Istanbul, Turkish Republic
| | - Sule Yildiz
- Department of Obstetrics and Gynecology, Koç University School of Medicine, Istanbul, Turkish Republic
| | - Antonio La Marca
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Wright AA, Fayad GN, Selgrade JF, Olufsen MS. Mechanistic model of hormonal contraception. PLoS Comput Biol 2020; 16:e1007848. [PMID: 32598357 PMCID: PMC7365466 DOI: 10.1371/journal.pcbi.1007848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 07/16/2020] [Accepted: 04/03/2020] [Indexed: 02/02/2023] Open
Abstract
Contraceptive drugs intended for family planning are used by the majority of married or in-union women in almost all regions of the world. The two most prevalent types of hormones associated with contraception are synthetic estrogens and progestins. Hormonal based contraceptives contain a dose of a synthetic progesterone (progestin) or a combination of a progestin and a synthetic estrogen. In this study we use mathematical modeling to understand better how these contraceptive paradigms prevent ovulation, special focus is on understanding how changes in dose impact hormonal cycling. To explain this phenomenon, we added two autocrine mechanisms essential to achieve contraception within our previous menstrual cycle models. This new model predicts mean daily blood concentrations of key hormones during a contraceptive state achieved by administering progestins, synthetic estrogens, or a combined treatment. Model outputs are compared with data from two clinical trials: one for a progestin only treatment and one for a combined hormonal treatment. Results show that contraception can be achieved with synthetic estrogen, with progestin, and by combining the two hormones. An advantage of the combined treatment is that a contraceptive state can be obtained at a lower dose of each hormone. The model studied here is qualitative in nature, but can be coupled with a pharmacokinetic/pharamacodynamic (PKPD) model providing the ability to fit exogenous inputs to specific bioavailability and affinity. A model of this type may allow insight into a specific drug's effects, which has potential to be useful in the pre-clinical trial stage identifying the lowest dose required to achieve contraception.
Collapse
Affiliation(s)
- A. Armean Wright
- Department of Mathematics and Program in Biomathematics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Ghassan N. Fayad
- Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - James F. Selgrade
- Department of Mathematics and Program in Biomathematics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Mette S. Olufsen
- Department of Mathematics and Program in Biomathematics, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
15
|
Use of progestins to inhibit spontaneous ovulation during ovarian stimulation: the beginning of a new era? Reprod Biomed Online 2019; 39:321-331. [DOI: 10.1016/j.rbmo.2019.03.212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/27/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
|
16
|
Del Río JP, Alliende MI, Molina N, Serrano FG, Molina S, Vigil P. Steroid Hormones and Their Action in Women's Brains: The Importance of Hormonal Balance. Front Public Health 2018; 6:141. [PMID: 29876339 PMCID: PMC5974145 DOI: 10.3389/fpubh.2018.00141] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/27/2018] [Indexed: 12/19/2022] Open
Abstract
Sex hormones significantly impact women's lives. Throughout the different stages of life, from menarche to menopause and all stages in between, women experience dramatic fluctuations in the levels of progesterone and estradiol, among other hormones. These fluctuations affect the body as a whole, including the central nervous system (CNS). In the CNS, sex hormones act via steroid receptors. They also have an effect on different neurotransmitters such as GABA, serotonin, dopamine, and glutamate. Additionally, studies show that sex hormones and their metabolites influence brain areas that regulate mood, behavior, and cognitive abilities. This review emphasizes the benefits a proper hormonal balance during the different stages of life has in the CNS. To achieve this goal, it is essential that hormone levels are evaluated considering a woman's age and ovulatory status, so that a correct diagnosis and treatment can be made. Knowledge of steroid hormone activity in the brain will give women and health providers an important tool for improving their health and well-being.
Collapse
Affiliation(s)
| | | | | | | | | | - Pilar Vigil
- Reproductive Health Research InstituteSantiago, Chile
- Vicerrectoría de ComunicacionesPontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
The role of combining medroxyprogesterone 17-acetate with human menopausal gonadotropin in mouse ovarian follicular development. Sci Rep 2018. [PMID: 29535409 PMCID: PMC5849710 DOI: 10.1038/s41598-018-22797-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Medroxyprogesterone 17-acetate (MPA) combined with human menopausal gonadotropin (hMG) has been effectively used for ovarian stimulation in clinical practice. However, the molecular mechanism of MPA + hMG treatment in follicular development is poorly described. Here we performed a study to investigate the impact of MPA + hMG on ovarian stimulation utilizing a mouse model in vivo. Forty female BALB/C mice were randomly divided into four groups of 10 each and treated during ciestrus stage and continued for 5 days: control group, MPA group, hMG group, and MPA + hMG group. Morphological and molecular biology methods were used for detecting serum hormones and ovarian function. MPA + hMG group exhibited increasing follicle stimulating hormone (FSH), antral follicle, FSH receptor (FSHR) and phosphorylated mammal target of rapamycin (p-mTOR), and decreasing luteinizing hormone (LH), estradiol (E2), progesterone (P), corpus luteum, phosphoinositide 3-kinase (PI3K), Akt and mTOR compared with control group. In contrast, MPA + hMG group showed reduced FSH, LH, E2, P, corpus luteum, LH receptor (LHR), and activated PI3K,/Akt/mTOR pathway compared with hMG group (P < 0.05). Collectively, these data definitively established that MPA plus hMG may modulate the hormone, hormone receptor and PI3K/Akt/mTOR signaling pathway to influence follicular development in the mouse ovary. Our study provides overwhelming support for MPA + hMG as an effective treatment for infertility in women.
Collapse
|
18
|
Massin N. New stimulation regimens: endogenous and exogenous progesterone use to block the LH surge during ovarian stimulation for IVF. Hum Reprod Update 2017; 23:211-220. [PMID: 28062551 DOI: 10.1093/humupd/dmw047] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/11/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The advent of embryo and oocyte vitrification today gives reproductive specialists an opportunity to consider new strategies for improving the practice and results of IVF attempts. As the freezing of entire cohorts does not compromise, and may even improve, the results of IVF attempts, it is possible to break away from the standard sequence of stimulation-retrieval-transfer. The constraints associated with ovarian stimulation in relation to the potential harmful effects of the hormonal environment on endometrial receptivity can be avoided. OBJECTIVE AND RATIONALE This review will look at the new stimulation protocols where progesterone is used to block the LH surge. Thanks to 'freeze all' strategies, the increase in progesterone could actually be no longer a cause for concern. There are two ways of using progesterone, whether it be endogenous, as in luteal phase stimulation, or exogenous, as in the use of progesterone in the follicular phase i.e. progestin primed ovarian stimulation. SEARCH METHODS A literature search was carried out (until September 2016) on MEDLINE. The following text words were utilized to generate the list of citations: progestin primed ovarian stimulation, luteal phase stimulation, luteal stimulation, duostim, double stimulation, random start. Articles and their references were then examined in order to identify other potential studies. All of the articles are reported in this review. OUTCOMES The use of progesterone during ovarian stimulation is effective in blocking the LH surge, whether endogenous or exogenous, and it does not affect the number of oocytes collected or the quality of the embryos obtained. Its main constraint is that it requires total freezing and delayed transfer. A variety of stimulation protocols can be derived from these two methods, and their implications are discussed, from fertility preservation to ovarian response profiles to organization for the patients and clincs. These new regimens enable more flexibility and are of emerging interest in daily practice. However, their medical and economic significance remains to be demonstrated. WIDER IMPLICATIONS The use of luteal phase or follicular phase protocols with progestins could rapidly develop in the context of oocyte donation and fertility preservation not related to oncology. Their place could develop even more in the general population of patients in IVF programs. The strategy of total freezing continues to develop, thanks to technical improvements, in particular vitrification and PGS on blastocysts, and thanks to studies showing improvements in embryo implantation when the transfer take place far removed from the hormonal changes caused by ovarian stimulation.
Collapse
Affiliation(s)
- Nathalie Massin
- Centre d'Assistance Médicale à la Procréation, Centre Hospitalier Intercommual, Université Paris 12, 40 Avenue de Verdun, 94000Créteil, France
| |
Collapse
|
19
|
Lira-Albarrán S, Larrea-Schiavon MF, González L, Durand M, Rangel C, Larrea F. The effects of levonorgestrel on FSH-stimulated primary rat granulosa cell cultures through gene expression profiling are associated to hormone and folliculogenesis processes. Mol Cell Endocrinol 2017; 439:337-345. [PMID: 27663078 DOI: 10.1016/j.mce.2016.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 12/18/2022]
Abstract
Levonorgestrel (LNG), a synthetic progestin, is used in emergency contraception (EC). The mechanism is preventing or delaying ovulation at the level of the hypothalamic pituitary unit; however, little knowledge exists on LNG effects at the ovary. The aim of this study was to identify the effects of LNG on FSH-induced 17β-estradiol (E2) production, including LNG-mediated changes on global gene expression in rat granulosa cells (GC). Isolated GC from female Wistar rats were incubated in vitro in the presence or absence of human FSH and progestins. At the end of incubations, culture media and cells were collected for E2 and mRNA quantitation. The results showed the ability of LNG to inhibit both hFSH-induced E2 production and aromatase gene expression. Microarray analysis revealed that LNG treatment affects GC functionality particularly that related to folliculogenesis and steroid metabolism. These results may offer additional evidence for the mechanisms of action of LNG as EC.
Collapse
Affiliation(s)
- Saúl Lira-Albarrán
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Avenida Vasco de Quiroga No. 15, Ciudad de México 14080, México.
| | - Marco F Larrea-Schiavon
- Department of Computational Genomics, Instituto Nacional de Medicina Genómica, Periférico Sur No. 4809, Ciudad de México 14610, México.
| | - Leticia González
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Avenida Vasco de Quiroga No. 15, Ciudad de México 14080, México.
| | - Marta Durand
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Avenida Vasco de Quiroga No. 15, Ciudad de México 14080, México.
| | - Claudia Rangel
- Department of Computational Genomics, Instituto Nacional de Medicina Genómica, Periférico Sur No. 4809, Ciudad de México 14610, México.
| | - Fernando Larrea
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Avenida Vasco de Quiroga No. 15, Ciudad de México 14080, México.
| |
Collapse
|
20
|
Chabbert-Buffet N, Pintiaux A, Bouchard P. The immninent dawn of SPRMs in obstetrics and gynecology. Mol Cell Endocrinol 2012; 358:232-43. [PMID: 22415029 DOI: 10.1016/j.mce.2012.02.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/21/2012] [Accepted: 02/22/2012] [Indexed: 12/30/2022]
Abstract
Selective progesterone receptor modulators (SPRMs) have been developed since the late 70s when mifepristone was first described. They act through nuclear progesterone receptors and can have agonist or mixed agonist antagonist actions depending on the cell and tissue. Mifepristone has unique major antagonist properties allowing its use for pregnancy termination. Ulipristal acetate has been marketed in 2009 for emergency contraception and has been recently approved for preoperative myoma treatment. Further perspectives for SPRMs use include long term estrogen free contraception, endometriosis treatment. However long term applications will be possible only after confirmation of endometrial safety.
Collapse
Affiliation(s)
- Nathalie Chabbert-Buffet
- Obstetrics, Gynecology and Reproductive Medicine Department, AP-HP, Hospital Tenon, UPMC Paris 06, Paris, France.
| | | | | |
Collapse
|
21
|
Nakama K, Akune A, Kawate N, Takahashi M, Inaba T, Sameshima H, Tamada H. Delay of ovulation due to diets containing levonorgestrel in cynomolgus monkeys (Macaca fascicularis). J Vet Med Sci 2012; 74:1453-60. [PMID: 22785380 DOI: 10.1292/jvms.12-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was undertaken to develop a simple and practical method to control the time of ovulation in cynomolgus monkeys. Diets containing a synthetic gestagen, levonorgestrel (LNG) were given daily to normally cycling female monkeys for 2 weeks, and plasma concentrations of estradiol-17β and progesterone were determined by EIA in order to estimate the time of ovulation. Doses of LNG (0, 3.2, 8, 20, 50, or 125 μg) were given from Day 2 (Day 0 =the first day of menstruation) through Day 15. The numbers of days from the last administration of LNG to the estimated ovulation in the groups treated with LNG at 20 μg and above were significantly greater than those in the controls, and the values in the group treated with LNG at 50 μg were within a narrow range. In a second experiment, LNG was administered at 50 μg in different phases of the menstrual cycle (Days 9-22, 16-29, and 23-36), and the results indicated that ovulation occurred more than 12 days after the last administration in all monkeys, and the number of days from the last administration of LNG to the estimated ovulation in the group treated on Days 16-29 (luteal phase) was significantly greater than that in the group treated on Days 23-36. These results indicate that daily provision of a diet containing 50 μg LNG could be applicable for delaying ovulation, and suggest that the total level of (exogenous and endogenous) progestins is critical for determining the length of ovulation delay in cynomolgus monkeys.
Collapse
Affiliation(s)
- Kazuhiro Nakama
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd, Kagoshima, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Nariai T, Fujita K, Mori M, Katayama S, Hori S, Matsui K. SM-368229, a novel selective and potent non-steroidal mineralocorticoid receptor antagonist with strong urinary Na+ excretion activity. J Pharmacol Sci 2011; 115:346-53. [PMID: 21358118 DOI: 10.1254/jphs.10285fp] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Mineralocorticoid receptor (MR) antagonists, such as spironolactone (SPI) and eplerenone (EPL), are useful for the treatment of hypertension and heart failure. However, the use of these two agents has been limited due to endocrine disturbance (SPI) and poor drug action (EPL). In our search for safer and more effective MR antagonists, we identified SM-368229 as a novel non-steroidal MR antagonist. SM-368229 showed strong MR inhibitory activity with IC(50) values of 0.021 and 0.13 µM in the binding assay and reporter-gene assay, respectively. The selectivity of SM-368229 for MR was 18-fold higher than that for other steroid receptors, such as androgen, progesterone, and glucocorticoid receptors. SM-368229 dose-dependently increased urinary Na(+)/K(+) ratio with an ED(50) value of 5.6 mg/kg in adrenalectomized rats treated with deoxycorticosterone acetate, and its efficacy was superior to that of SPI (ED(50) = 14 mg/kg) or EPL (ED(50) = 147 mg/kg). Moreover, even at high doses of 100 and 300 mg/kg, SM-368229 showed very weak anti-androgenic effect in methyltestosterone-treated male rats and no progestagenic effect in estrus cycle synchronized female rats. These findings indicate that SM-368229 may offer a new promising therapeutic option for the treatment of hypertension and heart failure.
Collapse
Affiliation(s)
- Tetsuro Nariai
- Pharmacology Research Laboratory, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan
| | | | | | | | | | | |
Collapse
|
23
|
LV XH, SHI DZ. Effects of Levonorgestrel on Reproductive Hormone Levels and Their Receptor Expression in Mongolian Gerbils (Meriones unguiculatus). Exp Anim 2011; 60:363-71. [DOI: 10.1538/expanim.60.363] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Xiao-Hui LV
- College of Agriculture and Biotechnology, China Agricultural University
| | - Da-Zhao SHI
- College of Agriculture and Biotechnology, China Agricultural University
| |
Collapse
|
24
|
Chabbert-Buffet N, Ouzounian S, Kairis AP, Bouchard P. Contraceptive applications of progesterone receptor modulators. EUR J CONTRACEP REPR 2009; 13:222-30. [PMID: 18821461 DOI: 10.1080/13625180802267060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Beijerink N, Bhatti S, Okkens A, Dieleman S, Duchateau L, Kooistra H. Pulsatile plasma profiles of FSH and LH before and during medroxyprogesterone acetate treatment in the bitch. Theriogenology 2008; 70:179-85. [DOI: 10.1016/j.theriogenology.2008.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 11/24/2007] [Accepted: 03/14/2008] [Indexed: 11/29/2022]
|
26
|
Fensome A, Adams WR, Adams AL, Berrodin TJ, Cohen J, Huselton C, Illenberger A, Kern JC, Hudak VA, Marella MA, Melenski EG, McComas CC, Mugford CA, Slayden OD, Yudt M, Zhang Z, Zhang P, Zhu Y, Winneker RC, Wrobel JE. Design, Synthesis, and SAR of New Pyrrole-Oxindole Progesterone Receptor Modulators Leading to 5-(7-Fluoro-3,3-dimethyl-2-oxo-2,3-dihydro-1H-indol-5-yl)-1-methyl-1H-pyrrole-2-carbonitrile (WAY-255348). J Med Chem 2008; 51:1861-73. [DOI: 10.1021/jm701080t] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Andrew Fensome
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - William R. Adams
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Andrea L. Adams
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Tom J. Berrodin
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Jeff Cohen
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Christine Huselton
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Arthur Illenberger
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Jeffrey C. Kern
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Valerie A. Hudak
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Michael A. Marella
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Edward G. Melenski
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Casey C. McComas
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Cheryl A. Mugford
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Ov D. Slayden
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Matthew Yudt
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Zhiming Zhang
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Puwen Zhang
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Yuan Zhu
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Richard C. Winneker
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| | - Jay E. Wrobel
- Chemical and Screening Sciences, Biotransformation Division, Drug Safety and Metabolism, Women’s Health and Musculoskeletal Biology, and BioResources, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, and Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97005
| |
Collapse
|
27
|
Chabbert-Buffet N, Pintiaux-Kairis A, Bouchard P. Effects of the progesterone receptor modulator VA2914 in a continuous low dose on the hypothalamic-pituitary-ovarian axis and endometrium in normal women: a prospective, randomized, placebo-controlled trial. J Clin Endocrinol Metab 2007; 92:3582-9. [PMID: 17579200 DOI: 10.1210/jc.2006-2816] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Progestin-only pills, the main hormonal alternative to ethinyl estradiol-containing pills in women bearing vascular risk factors, are poorly tolerated due to irregular bleeding. In contrast, progesterone receptor modulators can inhibit ovulation, alter endometrial receptivity, and improve cycle control. OBJECTIVE We evaluated the effects of a new progesterone receptor modulator, VA2914, administered continuously for 3 months, on ovulation and endometrial maturation. DESIGN, SETTINGS, AND PATIENTS Forty-six normal women were included in a prospective, placebo-controlled, randomized trial, conducted in four referral centers. INTERVENTION VA2914 (2.5, 5, or 10 mg/d) was administered continuously for 84 d. Pelvic ultrasound (treatment d 67 and 77), hormonal monitoring (FSH, LH, estradiol, and progesterone on treatment d 59, 63, 67, 70, 74, 77, 80, and 84), and endometrial biopsy (treatment d 77) were performed. MAIN OUTCOME MEASURE Ovulation inhibition was assessed by the absence of progesterone values above 3 ng/ml at any time during treatment month 3. RESULTS Anovulation was observed in 81.8% women in the 5-mg group and 80% in the 10-mg group, and amenorrhea occurred in 81.2 and 90% of cases in the 5- and 10-mg groups. We did not detect any cases of endometrial hyperplasia despite estradiol levels that remained in the physiological follicular phase range throughout treatment cycle 3. CONCLUSIONS Continuous low-dose VA2914 can induce amenorrhea and inhibit ovulation without down-regulating estradiol levels or inducing endometrial hyperplasia in normal women. Long-term studies with a larger population are required to confirm the contraceptive efficacy of this regimen.
Collapse
Affiliation(s)
- Nathalie Chabbert-Buffet
- Department of Obstetrics and Gynecology, Hospital Tenon, 4 Rue de la Chine, 75020 Paris, France.
| | | | | |
Collapse
|
28
|
Beijerink NJ, Bhatti SFM, Okkens AC, Dieleman SJ, Mol JA, Duchateau L, Van Ham LML, Kooistra HS. Adenohypophyseal function in bitches treated with medroxyprogesterone acetate. Domest Anim Endocrinol 2007; 32:63-78. [PMID: 16455223 DOI: 10.1016/j.domaniend.2005.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 12/21/2005] [Accepted: 12/21/2005] [Indexed: 10/25/2022]
Abstract
The aim of this study was to investigate the effects of treatment with medroxyprogesterone acetate (MPA) on canine adenohypophyseal function. Five Beagle bitches were treated with MPA (10mg/kg, every 4 weeks) and their adenohypophyseal function was assessed in a combined adenohypophyseal function test. Four hypophysiotropic hormones (CRH, GHRH, GnRH, and TRH) were administered before and 2, 5, 8, and 11 months after the start of MPA treatment, and blood samples for determination of the plasma concentrations of ACTH, cortisol, GH, IGF-1, LH, FSH, prolactin, alpha-MSH, and TSH were collected at -15, 0, 5, 10, 20, 30, and 45 min after suprapituitary stimulation. MPA successfully prevented the occurrence of estrus, ovulation, and a subsequent luteal phase. MPA treatment did not affect basal and GnRH-induced plasma LH concentrations. The basal plasma FSH concentration was significantly higher at 2 months after the start of MPA treatment than before or at 5, 8, and 11 months after the start of treatment. The maximal FSH increment and the AUC for FSH after suprapituitary stimulation were significantly higher before treatment than at 5, 8, and 11 months of MPA treatment. Differences in mean basal plasma GH concentrations before and during treatment were not significant, but MPA treatment resulted in significantly elevated basal plasma IGF-1 concentrations at 8 and 11 months. MPA treatment did not affect basal and stimulated plasma ACTH concentrations, with the exception of a decreased AUC for ACTH at 11 months. In contrast, the maximal cortisol increment and the AUC for cortisol after suprapituitary stimulation were significantly lower during MPA treatment than prior to treatment. MPA treatment did not affect basal plasma concentrations of prolactin, TSH, and alpha-MSH, with the exception of slightly increased basal plasma TSH concentrations at 8 months of treatment. MPA treatment did not affect TRH-induced plasma concentrations of prolactin and TSH. In conclusion, the effects of chronic MPA treatment on adenohypophyseal function included increased FSH secretion, unaffected LH secretion, activation of the mammary GH-induced IGF-I secretion, slightly activated TSH secretion, suppression of the hypothalamic-pituitary-adrenocortical axis, and unaffected secretion of prolactin and alpha-MSH.
Collapse
Affiliation(s)
- N J Beijerink
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Obruca A, Korver T, Huber J, Killick SR, Landgren B, Struijs MJ. Ovarian function during and after treatment with the new progestagen Org 30659. Fertil Steril 2001; 76:108-15. [PMID: 11438328 DOI: 10.1016/s0015-0282(01)01824-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To evaluate ovarian function during 21 days of oral administration of different doses of Org 30659, a novel selective progestagenic steroid. DESIGN Randomized, double-blind, dose-finding study. SETTINGS Three centers in Austria, Sweden, and the United Kingdom. PARTICIPANTS Eighty-one healthy women 19-40 years of age with regular ovulatory cycles. INTERVENTION Daily oral administration of 0.060, 0.120, 0.180, or 0.240 mg of Org 30659, or 0.075 mg desogestrel (reference group), for 21 days. MAIN OUTCOME MEASURE(S) Once-daily measurements of follicular diameter and 17-beta estradiol, progesterone, FSH, and LH levels. RESULT(S) Daily treatment with Org 30659 for 21 days caused dose-dependent suppression of ovarian activity. No ovulation was observed in any study group. On average, ovulation returned 16.5 to 22.1 days after treatment. The effects of desogestrel, 0.075 mg, were similar to those of 0.060 and 0.120 mg of Org 30659. All doses were well tolerated, as shown by the type of side effects that occurred, the absence of an effect on physical and laboratory findings, and the low rate of study discontinuation. CONCLUSION(S) Daily oral administration of 0.060-0.240 mg of Org 30659 suppresses ovarian function to a level sufficient to inhibit ovulation. This effect is dose-dependent, and the suppressive effect is readily reversible at all doses tested. Org 30659 can thus be safely administered orally for 21 days to healthy female volunteers in a dosage of 0.060 mg/d to 0.240 mg/d.
Collapse
Affiliation(s)
- A Obruca
- University of Vienna, General Hospital, Department of Obstetrics and Gynaecology, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
30
|
van Heusden AM, Killick SR, Coelingh Bennink HJ, Fauser BC. Single monthly administration of the anti-progestagen Org 31710 in users of the 75 microg desogestrel progestagen-only pill: effects on pituitary-ovarian activity. Hum Reprod 2000; 15:629-36. [PMID: 10686210 DOI: 10.1093/humrep/15.3.629] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Endocrine and ultrasound effects were studied of an intermittent (every 28 days) oral administration of 150 mg of the anti-progestagen Org 31710 during the continued daily use of 75 microg desogestrel (DSG) for progestagen-only contraception. A randomized, double-blind, placebo-controlled two-centre study was conducted in 50 healthy volunteers. Serum luteinizing hormone (LH), follicle stimulating hormone (FSH), oestradiol and progesterone concentrations, and follicle number and size were studied, as well as endometrial thickness, which was assessed by transvaginal sonography at least twice weekly during a single medication cycle (cycle 3-5). Forty-eight women were evaluated (Org 31710, n = 25; placebo, n = 23). Seven ovulations were observed in the treated group versus none in the placebo group. LH concentrations were higher on days 9 and 11 and oestradiol concentrations lower on day 3 in the treated group, irrespective of whether ovulation occurred. No parameter could predict ovulation. Endometrial thickness was greater on cycle days 7-13 and 19 in the treated group. However, within the Org 31710 group, no significant differences were found in volunteers who did or did not ovulate. Observed differences may be attributed to a competitive effect of Org 31710 with progestagen-induced suppression of the pituitary-ovarian axis, altered oestradiol feedback mechanisms, and/or altered receptor availability.
Collapse
Affiliation(s)
- A M van Heusden
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
31
|
Ko C, In YH, Park-Sarge OK. Role of progesterone receptor activation in pituitary adenylate cyclase activating polypeptide gene expression in rat ovary. Endocrinology 1999; 140:5185-94. [PMID: 10537148 DOI: 10.1210/endo.140.11.7149] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well known that the pituitary gonadotropin surge induces progesterone receptor (PR) gene expression in luteinizing granulosa cells and that PR activation is critical for successful ovulation. To further understand the molecular mechanism(s) by which PR plays a role critical for granulosa cell functions, we wanted to identify progesterone-induced genes in granulosa cells. We employed a PCR-based subtraction cloning strategy to screen for genes expressed differentially in granulosa cells that were challenged with forskolin in the presence of progesterone or ZK98299. One such differentially expressed clone was identified as the pituitary adenylate cyclase activating polypeptide (PACAP). To begin to understand the relationship between PR activation and PACAP gene expression in luteinizing granulosa cells, we examined whether PR and PACAP messenger RNA (mRNA) expression is temporally correlated. In cultured granulosa cells, both human CG and forskolin induced PR and PACAP mRNA levels in a dose-dependent manner, as determined by semi-quantitative RT-PCR assays. However, the peak expression for PR and PACAP mRNAs was observed at 3 h and 6 h after hormone treatment, respectively. This time difference in cAMP-responsive expression of the PR and PACAP genes is due, at least in part, to the requirement of ongoing protein synthesis for PACAP expression, as demonstrated by the inhibitory effect of cycloheximide on cAMP-induced PACAP, but not PR, mRNA levels. To determine whether PR synthesis is prerequisite for PACAP expression, we examined the effect of ZK98299, a specific PR antagonist, on cAMP-induced PACAP mRNA expression. This compound blocked cAMP-induced PACAP mRNA expression in a dose-dependent manner, indicating that PR activation is required for PACAP gene expression in granulosa cells. We then compared cellular localization and hormonal regulation of ovarian PR and PACAP gene expression in immature rats treated with gonadotropins as well as in adult rats during the preovulatory period by using in situ hybridization and semiquantitative RT-PCR assays. Results show that both PR and PACAP mRNAs are induced in granulosa cells of preovulatory follicles by human CG, but that the PR gene is expressed before the PACAP gene. Taken together, these results demonstrate that PRs mediate the LH-induced PACAP gene expression in rat granulosa cells.
Collapse
Affiliation(s)
- C Ko
- Department of Physiology, University of Kentucky, Lexington 40536-0084, USA
| | | | | |
Collapse
|
32
|
Burleigh DW, Williams RF, Gordon K, Hsiu JG, Hodgen GD. Screening for antiproliferative actions of mifepristone. Differential endometrial responses of primates versus rats. Contraception 1998; 58:45-50. [PMID: 9743896 DOI: 10.1016/s0010-7824(98)00060-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This laboratory has previously shown the capability of the antiprogestin, mifepristone, to noncompetitively inhibit estrogen-induced endometrial proliferation in nonhuman primates. In the following study, use of the rat uterine weight bioassay was compared against a primate (Macaca fascicularis) uterine bioassay to identify the noncompetitive/antiproliferative effects of mifepristone. These uterine bioassays were contrasted for reasons of identifying a comparative laboratory rodent model that could substitute for the need to use primate models in the screening of potential antiprogestins, thereby saving time, cost, and primate resources. Results of the primate experiment showed that mifepristone decreased endometrial proliferation in a dose-dependent manner; importantly, this decrease occurred in the presence of sustained physiologic serum 17 beta-estradiol (E2) levels. However, in the rat model, results showed that mifepristone altered uterine wet weight and blotted weight values only in those animals receiving pharmacological doses of E2 (p < 0.05). Based on the results summarized herein, use of this rat uterine weight bioassay as a substitute for primate models is not recommended for screening and identification of "interesting" antiprogestins. Apparently, the endometrial noncompetitive antiestrogenic/antiproliferative effects of mifepristone, observed repeatedly in these laboratory primates, do not operate in the rat uterine tissue.
Collapse
Affiliation(s)
- D W Burleigh
- Jones Institute for Reproductive Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical school. Norfolk, Virginia 23507, USA
| | | | | | | | | |
Collapse
|
33
|
Halikias I, Lytras A, Syriou V, Tolis G. Combined oral contraceptives and gonadotropin releasing hormone agonistic analogs in polycystic ovary syndrome: clinical and experimental studies. EUR J CONTRACEP REPR 1997; 2:213-24. [PMID: 9678076 DOI: 10.3109/13625189709165297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Polycystic ovary syndrome is a common endocrine disorder, presenting with menstrual irregularities, hirsutism, obesity, infertility and abnormal ovarian morphology. In addition, polycystic ovary syndrome is associated with a self-perpetuating imbalance involving the endocrine system and metabolic pathways, in which carbohydrates, lipids and growth factors are involved. Because of its chronicity, it is considered to be a substantial risk factor for atherogenesis and hormone-dependent neoplasia. The etiology and pathophysiology of the syndrome remain elusive. However, during the last decade, several clues have emerged from human and animal studies that may have significant repercussions in the treatment of polycystic ovary syndrome. Therapeutic maneuvers should be directed towards the dominant abnormalities present in individual patients with polycystic ovary syndrome. Gonadotropin releasing hormone (GnRH) agonists can directly affect the gonadotropin generator and secondary downstream derangements, whereas combined oral contraceptives (COCs) can modify hypothalamic as well as peripheral abnormalities. In view of the fact that GnRH agonistic analogs (GnRH-a) will induce hypoestrogenemia and its sequelae, the add-back strategy of estrogenic supplementation is recommended for preventive reasons and, as it transpires from some studies, for enhancement of GnRH-a effectiveness.
Collapse
Affiliation(s)
- I Halikias
- Athens University, Division of Endocrinology, Hippokration Hospital, Greece
| | | | | | | |
Collapse
|
34
|
Heikinheimo O, Hsiu JG, Gordon K, Kim S, Williams RF, Gibbons WE, Hodgen GD. Endometrial effects of RU486 in primates--antiproliferative action despite signs of estrogen action and increased cyclin-B expression. J Steroid Biochem Mol Biol 1996; 59:179-90. [PMID: 9010333 DOI: 10.1016/s0960-0760(96)00113-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Continuous antiprogestin administration to hormone replaced, castrate monkeys inhibits estrogen-induced endometrial proliferation through mechanisms which remains unclear. To elucidate the molecular mechanisms of RU486-induced endometrial suppression, we treated six intact female cynomolgus monkeys on cycle days 2-22 sequentially with placebo, RU486 (1 mg/kg/day) and levonorgestrel (LNG) (2 microg/kg/day) intramuscularly (i.m.), with uterine wedge sections and endometrial biopsies collected on day 22 of each cycle. The uterine sections were evaluated for morphology, mitosis and proliferating cell nuclear antigen (PCNA) immunohistochemistry. Changes in the mRNA levels of ER, PR, cyclin-B and tumour suppressor gene p21 were assessed using co-amplification with beta-actin by reverse transcriptase-polymerase chain reaction (RT-PCR). Administration of RU486 uniformly resulted in characteristic suppression of endometrium with few mitosis, dense stroma and simple glands, whereas the effects of LNG were less uniform. Following RU486 administration, the levels of endometrial ER and PR mRNA were comparable to proliferative phase endometrium, and significantly higher than those seen in the secretory endometrium, indicating that some of the biological actions of E2 were not inhibited during RU486 treatment. Despite scarce mitosis, PCNA was readily detectable in all samples. Curiously, in comparison to secretory phase controls, the levels of cyclin-B, but not p21, mRNA were markedly increased following RU486. The effects of LNG on the levels of these mRNA species varied, with mean levels falling between those of the secretory phase controls, and RU486-treated specimens. The increase in cyclin-B mRNA and lack of mitosis suggests that anti-proliferative actions of RU486 in the primate endometrium might be associated with a cell-cycle block at the G2-M interphase. Whether mechanisms similar to these are associated with the beneficial clinical effects of RU486 seen in the treatment of various hormone dependent maladies remains to be determined.
Collapse
Affiliation(s)
- O Heikinheimo
- The Jones Institute for Reproductive Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA 23517, USA
| | | | | | | | | | | | | |
Collapse
|