Next Article in Journal
High Expression of MRE11A Is Associated with Shorter Survival and a Higher Risk of Death in CRC Patients
Next Article in Special Issue
A Circular RNA Derived from the Pumilio 1 Gene Could Regulate PTEN in Human Cumulus Cells
Previous Article in Journal
Chromosome-Level Genome Assembly Provides Insights into the Evolution of the Special Morphology and Behaviour of Lepturacanthus savala
Previous Article in Special Issue
Vitrification with Dimethyl Sulfoxide Induces Transcriptomic Alteration of Gene and Transposable Element Expression in Immature Human Oocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Variants of Gonadotropins and Their Receptors Could Influence Controlled Ovarian Stimulation: IVF Data from a Prospective Multicenter Study

1
Department of Public Health, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
2
Merck Serono S.p.A., 00176 Rome, Italy
3
IRCCS San Raffaele Hospital, 00163 Milan, Italy
4
Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
5
Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 80131 Naples, Italy
6
Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
7
Azienda Ospedaliera Universitaria Federico II di Napoli, 80131 Naples, Italy
8
Faculty of Medicine, Department of Surgery and Cancer, London W12 0NN, UK
9
Institute of Biomedicine, University of Turku, 20014 Turku, Finland
10
Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
11
Fertility Clinic at Skive Regional Hospital, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
*
Author to whom correspondence should be addressed.
Genes 2023, 14(6), 1269; https://doi.org/10.3390/genes14061269
Submission received: 3 March 2023 / Revised: 19 April 2023 / Accepted: 22 May 2023 / Published: 15 June 2023
(This article belongs to the Special Issue Genetics and Genomics of Female Reproduction)

Abstract

:
Background: Specific polymorphisms might influence controlled ovarian stimulation in women undergoing assisted reproductive technologies (ARTs). Data regarding possible interactions of these polymorphisms are still scanty. The aim of this analysis was to evaluate the effect of polymorphisms of gonadotropins and their receptors in women undergoing ART. Methods: A total of 94 normogonadotropic patients from three public ART units were enrolled. Patients underwent a gonadotropin releasing hormone (GnRH) long down-regulation protocol with a starting dose of 150 IU of recombinant follicular stimulating hormone (FSH) daily. Eight polymorphisms were genotyped. Results: A total of 94 women (mean age 30.71 ± 2.61) were recruited. Fewer fertilized and mature oocytes were retrieved in homozygous carriers of luteinizing hormone/choriogonadotropin receptor (LHCGR) 291 (T/T) than in heterozygous C/T carriers (p = 0.035 and p = 0.05, respectively). In FSH receptor (FSHR) rs6165 and FSHR rs6166 carriers, the ratio between total gonadotropin consumption and number of oocytes retrieved differed significantly among three genotypes (p = 0.050), and the ratio was lower in homozygous A/A carriers than in homozygous G/G and heterozygous carriers. Women who co-expressed allele G in FSHR-29 rs1394205 and FSHR rs6166 and allele C LHCGR 291 rs12470652 are characterized by an increased ratio between total FSH dosage and number of oocytes collected after ovarian stimulation (risk ratio: 5.44, CI 95%: 3.18–7.71, p < 0.001). Conclusions: Our study demonstrated that specific polymorphisms affect the response to ovarian stimulation. Despite this finding, more robust studies are required to establish the clinical utility of genotype analysis before ovarian stimulation.

1. Introduction

Several studies demonstrated that individual gonadotropins and the genotype of their receptors could affect ovarian stimulation (OS) [1,2,3]. Among these receptors, the FSHR polymorphism located in amino acid position 680 of exon 10 (FSHR N680S; rs6166) has been the most widely investigated. In fact, previous studies showed that FSHR A680G G homozygous women need a higher amount of exogenous gonadotropin during OS [4,5,6] and had higher basal FSH levels than women carrying other haplotypes [2,7,8]. In addition, the polymorphism in the 5′ untranslated region of FSHR (rs1394205) seems to exert an influence on ovarian response to exogenous gonadotropin. In detail, FSHR-29 (rs1394205) A homozygous women had fewer oocytes retrieved and a lower clinical pregnancy rate than did GG homozygous women [9,10]. Similarly, fewer mature (MII) oocytes were retrieved in carriers of the FSHR-29 (rs1394205) AA genotype than in carriers of the GG genotype [10,11]. However, a higher live birth rate was reported in FSHR-29 (rs1394205) A homozygous women than in women with other haplotypes in a retrospective analysis of 603 women undergoing ART [12].
An increased exogenous FSH consumption was observed in carriers of the luteinizing B subunit (LH-B) W8R/I15T rs1800447 genetic variant of the luteinizing hormone (LH) β subunit [13] (Alviggi et al., 2013). This polymorphism presents a reduced half-life in vivo in contrast with the wild-type form [14] and is widely expressed in Northern Europe and in Australian aboriginal populations [15].
In another investigation involving 384 women undergoing ART, the pregnancy rate was higher in carriers of the LHCGR 312G polymorphism than in A312 carriers (LHCGR A312G rs2293275) [16]. Furthermore, LHCGR 312 G homozygotes required a higher amount of gonadotropins for follicular recruitment than did A homozygotes. The LHCGR A312G rs2293275 polymorphism was also implicated in polycystic ovarian syndrome development (PCOS), with higher risk of PCOS in AA homozygotes [17]. When combined, gonadotropin receptor polymorphisms could also modulate OS [18]. For example, the pregnancy rate was higher in women homozygous for G than in women homozygous for A in both the FSHR A680G and LHCGR A312G polymorphisms [16]. Furthermore, in a retrospective study, homozygotes of both AA FSHR-29 and AA FSHR A680G had an increased risk of impaired ovarian response after OS [19]. Moreover, carriers of the FSH B subunit (FSHB)-211 GT plus FSHR 2039 AA genotype had significantly lower day 3 FSH levels versus carriers of the FSHB-211 GG/FSHR 2039 GG genotype [20]. However, most of the studies that investigated the role of polymorphisms in OS have some limitations. For instance, most studies were retrospective and were widely heterogeneous in terms of the ART protocols used and patients recruited [4,6,9,10,12]. Moreover, little is known about the interactions, if any, among these genetic variants.
The aim this multicenter prospective study was to assess the effect of multiple genetic variants of gonadotropins and their receptors in women undergoing OS with the GnRH agonist long down-regulation protocol and a fixed FSH starting dose.

2. Materials and Methods

2.1. Study Population

Caucasian women from three ART centers (University of Naples Federico II, IRCCS San Raffaele Hospital, Milan, and Aarhus University, The Fertility Clinic, Skive Regional Hospital, Denmark) were enrolled in this study. We recruited women with the following characteristics: age from 20–35 years; body mass index (BMI) from 20–27 kg/m2; basal FSH ≤ 10 IU/L; indication for ART; presence of both normal ovaries; normal ovarian reserve according to Patient-Oriented Strategies Encompassing IndividualizeD Oocyte Number (POSEIDON) [21,22] (antral follicle count > 5). Women with the following characteristics were excluded from the analysis: abnormal uterine cavity; endocrine, genetic, or systemic inflammatory-immunological disease; presence of PCOS according to the Rotterdam criteria; and diagnosis of endometriosis. Moreover, we excluded women with a history of more than two previous ART procedures that had a good ovarian response or a previous stimulation cycle that had been suspended due to an inadequate ovarian response or in whom < 4 oocytes had been obtained.

2.2. Stimulation Protocol

The GnRH-a long down-regulation protocol was adopted using buserelin acetate as follows: 0.5 mg s.c. daily from luteal phase for 12–14 days. Fourteen days later, transvaginal ultrasound and biochemical evaluations were performed to verify the pituitary suppression. Only women with a serum estradiol (E2) level ≤ 40 pg/mL, an endometrial thickness ≤ 5 mm, and arrested follicular development were admitted for controlled OS. Women with delayed pituitary suppression (including subjects who developed ovarian cysts after GnRH-a administration) were not included. A fixed starting dosage of 150 IU of recombinant human FSH (r-hFSH) was adopted for all participants (Gonal-F®; Merck Serono S.p.A, Rome, Italy) for at least four days. E2 was measured on day five of stimulation. On that day, the daily dose of gonadotropin was modified when E2 concentration > 180 pg/mL. Only in these cases was a daily dose of r-hFSH of 112.5 IU adopted according to standard clinical practice. Follicular growth was evaluated on day 8 of stimulation by transvaginal ultrasound. Only patients who had at least 6 follicles ranging between 6 and 10 mm in diameter, but no follicle with a mean diameter > 10 mm, received an increased dosage. Specifically, the r-hFSH dose was increased by 150 IU per day, for a cumulative daily dose of 300 IU. Women who had their daily dose of gonadotropin reduced on the fifth day of stimulation and who required another increase on day 8 were excluded from the study. Women who required “coasting” to reduce the risk of OHSS were also excluded. E2 serum levels were measured on days 1, 5, and 8 of stimulation and on the day of administration of human CG (hCG). According to clinical practice, the ovulatory dose of 10,000 IU of hCG or 250 mcg of recombinant hCG was prescribed in women who had three follicles with a mean diameter of at least 17 mm. Oocytes were collected by the transvaginal ultrasound approach 34–36 h after hCG injection. Embryo transfer and luteal phase support were conducted as previously mentioned [13,23].

2.3. Polymorphism Analyses

Venous blood samples were clotted and centrifuged at 400× g for 10 min. Serum was divided into a maximum of four aliquots and frozen. Pellets were also separated into four aliquots and stocked at −80 °C.
We used an amplicon-based next-generation sequencing assay (TruSeq Custom Amplicon v1.5, Illumina, San Diego, CA, USA) to genotype the following SNPs: (i) FSHR 307 rs6165; (ii) FSHR 680 rs6166; (iii) FSHR-29 rs1394205; (iv) LHCGR intronic rs4073366; (v) LHCGR rs2293275; (vi) FSHB 2623 rs6169; (vii) v-LH rs1800447. The probes for custom panels targeting the LHB, FSHR, LHCGR, and FSHB genes were designed with Design Studio (Illumina, San Diego, CA, USA) and consisted of 44 amplicons with an average size of 250 bp and a cumulative targeted region of 5.7 kb. Polymorphisms were avoided in the primer design. The pooled libraries were paired-end sequenced (2 × 151 bases) with V2 chemistry on a MiSeq instrument. The PhiX control library (5%) was spiked in each run to estimate the sequencing error rate. Polymorphisms were analyzed with VariantStudio (Illumina, San Diego, CA, USA). The HCGR 291 rs12470652 polymorphism was genotyped with bidirectional Sanger sequencing by using a BigDye Terminator v3.1 Cycle Sequencing Kit (Applied Biosystems, Foster City, CA, USA; Eurofins Genomics, Ebersberg, Germany).

2.4. Primary and Secondary Endpoints

The primary endpoint was the ratio between cumulative r-hFSH dosage and mean number of oocytes obtained. Secondary endpoints were: E2 levels on the day of hCG, cumulative dosage of r-hFSH, number of preovulatory follicles, number of oocytes retrieved, the number of mature oocytes retrieved (MII oocytes), number of fertilized oocytes, number of embryos transferred, implantation rate, pregnancy rate per cycle, pregnancy rate per transfer, clinical pregnancy rate per started cycle (presence of embryos with heartbeat), clinical pregnancy rate per transfer (presence of embryos with heartbeat).

2.5. Statistical Analysis

Genotype distributions of the SNPs evaluated were collected by direct computing, and linkage disequilibrium was evaluated using SNPStat. Hardy–Weinberg equilibrium was measured [24]. The chi-square test was conducted to compare the frequencies of SNPs in the enrolled patients to those of the general population using widely available databases. For each polymorphism evaluated, the codominance model was considered, supposing that the contribution of both alleles was visible in the phenotype.
The Kolmogorov–Smirnov test was carried out to evaluate the distribution of variables. Differences in continuous variables among groups were evaluated with an ANOVA univariate model for normally distributed variables, and the Kruskal–Wallis or Mann–Whitney test for not normally distributed variables. The Dunnet test was used as a post hoc test. Spearman’s rho regression was performed for bivariate correlation. Statistical analysis was carried out using the Statistical Package for the Social Sciences software for Macintosh (SPSS Inc version 20.0, Chicago, IL, USA).

3. Results

Ninety-four women with a mean age of 30.71 ± 2.61 years and a mean BMI of 23.9 ± 2.35 kg/m2 undergoing ART cycles were enrolled. Fifty-three was recruited from Federico II University, twenty-nine from Aarhus University, and twelve from the San Raffaele Institution. Hardy–Weinberg equilibrium was verified and the allele frequencies did not differ between the study group and the general population (Table 1). Baseline FSH serum levels were 6.73 ± 1.98 IU/L. All patients underwent r-HFSH 150 IU daily, according to the study protocol and the mean total r-hFSH dosage prescribed was 1725.33 ± 520.15 IU. Stimulation had an average duration of 11.24 ± 1.69 days (Table 2). Only two cycles (2.4%) were interrupted due to risk of OHSS, whereas no cycles were interrupted for non-response to OS. After OS, mean E2 serum levels were 1655.43 ± 895.59 pg/mL, and women underwent IVF in 28.7% of cases (27 women) and intracytoplasmatic sperm injection in 71.2% of cases (67 women) (Table 2). Forty pregnancies per cycle (42.5%) were obtained according to β-hCG measurement and 32 (34.04%) of these were diagnosed by ultrasound assessment (Table 2).

3.1. FSHR 307 (rs6165) and FSHR 680 (rs6166)

The total number of oocytes retrieved did not differ between carriers of FSHR rs6165 (p = 0.510) and carriers of FSHR rs6166 (p = 0.170). The ratio between total gonadotropin consumption and number of oocytes retrieved differed almost significantly among three genotypes (p = 0.050). As shown in Table 3 and Table 4, the following features did not differ between the two SNPs: total r-hFSH dosage used, the ratio between fertilized and inseminated oocytes, E2 levels on the day of hCG, cumulative dosage of r-hFSH, number of preovulatory follicles, mature oocytes retrieved (MII oocytes), number of fertilized oocytes, number of embryos transferred, implantation rate, pregnancy rate per cycle, pregnancy rate per transfer, clinical pregnancy rate for started cycle, clinical pregnancy rate per transfer.

3.2. FSHR-29 (rs1394205)

Treatment outcomes did not differ among models generated according to genotype frequencies (Table 5).

3.3. LHCGR 291 (rs12470652)

LHCGR heterozygous women had higher E2 levels on the day of hCG administration (p = 0.005) than wild-type carriers (Table 6). Similarly, the numbers of total retrieved oocytes (p = 0.035), MII (p = 0.002), inseminated oocytes (p = 0.001), fertilized oocytes (p = 0.001), and cryopreserved embryos (p = 0.001) were higher in heterozygous women than in wild-type carriers (Table 6). No significant differences were found among the other variables (Table 6).

3.4. LHCGR Intronic (rs4073366), LHCGR 312 (rs2293275), FSHB 2623 (rs6169), and v-LH (rs1800447)

The parameters evaluated did not differ significantly among LHCGR rs4073366, LHCGR rs2293275, FSHB rs10835638, and v-LH rs1800447 carriers (Supplemental Tables S1–S4).

3.5. Multivariate Analysis

In multivariate analysis, the copresence of allele G of FSHR-29 rs1394205 and allele C of LHCGR 291 rs12470652 was related to an increased ratio between cumulative r-hFSH dose and total number of oocytes retrieved (5.47, CI 95%: 3.13–7.81, p < 0.001). Furthermore, the copresence of allele G of both FSHR-29 rs1394205 and FSHR rs6166 and allele C LHCGR 291 rs12470652 was related to an increased ratio between cumulative FSH dose and total number of oocytes retrieved (5.44, CI: 3.18–7.71, p < 0.001).

4. Discussion

Our data support the concept that ovarian response to exogenous gonadotropin could be influenced by specific genetic traits. In detail, we observed that two common FSHR (FSHR rs6165 and rs6166) SNPs and one LHCGR (rs12470652) SNP might affect OS outcomes in terms of number of oocytes retrieved, cumulative r-hFSH dosage, and oocyte ratio. Furthermore, our multivariate analysis suggests that the interaction of specific genetic traits could also influence ovarian sensitivity to exogenous gonadotropin. To our knowledge, this is first time that LHCGR 291 (rs12470652) has been implicated in response to OS.
In contrast to Ackrekar et al. [9], we did not find any association between FSHR-29 polymorphism and OS response. This discordance might be due to the differences in study design, inclusion criteria, and protocols between the two studies. In fact, we conducted a prospective analysis adopting very strict inclusion criteria. In addition, we used a standardized OS protocol in which recombinant gonadotropin was used at a fixed starting dose.
Our finding that GG carriers are resistant to both endogenous and exogenous FSH is consistent with previous studies [2,6,7,8]. This resistance seems to be related to specific molecular characteristics. In fact, in vitro studies showed that women with a GG haplotype (rs6166) genotype have reduced sensitivity compared with the AA haplotype [25,26]. Unlike our previous studies [13,27], we did not detect an association between OS and the LH β polymorphism rs1800447. This incongruity could reflect the absence of homozygous carriers of this polymorphic trait in the present study and the reduced number of women recruited, namely, 94 patients versus 200 cases reported in 2013 [13]. It could also reflect the different designs of the two studies.
Our study has several strengths. Firstly, it was a multicenter prospective analysis conducted using detailed inclusion/exclusion criteria. Indeed, we enrolled only patients with an adequate ovarian reserve using a fixed starting daily dose who did not require “coasting” for hyper-response or multiple dosage adjustments during OS. We adopted these inclusion criteria to avoid any misinterpretation of data. Indeed, some authors demonstrated that elevated FSH dosage, as well as dose adjustment during OS, could mitigate the effect of genotype on ART [28]. Furthermore, we also carried out a multivariate analysis to establish whether interactions among different polymorphisms could influence ovarian response. Most previous studies focused on a single polymorphism [8,9,28,29], and only the most recent studies considered no more than two polymorphisms together [16,19]. Furthermore, ours is the first study to evaluate the LHCGR intronic (rs4073366) and FSHB 2623 (rs10835638) polymorphisms in ART.
The most important limitation of our investigation resides in the relatively small number of women enrolled. In addition, we were not able to follow patients up until childbirth, although we do provide data on the ongoing pregnancy rate. Like other studies, we did not find a significant association between gonadotropins and their polymorphisms and pregnancy outcomes [13,29,30]. However, this does not necessarily mean that gonadotropins did not affect OS. In fact, we believe that the number of oocytes retrieved and the amount of gonadotropin consumed during OS are more appropriate measures than ART births of the influence of gonadotropins and their receptor genetic variants in an in vitro fertilization setting. Indeed, ART births are influenced by such other factors as embryo quality and maternal age and factors that occur during pregnancy, namely, intrauterine growth restriction, that are poorly related to genetic variants affecting gonadotropins and their receptors. Furthermore, we enrolled women who carried out a long analogue protocol and consequently we cannot provide data about antagonist protocols that are currently widely adopted. Finally, another limitation is that we limited our analysis to young, good prognosis patients, thus our data could not be generalized to advanced age women that often require medically assisted reproduction.

5. Conclusions

In conclusion, our analysis supports the concept that distinct polymorphisms could modulate the response to OS. Moreover, we demonstrate that simultaneous analysis of multiple polymorphisms provides useful information about the response to controlled ovarian response. Our data need to be confirmed by further investigations, especially for LHCGR intronic (s4073366) and FSHB 2623 (rs10835638) polymorphisms. Despite our findings, more robust studies are required to establish the clinical utility of genotype analysis before ovarian stimulation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14061269/s1, Table S1: Treatment outcomes. Patients stratified according to the rs LHCGR intronic polymorphism (rs4073366); Table S2: Treatment outcomes in patients stratified according to the LHCGR 312 (rs2293275) polymorphism; Table S3: Treatment outcomes in patients stratified according to the rs FSHB 2623 (rs6169) polymorphism; Table S4: Treatment outcomes in patients stratified according to the LHB (rs1800447) polymorphism.

Author Contributions

C.A., S.L., T.D., P.H. and A.C.: Conceptualization; Writing—original draft; Writing—review and editing; Supervision. S.A., V.S.V., M.R.C., P.D.R. and I.S.: Writing—review and editing; Resources. I.H. and J.-P.P.: Writing—original draft; Writing—review and editing; Methodology. D.S.: Formal analysis; Writing—original draft. Software; E.P.: Conceptualization; Writing—review and editing; Supervision. C.A.: Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This is an investigator-sponsored study supported by an unrestricted grant from Merck Serono S.p.A, Italy.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board and Ethics Committee of the University of Naples Federico II, Naples, Italy.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are unavailable due to privacy restrictions.

Acknowledgments

The authors thank Jean Ann Gilder (Scientific Communication srl., Naples, Italy) for revising and editing the manuscript.

Conflicts of Interest

A.C. and C.A. report personal fees honoraria from Merck outside the submitted work. P.H. reports unrestricted research grants from MSD, Merck, and Ferring as well as personal fees from honoraria for lectures from MSD, Merck, and Finox, outside the submitted work. E.P. reports grants, personal fees, and non-financial support from Merck, grants, personal fees, and non-financial support from MSD, grants, personal fees, and non-financial support from Ferring, grants from Finox, and grants and non-financial support from IBSA outside the submitted work; D.S., G.D., V.S.V., I.S., I.H., P.D.R., S.A. and J.P. have nothing to disclose.

References

  1. Altmäe, S.; Hovatta, O.; Stavreus-Evers, A.; Salumets, A. Genetic Predictors of Controlled Ovarian Hyperstimulation: Where Do We Stand Today? Hum. Reprod. Update 2011, 17, 813–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Alviggi, C.; Conforti, A.; Santi, D.; Esteves, S.C.; Andersen, C.Y.; Humaidan, P.; Chiodini, P.; De Placido, G.; Simoni, M. Clinical Relevance of Genetic Variants of Gonadotrophins and Their Receptors in Controlled Ovarian Stimulation: A Systematic Review and Meta-Analysis. Hum. Reprod. Update 2018, 24, 599–614. [Google Scholar] [CrossRef]
  3. Conforti, A.; Tüttelmann, F.; Alviggi, C.; Behre, H.M.; Fischer, R.; Hu, L.; Polyzos, N.P.; Chuderland, D.; Rama Raju, G.A.; D’Hooghe, T.; et al. Effect of Genetic Variants of Gonadotropins and Their Receptors on Ovarian Stimulation Outcomes: A Delphi Consensus. Front. Endocrinol. 2021, 12, 797365. [Google Scholar] [CrossRef] [PubMed]
  4. Sudo, S.; Kudo, M.; Wada, S.; Sato, O.; Hsueh, A.J.W.; Fujimoto, S. Genetic and Functional Analyses of Polymorphisms in the Human FSH Receptor Gene. Mol. Hum. Reprod. 2002, 8, 893–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Behre, H.M.; Greb, R.R.; Mempel, A.; Sonntag, B.; Kiesel, L.; Kaltwasser, P.; Seliger, E.; Röpke, F.; Gromoll, J.; Nieschlag, E.; et al. Significance of a Common Single Nucleotide Polymorphism in Exon 10 of the Follicle-Stimulating Hormone (FSH) Receptor Gene for the Ovarian Response to FSH: A Pharmacogenetic Approach to Controlled Ovarian Hyperstimulation. Pharm. Genom. 2005, 15, 451–456. [Google Scholar] [CrossRef] [PubMed]
  6. Alviggi, C.; Conforti, A.; Caprio, F.; Gizzo, S.; Noventa, M.; Strina, I.; Pagano, T.; De Rosa, P.; Carbone, F.; Colacurci, N.; et al. In Estimated Good Prognosis Patients Could Unexpected “Hyporesponse” to Controlled Ovarian Stimulation Be Related to Genetic Polymorphisms of FSH Receptor? Reprod. Sci. 2016, 23, 1103–1108. [Google Scholar] [CrossRef]
  7. Perez Mayorga, M.; Gromoll, J.; Behre, H.M.; Gassner, C.; Nieschlag, E.; Simoni, M. Ovarian Response to Follicle-Stimulating Hormone (FSH) Stimulation Depends on the FSH Receptor Genotype. J. Clin. Endocrinol. Metab. 2000, 85, 3365–3369. [Google Scholar] [CrossRef]
  8. Yan, Y.; Gong, Z.; Zhang, L.; Li, Y.; Li, X.; Zhu, L.; Sun, L. Association of Follicle-Stimulating Hormone Receptor Polymorphisms with Ovarian Response in Chinese Women: A Prospective Clinical Study. PLoS ONE 2013, 8, e78138. [Google Scholar] [CrossRef] [Green Version]
  9. Achrekar, S.K.; Modi, D.N.; Desai, S.K.; Mangoli, V.S.; Mangoli, R.V.; Mahale, S.D. Poor Ovarian Response to Gonadotrophin Stimulation Is Associated with FSH Receptor Polymorphism. Reprod. Biomed. Online 2009, 18, 509–515. [Google Scholar] [CrossRef]
  10. Polyzos, N.P.; Neves, A.R.; Drakopoulos, P.; Spits, C.; Alvaro Mercadal, B.; Garcia, S.; Ma, P.Q.M.; Le, L.H.; Ho, M.T.; Mertens, J.; et al. The Effect of Polymorphisms in FSHR and FSHB Genes on Ovarian Response: A Prospective Multicenter Multinational Study in Europe and Asia. Hum. Reprod. 2021, 36, 1711–1721. [Google Scholar] [CrossRef]
  11. Desai, S.S.; Achrekar, S.K.; Pathak, B.R.; Desai, S.K.; Mangoli, V.S.; Mangoli, R.V.; Mahale, S.D. Follicle-Stimulating Hormone Receptor Polymorphism (G-29A) Is Associated with Altered Level of Receptor Expression in Granulosa Cells. J. Clin. Endocrinol. Metab. 2011, 96, 2805–2812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Tohlob, D.; Abo Hashem, E.; Ghareeb, N.; Ghanem, M.; Elfarahaty, R.; Byers, H.; Pemberton, P.; Roberts, S.A.; Newman, W.G.; Mohiyiddeen, L. Association of a Promoter Polymorphism in FSHR with Ovarian Reserve and Response to Ovarian Stimulation in Women Undergoing Assisted Reproductive Treatment. Reprod. Biomed. Online 2016, 33, 391–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Alviggi, C.; Pettersson, K.; Longobardi, S.; Andersen, C.Y.; Conforti, A.; De Rosa, P.; Clarizia, R.; Strina, I.; Mollo, A.; De Placido, G.; et al. A Common Polymorphic Allele of the LH Beta-Subunit Gene Is Associated with Higher Exogenous FSH Consumption during Controlled Ovarian Stimulation for Assisted Reproductive Technology. Reprod. Biol. Endocrinol. 2013, 11, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Haavisto, A.M.; Pettersson, K.; Bergendahl, M.; Virkamäki, A.; Huhtaniemi, I. Occurrence and Biological Properties of a Common Genetic Variant of Luteinizing Hormone. J. Clin. Endocrinol. Metab. 1995, 80, 1257–1263. [Google Scholar] [CrossRef] [PubMed]
  15. Nilsson, C.; Jiang, M.; Pettersson, K.; Iitiä, A.; Mäkelä, M.; Simonsen, H.; Easteal, S.; Herrera, R.J.; Huhtaniemi, I. Determination of a Common Genetic Variant of Luteinizing Hormone Using DNA Hybridization and Immunoassays. Clin. Endocrinol. 1998, 49, 369–376. [Google Scholar] [CrossRef]
  16. Lindgren, I.; Bååth, M.; Uvebrant, K.; Dejmek, A.; Kjaer, L.; Henic, E.; Bungum, M.; Bungum, L.; Cilio, C.; Leijonhufvud, I.; et al. Combined Assessment of Polymorphisms in the LHCGR and FSHR Genes Predict Chance of Pregnancy after in Vitro Fertilization. Hum. Reprod. 2016, 31, 672–683. [Google Scholar] [CrossRef] [Green Version]
  17. Capalbo, A.; Sagnella, F.; Apa, R.; Fulghesu, A.M.; Lanzone, A.; Morciano, A.; Farcomeni, A.; Gangale, M.F.; Moro, F.; Martinez, D.; et al. The 312N Variant of the Luteinizing Hormone/Choriogonadotropin Receptor Gene (LHCGR) Confers up to 2·7-Fold Increased Risk of Polycystic Ovary Syndrome in a Sardinian Population. Clin. Endocrinol. 2012, 77, 113–119. [Google Scholar] [CrossRef]
  18. Conforti, A.; Alfano, S.; De Rosa, P.; Alviggi, C.; De Placido, G. The Role of Gonadotropin Polymorphisms and Their Receptors in Assisted Reproductive Technologies and Controlled Ovarian Stimulation: A Prospective Observational Study. Ital. J. Gynaecol. Obstet. 2017, 29, 15–21. [Google Scholar]
  19. Desai, S.S.; Achrekar, S.K.; Paranjape, S.R.; Desai, S.K.; Mangoli, V.S.; Mahale, S.D. Association of Allelic Combinations of FSHR Gene Polymorphisms with Ovarian Response. Reprod. Biomed. Online 2013, 27, 400–406. [Google Scholar] [CrossRef] [Green Version]
  20. La Marca, A.; Papaleo, E.; Alviggi, C.; Ruvolo, G.; De Placido, G.; Candiani, M.; Cittadini, E.; De Michele, F.; Moriondo, V.; Catellani, V.; et al. The Combination of Genetic Variants of the FSHB and FSHR Genes Affects Serum FSH in Women of Reproductive Age. Hum. Reprod. 2013, 28, 1369–1374. [Google Scholar] [CrossRef]
  21. Esteves, S.C.; Conforti, A.; Sunkara, S.K.; Carbone, L.; Picarelli, S.; Vaiarelli, A.; Cimadomo, D.; Rienzi, L.; Ubaldi, F.M.; Zullo, F.; et al. Improving Reporting of Clinical Studies Using the POSEIDON Criteria: POSORT Guidelines. Front. Endocrinol. 2021, 12, 587051. [Google Scholar] [CrossRef] [PubMed]
  22. Esteves, S.C.; Yarali, H.; Vuong, L.N.; Conforti, A.; Humaidan, P.; Alviggi, C. POSEIDON Groups and Their Distinct Reproductive Outcomes: Effectiveness and Cost-Effectiveness Insights from Real-World Data Research. Best Pract. Res. Clin. Obstet. Gynaecol. 2022, 85, 159–187. [Google Scholar] [CrossRef] [PubMed]
  23. Alviggi, C.; Guadagni, R.; Conforti, A.; Coppola, G.; Picarelli, S.; De Rosa, P.; Vallone, R.; Strina, I.; Pagano, T.; Mollo, A.; et al. Association between Intrafollicular Concentration of Benzene and Outcome of Controlled Ovarian Stimulation in IVF/ICSI Cycles: A Pilot Study. J. Ovarian Res. 2014, 7, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Solé, X.; Guinó, E.; Valls, J.; Iniesta, R.; Moreno, V. SNPStats: A Web Tool for the Analysis of Association Studies. Bioinformatics 2006, 22, 1928–1929. [Google Scholar] [CrossRef] [Green Version]
  25. Casarini, L.; Moriondo, V.; Marino, M.; Adversi, F.; Capodanno, F.; Grisolia, C.; La Marca, A.; La Sala, G.B.; Simoni, M. FSHR Polymorphism p.N680S Mediates Different Responses to FSH in Vitro. Mol. Cell. Endocrinol. 2014, 393, 83–91. [Google Scholar] [CrossRef]
  26. Casarini, L.; Santi, D.; Marino, M. Impact of Gene Polymorphisms of Gonadotropins and Their Receptors on Human Reproductive Success. Reproduction 2015, 150, R175–R184. [Google Scholar] [CrossRef] [Green Version]
  27. Alviggi, C.; Clarizia, R.; Pettersson, K.; Mollo, A.; Humaidan, P.; Strina, I.; Coppola, M.; Ranieri, A.; D’Uva, M.; De Placido, G. Suboptimal Response to GnRHa Long Protocol Is Associated with a Common LH Polymorphism. Reprod. Biomed. Online 2011, 22, 67–72. [Google Scholar] [CrossRef]
  28. Genro, V.K.; Matte, U.; De Conto, E.; Cunha-Filho, J.S.; Fanchin, R. Frequent Polymorphisms of FSH Receptor Do Not Influence Antral Follicle Responsiveness to Follicle-Stimulating Hormone Administration as Assessed by the Follicular Output RaTe (FORT). J. Assist. Reprod. Genet. 2012, 29, 657–663. [Google Scholar] [CrossRef] [Green Version]
  29. Huang, X.; Li, L.; Hong, L.; Zhou, W.; Shi, H.; Zhang, H.; Zhang, Z.; Sun, X.; Du, J. The Ser680Asn Polymorphism in the Follicle-Stimulating Hormone Receptor Gene Is Associated with the Ovarian Response in Controlled Ovarian Hyperstimulation. Clin. Endocrinol. 2015, 82, 577–583. [Google Scholar] [CrossRef]
  30. Mohiyiddeen, L.; Newman, W.G.; Cerra, C.; McBurney, H.; Mulugeta, B.; Roberts, S.A.; Nardo, L.G. A Common Asn680Ser Polymorphism in the Follicle-Stimulating Hormone Receptor Gene Is Not Associated with Ovarian Response to Gonadotropin Stimulation in Patients Undergoing in Vitro Fertilization. Fertil. Steril. 2013, 99, 149–155. [Google Scholar] [CrossRef]
Table 1. Baseline characteristics of the population studied (n = 94).
Table 1. Baseline characteristics of the population studied (n = 94).
Basal CharacteristicsValues
Age (years)30.71 ± 2.61
BMI (kg/m2)22.94 ± 2.35
AMH (ng/mL)2.70 ± 1.76
Antral follicle count12.36 ± 3.63
Basal FSH (IU/L)6.73 ± 1.98
Basal estradiol (pg/mL)80.65 ± 101.16
Table 2. Treatment outcomes (n = 94).
Table 2. Treatment outcomes (n = 94).
Treatment Outcomes *Values
Total FSH doses (IU)1725.33 ± 520.15
Days of stimulation11.24 ± 1.69
Estradiol at the day of hCG (pg/mL)1655.43 ± 895.59
Follicles > 10 mm11.04 ± 4.41
Follicles > 16 mm7.72 ± 3.15
Oocyte number9.51 ± 3.82
Mature oocyte number7.78 ± 3.39
Oocytes inseminated5.35 ± 3.50
Oocytes fertilized3.61 ± 2.55
Oocytes cryopreserved0.35 ± 1.36
Embryos cryopreserved6.73 ± 1.98
Embryos transferred1.65 ± 0.80
Cycles canceled for hyper-response2 (2.1%)
OHSS1 (1.1%)
Implantation rate45.7%
Pregnancy rate (β-hCG) per cycle42.5%
Ongoing pregnancy rate per cycle34.04%
Pregnancy rate (β-hCG) per embryos transferred30.5%
Ongoing pregnancy rate per embryos transferred25.3%
Miscarriage rate per cycle9.4%
* Continuous data are expressed as mean ± standard deviation; categorical data as percentage.
Table 3. Treatment outcomes in patients stratified according to the FSHR 307 (rs6165) polymorphism.
Table 3. Treatment outcomes in patients stratified according to the FSHR 307 (rs6165) polymorphism.
Homozygous
A/A (n = 24)
Heterozygous
A/G (n = 50)
Homozygous
G/G (n = 20)
p-Value
Total FSH doses (IU)1781.23 ± 568.451730.04 ± 550.191647.17 ± 383.580.536
FSH/oocytes243.42 ± 97.60338.52 ± 251.80252.60 ± 166.330.050
Days of stimulation11.13 ± 1.6811.35 ± 1.8211.10 ± 1.410.769
Endometrial thickness (mm)9.70 ± 1.1510.38 ± 2.0010.28 ± 2.090.547
Estradiol on the day of hCG (pg/mL)1555.24 ± 663.851607.54 ± 906.211859.42 ± 1092.750.513
Follicles ≥ 16 mm on the day of hCG7.63 ± 2.727.73 ± 3.267.80 ± 3.500.983
Oocyte number9.58 ± 3.329.24 ± 3.5710.10 ± 4.980.685
Mature oocyte number8.13 ± 2.727.50 ± 3.718.06 ± 3.470.643
Oocytes inseminated6.08 ± 3.265.18 ± 3.604.90 ± 3.550.346
Oocytes fertilized3.92 ± 2.533.60 ± 2.663.25 ± 2.380.537
Oocytes cryopreserved0.21 ± 1.020.36 ± 1.440.50 ± 1.540.802
Embryos cryopreserved0.96 ± 1.811.16 ± 2.050.65 ± 1.310.534
Embryos transferred1.63 ± 0.771.56 ± 0.791.90 ± 0.850.236
Implantation rate10/3923/7710/380.795
Pregnancy rate per embryos transferred12/3924/7711/380.867
Ongoing pregnancy rate per embryos transferred 9/3921/779/380.792
Pregnancy rate per cycle12/2424/5011/200.930
Ongoing pregnancy rate per cycle9/2421/509/200.863
Bold indicates statistical significance.
Table 4. Treatment outcomes in patients stratified according to the FSHR 680 (rs6166) polymorphism.
Table 4. Treatment outcomes in patients stratified according to the FSHR 680 (rs6166) polymorphism.
Homozygous
A/A (n = 24)
Heterozygous
A/G (n = 49)
Homozygous
G/G (n = 21)
p-Value
Total FSH doses (IU)1809.76 ± 563.381725.25 ± 554.531633.02 ± 379.450.698
FSH/oocytes248.80 ± 96.34333.44 ± 250.88252.60 ± 166.330.049
Days of stimulation11.42 ± 1.7211.23 ± 1.8111.50 ± 1.400.804
Endometrial thickness (mm)9.70 ± 1.1510.38 ± 2.0010.28 ± 2.090.547
Estradiol on the day of hCG (pg/mL)1624.09 ± 722.401568.46 ± 880.901859.42 ± 1092.750.514
Follicles ≥ 16 mm on the day of hCG7.58 ± 2.707.71 ± 3.287.90 ± 3.450.944
Oocyte number9.67 ± 3.339.20 ± 3.5910.05 ± 4.860.697
Mature oocyte number8.22 ± 2.787.45 ± 3.678.06 ± 3.470.725
Oocytes inseminated6.21 ± 3.355.18 ± 3.554.76 ± 3.520.476
Oocytes fertilized4.04 ± 2.563.57 ± 2.653.19 ± 2.380.694
Oocytes cryopreserved0.21 ± 1.020.37 ± 1.450.48 ± 1.500.779
Embryos cryopreserved1.00 ± 1.821.16 ± 2.060.62 ± 1.280.581
Embryos transferred1.63 ± 0.771.55 ± 0.791.90 ± 0.830.273
Implantation rate10/3721/7712/400.844
Pregnancy rate per embryos transferred12/3722/7713/400.839
Ongoing pregnancy rate per embryos transferred9/3719/7711/400.848
Pregnancy rate per cycle12/2323/5013/210.812
Ongoing pregnancy rate per cycle9/2319/5011/210.867
Bold indicates statistical significance.
Table 5. Treatment outcomes in patients stratified according to the FSHR-29 (rs1394205).
Table 5. Treatment outcomes in patients stratified according to the FSHR-29 (rs1394205).
Homozygous
G/G (n = 54)
Heterozygous
G/A (n = 31)
Homozygous
A/A (n = 8)
p-Value
Total FSH doses (IU)1730.04 ± 554.551745.19 ± 476.551601.71 ± 487.460.804
FSH/oocytes322.78 ± 239.49229.73 ± 109.79312.83 ± 161.030.186
Days of stimulation11.20 ± 1.5211.39 ± 1.8711.50 ± 1.400.733
Endometrial thickness (mm)10.29 ± 2.009.91 ± 1.5510.47 ± 1.360.776
Estradiol on the day of hCG (pg/mL)1518.52 ± 742.111832.04 ± 1184.502039.14 ± 706.300.197
Follicles ≥ 16 mm on the day of hCG8.07 ± 3.236.90 ± 2.838.50 ± 3.500.197
Oocyte number9.60 ± 3.909.58 ± 3.848.63 ± 3.540.794
Mature oocyte number7.67 ± 3.658.46 ± 2.906.50 ± 2.830.344
Oocytes inseminated5.22 ± 3.495.90 ± 3.674.13 ± 2.700.404
Oocytes fertilized3.47 ± 2.673.97 ± 2.603.13 ± 1.360.595
Oocytes cryopreserved0.38 ± 1.380.19 ± 1.080.75 ± 2.120.572
Embryos cryopreserved1.05 ± 2.031.03 ± 1.720.50 ± 0.760.730
Embryos transferred1.62 ± 0.851.61 ± 0.762.00 ± 0.530.435
Implantation rate18/8821/504/160.934
Pregnancy rate per embryos transferred22/8821/504/160.754
Ongoing pregnancy rate per embryos transferred16/8819/504/160.770
Pregnancy rate per cycle22/5521/314/80.879
Ongoing pregnancy rate per cycle16/5519/314/80.435
Table 6. Treatment outcomes in patients stratified according to the LHCGR 291 (rs12470652) polymorphism.
Table 6. Treatment outcomes in patients stratified according to the LHCGR 291 (rs12470652) polymorphism.
T/T (n = 87)C/T (n = 7)p-Value
Total FSH doses (IU)1736.38 ± 534.531568.75 ± 196.170.449
FSH/oocytes305.86 ± 208.66147.65 ± 46.770.069
Days of stimulation11.21 ± 1.7011.57 ± 1.620.588
Endometrial thickness (mm)9.97 ± 1.4911.04 ± 2.760.146
Estradiol on the day of hCG (pg/mL)1580.60 ± 860.032733.00 ± 747.230.005
Follicles ≥ 16 mm on the day of hCG7.80 ± 3.166.71 ± 2.980.382
Oocyte number9.28 ± 3.8112.43 ± 2.820.035
Mature oocyte number7.45 ± 3.2111.43 ± 3.410.002
Oocytes inseminated4.92 ± 3.2010.71 ± 2.560.001
Oocytes fertilized3.24 ± 2.168.14 ± 2.910.001
Oocytes cryopreserved0.38 ± 1.410.00 ± 0.000.480
Embryos cryopreserved0.75 ± 1.474.14 ± 3.080.001
Embryos transferred1.68 ± 0.811.29 ± 0.490.213
Implantation rate41/1452/90.992
Pregnancy rate per embryos transferred45/1454/90.639
Ongoing pregnancy rate per embryos transferred 37/1452/90.861
Pregnancy rate per cycle45/874/70.907
Ongoing pregnancy rate per cycle37/872/70.747
Bold indicates statistical significance.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alviggi, C.; Longobardi, S.; Papaleo, E.; Santi, D.; Alfano, S.; Vanni, V.S.; Campitiello, M.R.; De Rosa, P.; Strina, I.; Huhtaniemi, I.; et al. Genetic Variants of Gonadotropins and Their Receptors Could Influence Controlled Ovarian Stimulation: IVF Data from a Prospective Multicenter Study. Genes 2023, 14, 1269. https://doi.org/10.3390/genes14061269

AMA Style

Alviggi C, Longobardi S, Papaleo E, Santi D, Alfano S, Vanni VS, Campitiello MR, De Rosa P, Strina I, Huhtaniemi I, et al. Genetic Variants of Gonadotropins and Their Receptors Could Influence Controlled Ovarian Stimulation: IVF Data from a Prospective Multicenter Study. Genes. 2023; 14(6):1269. https://doi.org/10.3390/genes14061269

Chicago/Turabian Style

Alviggi, Carlo, Salvatore Longobardi, Enrico Papaleo, Daniele Santi, Simona Alfano, Valeria Stella Vanni, Maria Rosaria Campitiello, Pasquale De Rosa, Ida Strina, Ilpo Huhtaniemi, and et al. 2023. "Genetic Variants of Gonadotropins and Their Receptors Could Influence Controlled Ovarian Stimulation: IVF Data from a Prospective Multicenter Study" Genes 14, no. 6: 1269. https://doi.org/10.3390/genes14061269

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop