Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Hum Reprod ; 34(12): 2495-2512, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31820802

ABSTRACT

STUDY QUESTION: Can kisspeptin treatment induce gonadotrophin responses and ovulation in preclinical models and anovulatory women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER: Kisspeptin administration in some anovulatory preclinical models and women with PCOS can stimulate reproductive hormone secretion and ovulation, albeit with incomplete efficacy. WHAT IS KNOWN ALREADY: PCOS is a prevalent, heterogeneous endocrine disorder, characterized by ovulatory dysfunction, hyperandrogenism and deregulated gonadotrophin secretion, in need of improved therapeutic options. Kisspeptins (encoded by Kiss1) are master regulators of the reproductive axis, acting mainly at GnRH neurons, with kisspeptins being an essential drive for gonadotrophin-driven ovarian follicular maturation and ovulation. Altered Kiss1 expression has been found in rodent models of PCOS, although the eventual pathophysiological role of kisspeptins in PCOS remains unknown. STUDY DESIGN, SIZE, DURATION: Gonadotrophin and ovarian/ovulatory responses to kisspeptin-54 (KP-54) were evaluated in three preclinical models of PCOS, generated by androgen exposures at different developmental windows, and a pilot exploratory cohort of anovulatory women with PCOS. PARTICIPANTS/MATERIALS, SETTING, METHODS: Three models of PCOS were generated by exposure of female rats to androgens at different periods of development: PNA (prenatal androgenization; N = 20), NeNA (neonatal androgenization; N = 20) and PWA (post-weaning androgenization; N = 20). At adulthood (postnatal day 100), rats were subjected to daily treatments with a bolus of KP-54 (100 µg/kg, s.c.) or vehicle for 11 days (N = 10 per model and treatment). On Days 1, 4, 7 and 11, LH and FSH responses were assessed at different time-points within 4 h after KP-54 injection, while ovarian responses, in terms of follicular maturation and ovulation, were measured at the end of the treatment. In addition, hormonal (gonadotrophin, estrogen and inhibin B) and ovulatory responses to repeated KP-54 administration, at doses of 6.4-12.8 nmol/kg, s.c. bd for 21 days, were evaluated in a pilot cohort of anovulatory women (N = 12) diagnosed with PCOS, according to the Rotterdam criteria. MAIN RESULTS AND THE ROLE OF CHANCE: Deregulated reproductive indices were detected in all PCOS models: PNA, NeNA and PWA. Yet, anovulation was observed only in NeNA and PWA rats. However, while anovulatory NeNA rats displayed significant LH and FSH responses to KP-54 (P < 0.05), which rescued ovulation, PWA rats showed blunted LH secretion after repeated KP-54 injection and failed to ovulate. In women with PCOS, KP-54 resulted in a small rise in LH (P < 0.05), with an equivalent elevation in serum estradiol levels (P < 0.05). Two women showed growth of a dominant follicle with subsequent ovulation, one woman displayed follicle growth but not ovulation and desensitization was observed in another patient. No follicular response was detected in the other women. LIMITATIONS, REASONS FOR CAUTION: While three different preclinical PCOS models were used in order to capture the heterogeneity of clinical presentations of the syndrome, it must be noted that rat models recapitulate many but not all the features of this condition. Additionally, our pilot study was intended as proof of principle, and the number of participants is low, but the convergent findings in preclinical and clinical studies reinforce the validity of our conclusions. WIDER IMPLICATIONS OF THE FINDINGS: Our first-in-rodent and -human studies demonstrate that KP-54 administration in anovulatory preclinical models and women with PCOS can stimulate reproductive hormone secretion and ovulation, albeit with incomplete efficacy. As our rat models likely reflect the diversity of PCOS phenotypes, our results argue for the need of personalized management of anovulatory dysfunction in women with PCOS, some of whom may benefit from kisspeptin-based treatments. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research agreements between Ferring Research Institute and the Universities of Cordoba and Edinburgh. K.S. was supported by the Wellcome Trust Scottish Translational Medicine and Therapeutics Initiative (STMTI). Some of this work was undertaken in the MRC Centre for Reproductive Health which is funded by the MRC Centre grant MR/N022556/1. M.T.-S. is a member of CIBER Fisiopatología de la Obesidad y Nutrición, which is an initiative of Instituto de Salud Carlos III. Dr Mannaerts is an employee of Ferring International PharmaScience Center (Copenhagen, Denmark), and Drs Qi, van Duin and Kohout are employees of the Ferring Research Institute (San Diego, USA). Dr Anderson and Dr Tena-Sempere were recipients of a grant support from the Ferring Research Institute, and Dr Anderson has undertaken consultancy work and received speaker fees outside this study from Merck, IBSA, Roche Diagnostics, NeRRe Therapeutics and Sojournix Inc. Dr Skorupskaite was supported by the Wellcome Trust through the Scottish Translational Medicine and Therapeutics Initiative 102419/Z/13/A. The other authors have no competing interest.


Subject(s)
Kisspeptins/therapeutic use , Ovulation/drug effects , Polycystic Ovary Syndrome/drug therapy , Adult , Animals , Disease Models, Animal , Female , Follicle Stimulating Hormone/blood , Humans , Kisspeptins/pharmacology , Luteinizing Hormone/blood , Pilot Projects , Polycystic Ovary Syndrome/blood , Rats, Wistar , Young Adult
2.
Clin Pharmacol Ther ; 88(1): 79-87, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20520605

ABSTRACT

In controlled ovarian stimulation (COS), a single subcutaneous dose of corifollitropin alfa is used to initiate and sustain multifollicular growth for 7 days. The objective of this study was to determine the optimal dose of corifollitropin alfa. A pharmacokinetic model was developed to describe the time profile of corifollitropin alfa concentrations. Multiple parameters reflecting ovarian response were included in a pharmacokinetic-pharmacodynamic (PK-PD) model framework. An early decline in serum inhibin B was shown to be a sensitive marker for COS failure. Simulations were performed to select the lowest corifollitropin alfa dose that would result in a minimal cancellation rate: 100 microg for a group of women weighing 60 kg. With these doses, the predicted mean number of oocytes per started COS cycle was similar in the two groups, i.e., 12.1 and 13.2, respectively. The selected doses were tested in prospective clinical trials and were proven to be adequate.


Subject(s)
Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/pharmacology , Ovary/drug effects , Adult , Algorithms , Area Under Curve , Computer Simulation , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone, Human/pharmacokinetics , Gonadotropin-Releasing Hormone/pharmacology , Humans , Inhibins/metabolism , Models, Statistical , Oocytes/drug effects , Ovarian Follicle/drug effects , Stimulation, Chemical
3.
Hum Reprod ; 24(12): 3063-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19684043

ABSTRACT

BACKGROUND: Corifollitropin alfa, a fusion protein lacking LH activity, has a longer elimination half-life and extended time to peak levels than recombinant FSH (rFSH). A single injection of corifollitropin alfa may replace seven daily gonadotrophin injections during the first week of ovarian stimulation. METHODS: In this large, double-blind, randomized, non-inferiority trial the ongoing pregnancy rates were assessed after one injection of 150 microg corifollitropin alfa during the first week of stimulation and compared with daily injections of 200 IU rFSH using a standard GnRH antagonist protocol. RESULTS: The study population comprised 1506 treated patients with mean age of 31.5 years and body weight of 68.6 kg. Ongoing pregnancy rates of 38.9% for the corifollitropin alfa group and 38.1% for rFSH were achieved, with an estimated non-significant difference of 0.9% [95% confidence interval (CI): -3.9; 5.7] in favor of corifollitropin alfa. Stratified analyses of pregnancy rates confirmed robustness of this primary outcome by showing similar results regardless of IVF or ICSI, or number of embryos transferred. A slightly higher follicular response with corifollitropin alfa resulted in a higher number of cumulus-oocyte-complexes compared with rFSH [estimated difference 1.2 (95% CI: 0.5; 1.9)], whereas median duration of stimulation was equal (9 days) and incidence of (moderate/severe) ovarian hyperstimulation syndrome was the same (4.1 and 2.7%, respectively P = 0.15). CONCLUSION: Corifollitropin alfa is a novel and effective treatment option for potential normal responder patients undergoing ovarian stimulation with GnRH antagonist co-treatment for IVF resulting in a high ongoing pregnancy rate, equal to that achieved with daily rFSH. The trial was registered under ClinicalTrials.gov identifier NTC00696800.


Subject(s)
Follicle Stimulating Hormone, Human/therapeutic use , Follicle Stimulating Hormone, beta Subunit/therapeutic use , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Infertility, Female/therapy , Ovulation Induction/methods , Adolescent , Adult , Clinical Protocols , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination/statistics & numerical data , Female , Fertilization in Vitro/methods , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, beta Subunit/administration & dosage , Humans , Injections, Subcutaneous , Oocyte Retrieval/statistics & numerical data , Pregnancy , Pregnancy Rate , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Young Adult
4.
Hum Reprod Update ; 15(3): 309-21, 2009.
Article in English | MEDLINE | ID: mdl-19182099

ABSTRACT

BACKGROUND: Recombinant DNA technologies have been used to develop longer-acting therapeutic proteins. One approach is to introduce sequences containing additional glycosylation sites. Using this technique, a new chimeric gene has been developed containing the coding sequences of the FSH beta-subunit and the C-terminal peptide of the hCG beta-subunit, which bears four O-linked oligosaccharide binding sites. Co-expression of the alpha-subunit and the chimeric FSH beta-subunit produces a new recombinant molecule, named corifollitropin alfa, with a prolonged elimination half-life and enhanced in vivo bioactivity compared with wild-type FSH. METHODS: Medline searches by subject and additional searching by hand. RESULTS: Initial studies in pituitary suppressed female volunteers confirmed the extended half-life of the compound. Phase II studies have shown that corifollitropin alfa is able to induce and sustain multi-follicular growth for an entire week in women undergoing ovarian stimulation using GnRH antagonist co-treatment for IVF. Corifollitropin alfa regimens have been developed with dosages of 100 and 150 microg, for patients with body weight 60 kg, respectively. CONCLUSIONS: Corifollitropin alfa is the first long-acting hybrid molecule with sustained follicle-stimulating activity developed for the induction of multi-follicular growth along with GnRH antagonist co-treatment for IVF. This new treatment option may be simpler and more convenient for patients compared with conventional long protocols of daily FSH injections in combination with GnRH agonist co-treatment. The safety and efficacy of such regimens is currently being evaluated in large comparative phase III clinical trials. The development of corifollitropin alfa is the first step towards a new generation of recombinant gonadotrophins.


Subject(s)
DNA, Recombinant/chemistry , Follicle Stimulating Hormone, Human/pharmacology , Ovarian Follicle/drug effects , Ovulation Induction/methods , Recombinant Fusion Proteins/pharmacokinetics , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Fertilization in Vitro , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/pharmacokinetics , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/pharmacokinetics , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Half-Life , Humans , Injections, Subcutaneous , Ovarian Follicle/growth & development
5.
J Endocrinol Invest ; 28(11): 990-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16483177

ABSTRACT

Elevated LH concentration is a common feature in polycystic ovary syndrome (PCOS). This study was designed to establish whether elevated LH levels in PCOS might be suppressed to normal range values by the administration of different low doses of GnRH antagonist, which subsequently might reverse the anovulatory status of these patients. Twenty-four PCOS patients with elevated endogenous LH concentrations were randomized into 3 different dose groups, receiving either 0.125 mg (Group A), 0.250 mg (Group B) or 0.500 mg (Group C) ganirelix sc daily for 7 subsequent days. During the first day of treatment, LH and FSH levels were assessed at 20 min intervals, during 8 h. Thereafter LH, FSH, androgens, estradiol (E2) and inhibins were assessed daily and frequent ultrasound scans were performed for 7 days to record follicle development. Repeated GnRH antagonist administration induced a significant suppression of LH (and to a lesser extent of FSH) serum levels, which was comparable between the different doses. Six hours after ganirelix administration, endogenous LH was suppressed by 49, 69 and 75%, and endogenous FSH was suppressed by 23, 19 and 25%, respectively. The decrease in serum LH and FSH levels was transient and lasted for 12 h, after which serum levels returned to baseline levels at 24 h after drug administration. Androgen levels were not significantly suppressed using this regimen. E2 levels decreased significantly (p < 0.001) and suppression was most pronounced in Group C. Spontaneous follicle development or ovulations were not recorded during the course of treatment. In conclusion, the present study demonstrates that the GnRH antagonist ganirelix is capable of normalising elevated LH levels in PCOS patients, in doses similar to the ones previously shown to prevent a premature LH rise during ovarian hyperstimulation for in vitro fertilization (IVF). In addition, the transient suppression of elevated endogenous LH levels per se does not re-establish normal follicle development in PCOS. However, follicle development may be insufficiently supported by the accompanied subtle suppression of endogenous FSH. Similarly, a transient decline in E2 levels does not effectively restore normal pituitary ovarian feedback. Moreover, these results support the contention of a limited role of LH in the pathogenesis of PCOS.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Luteinizing Hormone/antagonists & inhibitors , Neurosecretory Systems/drug effects , Ovary/physiopathology , Polycystic Ovary Syndrome/drug therapy , Adult , Androgens/metabolism , Estradiol/metabolism , Female , Follicle Stimulating Hormone/antagonists & inhibitors , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Inhibins/metabolism , Luteinizing Hormone/metabolism , Ovary/drug effects , Ovulation/drug effects , Polycystic Ovary Syndrome/diagnosis
6.
J Clin Endocrinol Metab ; 89(12): 6297-304, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579793

ABSTRACT

In a double-blind, placebo-controlled, randomized study, 55 anovulatory subjects received a single s.c. injection of placebo (n = 10) or recombinant long-acting FSH [FSH-carboxy terminal peptide (CTP), ORG 36286, corifollitropin alfa; NV Organon, The Netherlands] in doses of 7.5 (n = 13), 15 (n = 10), 30 (n = 11), or 60 microg (n = 11). The injection was given 2 or 3 d after the onset of a spontaneous or progestagen-induced withdrawal bleed. After drug administration, the induced follicular response varied widely among subjects in each dose group. The percentage of subjects with a follicular response (at least one follicle > or = 10.0 mm) increased with the dose (P < 0.01) and was 10, 31, 70, 73, and 82% in the placebo and 7.5-, 15-, 30-, and 60-microg treatment groups, respectively. In responding subjects, the average maximum number of follicles was 4.0, 7.6, 13.4, and 20.0, respectively, which was reached at 6.5, 6.9, 6.6, and 8.2 d after a single dose of 7.5, 15, 30, and 60 microg FSH-CTP, respectively. The dose-response for the number of follicles was statistically significant within the dose range tested (P < 0.01). Peak serum inhibin-B levels were significantly correlated with serum estradiol (E2) levels (r = 0.84, P < 0.01), and peak concentrations of inhibin-B and E2 correlated with the number of follicles observed at the same time point (for both hormones; r = 0.47, P < 0.01). Overall per treatment group, serum E2 and inhibin B concentrations significantly increased only in the two highest FSH-CTP dose groups, reaching peak concentrations at d 3 in the 30-microg group and at d 5 in the 60-microg group. Thereafter these hormone values declined rapidly, returning to baseline within 1 wk after FSH-CTP administration. In total, nine of the 55 treated subjects (16.4%) ovulated after drug administration: one subject in the placebo group, two subjects in the 7.5-microg group, three subjects in the 15-microg group, two in the 30-microg group, and one in the 60-microg group. Three subjects had monofollicular ovulation after placebo (n = 1) and a single dose of 15 microg FSH-CTP (n = 2). In two subjects with too many preovulatory follicles, (multiple) ovulation was prevented by GnRH antagonist administration. Thus, a single low dose of long-acting FSH-CTP was able to induce one or more follicles to grow up to ovulatory sizes, but the anovulatory status was not reversed because the incidence of subsequent (mono)ovulations was low.


Subject(s)
Anovulation/complications , Anovulation/drug therapy , Follicle Stimulating Hormone, Human/administration & dosage , Infertility, Female/etiology , Adult , Anovulation/classification , Anovulation/physiopathology , Dose-Response Relationship, Drug , Double-Blind Method , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone, Human/adverse effects , Follicle Stimulating Hormone, Human/blood , Follicle Stimulating Hormone, Human/therapeutic use , Humans , Injections, Subcutaneous , Luteinizing Hormone/blood , Ovarian Follicle/drug effects , Ovarian Follicle/physiopathology , Ovulation , World Health Organization
7.
J Clin Endocrinol Metab ; 89(5): 2062-70, 2004 May.
Article in English | MEDLINE | ID: mdl-15126522

ABSTRACT

In a first feasibility study, the efficacy and safety of a single dose of recombinant long-acting FSH (FSH-CTP) were investigated in in vitro fertilization (IVF) patients undergoing controlled ovarian stimulation with a flexible GnRH antagonist protocol. Eligible subjects were randomized to receive a single dose of 120 micro g (n = 25), 180 microg (n = 24), or 240 microg (n = 25) corifollitropin alfa (FSH-CTP) or to start daily fixed doses of 150 IU recombinant FSH (rFSH) (n = 24, reference). Subjects who received a single dose of FSH-CTP continued 1 wk after injection (treatment d 8) with fixed daily doses of 150 IU rFSH (Puregon/Follistim) until the day of triggering final oocyte maturation. The terminal half-life of FSH-CTP was, on average, 65 h and dose independent. Cycle cancellation before human chorionic gonadotropin (hCG) administration occurred in only three subjects treated with FSH-CTP. The median duration of stimulation was 10.0 d in each FSH-CTP group and 9.0 d in the daily rFSH group. The total number of follicles at least 11 mm at stimulation d 8 and at the day of hCG administration tended to increase with dose of FSH-CTP, although a significant dose-response relationship was revealed only for the number of follicles at least 15 mm on the day of hCG (P = 0.03). Serum estradiol levels and inhibin-B levels were not significantly different between the four groups on d 8 and on the day of hCG. In total, 12 subjects (17.6%) in the FSH-CTP groups and two subjects (8.3%) in the rFSH group experienced a premature LH rise (defined as LH >or= 10 IU/liter) before the start of the GnRH antagonist (P value not significant between groups). This relatively high incidence of women demonstrating an early LH rise in the FSH-CTP groups may be related to the higher initial rises of serum estradiol and the use of a flexible GnRH antagonist protocol. The mean number of oocytes recovered per started cycle was higher in FSH-CTP-treated subjects compared with rFSH-treated subjects (significant at P = 0.03 for the 240- microg FSH-CTP group), but no difference could be noted between the number of good quality embryos (range of means, 3.8-4.8 per attempt), and equal numbers of embryos were available for embryo transfer. In summary, FSH-CTP appeared to be a potent inducer of multiple follicular growth; additional research will be needed to select the optimal FSH-CTP dose and treatment time interval.


Subject(s)
Fertilization in Vitro , Follicle Stimulating Hormone, Human/therapeutic use , Ovarian Follicle/physiology , Ovulation Induction/methods , Adult , Drug Administration Schedule , Female , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/adverse effects , Follicle Stimulating Hormone, Human/blood , Follicular Phase/blood , Hormones/blood , Humans , Luteal Phase/blood , Luteinizing Hormone/blood , Pregnancy , Pregnancy Rate , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/blood , Recombinant Proteins/therapeutic use , Treatment Outcome
9.
Fertil Steril ; 75(4): 688-93, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11287020

ABSTRACT

OBJECTIVE: To investigate relations between dose of GnRH antagonist and follicular phase characteristics. DESIGN: Randomized controlled multicenter trial. SETTING: Tertiary referral fertility centers. PATIENT(S): Three hundred and twenty-nine IVF patients. INTERVENTION(S): Ovarian stimulation for IVF with recombinant FSH starting on cycle day 2. From cycle day 7 onwards, cotreatment was provided with 0.0625, 0.125, 0.25, 0.5, 1.0, or 2.0 mg/d GnRH antagonist. MAIN OUTCOME MEASURE(S): Number of follicles, total follicular surface area, gonadotropin, and serum steroid concentrations. RESULT(S): In 311 patients, similar follicular growth was observed in all treatment groups. FSH levels increased during the follicular phase. Late follicular phase LH, androstenedione (AD), and E(2) levels showed a GnRH antagonist dose-related decrease (P<0.05). Late follicular phase E(2) levels correlated with total follicular surface area, AD, LH, and FSH (all P<0.001). Increasing GnRH antagonist doses exhibited additional suppressive action on E(2) levels. CONCLUSION(S): Follicular growth was unaffected by the dose of GnRH antagonist. A rise in follicular phase FSH serum concentrations during the follicular phase, largely related to exogenous FSH, enabled ongoing follicular growth in all treatment groups. The effect of GnRH antagonist on late follicular phase E(2) levels could not be exclusively attributed to suppression of LH.


Subject(s)
Fertilization in Vitro/methods , Follicle Stimulating Hormone/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/therapeutic use , Ovarian Follicle/physiology , Ovulation Induction/methods , Recombinant Proteins/therapeutic use , Adolescent , Adult , Embryo Transfer , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone, Human , Hormone Antagonists/therapeutic use , Humans , Luteinizing Hormone/blood , Menstrual Cycle/physiology , Ovarian Follicle/diagnostic imaging , Ovarian Follicle/drug effects , Pregnancy , Progesterone/blood , Regression Analysis , Treatment Outcome , Ultrasonography
10.
Fertil Steril ; 73(6): 1187-93, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10856481

ABSTRACT

OBJECTIVE: To assess pharmacokinetics (PK) and pharmacodynamics (PD) of subcutaneous (s.c.) administration of recombinant FSH in comparison with the intramuscular (i.m.) route. DESIGN: Open, group-comparative, randomized, multiple-dose study. SETTING: Phase I Clinical Research Unit.Volunteer(s): Forty-six healthy female volunteers. INTERVENTION(S): All volunteers were treated with Lyndiol contraceptive pills for 6 weeks to suppress pituitary function. After 3 weeks of Lyndiol, volunteers were randomized to 75 IU, 150 IU, or 225 IU s.c. or 150 IU i.m. of recombinant FSH, administered once daily for 7 days. Serum samples were collected to determine immunoreactive FSH, LH, and E(2) levels. Ultrasonography was performed for measurement of follicular growth. MAIN OUTCOME MEASURE(S): FSH pharmacokinetic parameters, number, and size of follicles. RESULT(S): The s.c. doses tested showed dose-proportional pharmacokinetics. Subcutaneous and i.m. administration of 150 IU of recombinant FSH were bioequivalent. For the 75-IU group almost no follicles >/=10 mm were found. The mean (+/-SD) number of follicles >/=8 mm on the day of maximum stimulation in the 150 IU and 225 IU s. c. and 150 IU i.m. groups were 14.0 +/- 7.1, 14.3 +/- 8.2, and 6.5 +/- 4.7. CONCLUSION(S): Pharmacokinetics of recombinant FSH were dose proportional within the dose range studied (75-225 IU). Subcutaneous and i.m. administration of 150 IU was bioequivalent with respect to pharmacokinetics, but after s.c. administration the number of growing follicles and estradiol response were higher.


Subject(s)
Follicle Stimulating Hormone/therapeutic use , Adult , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/pharmacokinetics , Follicle Stimulating Hormone, Human , Humans , Injections, Intramuscular , Injections, Subcutaneous , Ovarian Follicle/anatomy & histology , Ovarian Follicle/drug effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Reference Values , Therapeutic Equivalency
11.
Fertil Steril ; 72(6): 1001-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10593371

ABSTRACT

OBJECTIVE: To assess the absolute bioavailability of ganirelix (Antagon/Orgalutran; NV Organon, Oss, the Netherlands) after a single SC injection. DESIGN: Randomized, crossover, pharmacokinetic study. SETTING: Phase I clinical research unit. PATIENT(S): Nineteen healthy female volunteers of reproductive age. INTERVENTION(S): Two separate injections of 0.25 mg of ganirelix were given, one subcutaneously and one intravenously, with a washout period of 1 week between injections. Blood samples were taken for assessment of serum ganirelix concentrations, and blood pressure, heart rate, and adverse events were monitored. MAIN OUTCOME MEASURE(S): Pharmacokinetic parameters. RESULT(S): Fifteen subjects were evaluated. The mean concentration-time profile after SC administration was comparable to that after IV administration. The mean (+/- SD) peak concentration and time of occurrence after SC administration were 14.8+/-3.2 ng/mL and 1.1+/-0.3 hours, respectively. The mean (+/- SD) half-lives after IV administration and SC administration were highly similar (12.7+/-3.7 hours and 12.8+/-4.3 hours, respectively). Mean (+/- SD) AUC0-infinity (area under the concentration-time curve) values of 105+/-11 ng/mL x hours and 96+/-12 ng/mL x hours were calculated for IV administration and SC administration, respectively, resulting in an absolute mean (+/- SD) bioavailability of 91.3%+/-6.7%. Both treatments were well tolerated. CONCLUSION(S): Ganirelix is absorbed rapidly and extensively after SC administration, resulting in a high absolute bioavailability of >90%.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Adult , Biological Availability , Cross-Over Studies , Female , Gonadotropin-Releasing Hormone/adverse effects , Gonadotropin-Releasing Hormone/pharmacokinetics , Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Injections, Intravenous , Injections, Subcutaneous , Reference Values
12.
Fertil Steril ; 72(6): 1006-12, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10593372

ABSTRACT

OBJECTIVE: To assess the dose-proportionality and pharmacodynamic properties of multiple doses of ganirelix (Antagon/Orgalutran; NV Organon, Oss, the Netherlands). DESIGN: Randomized, parallel, pharmacokinetic, and pharmacodynamic study. SETTING: Phase I clinical research unit. PATIENT(S): Three groups of 15 healthy female volunteers of reproductive age. INTERVENTION(S): Subcutaneous injections of 0.125 mg, 0.25 mg, or 0.50 mg of ganirelix were given once daily for 7 days. Blood samples were taken to assess serum ganirelix, LH, FSH, and E2 concentrations. MAIN OUTCOME MEASURE(S): Pharmacokinetic parameters and hormone suppression. RESULT(S): Steady-state levels were reached between days 2 and 3. Peak concentrations, which occurred approximately 1 hour after dosing, increased in a dose-proportional manner and averaged 5.2 ng/mL, 11.2 ng/mL, and 22.2 ng/mL for the 0.125-mg, 0.25-mg, and 0.50-mg doses, respectively. Corresponding mean values for the area under the curve over one dosing interval (24 hours) were 33 ng x h/mL, 77.1 ng x h/mL, and 137.8 ng x h/mL, respectively. After the last 0.25-mg dose of ganirelix, serum LH, FSH, and E2 concentrations were maximally decreased (by 74%, 32%, and 25% at 4 hours, 16 hours, and 16 hours after injection, respectively). Serum hormone levels returned to pretreatment values within 2 days after the last injection. CONCLUSION(S): The pharmacokinetics of ganirelix were dose-proportional within the dose range studied. Multiple injections resulted in immediate suppression of gonadotropins, which was rapidly reversed after treatment discontinuation.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Adult , Depression, Chemical , Dose-Response Relationship, Drug , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/adverse effects , Gonadotropin-Releasing Hormone/pharmacokinetics , Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Luteinizing Hormone/blood , Reference Values
13.
Hum Reprod ; 13(6): 1461-4, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9688371

ABSTRACT

The objective of this study was to compare the bioavailability of s.c. and i.m. administration of human chorionic gonadotrophin (HCG; Pregnyl). In a randomized, single-centre, three-way cross-over study, 18 healthy pituitary-suppressed volunteers were assigned to single HCG injections of 5000 and 10,000 IU i.m. and 10,000 IU s.c. Rate (Cmax, t(max)) and extent [area under curve from zero to infinity (AUC(0-infinity))] of absorption of HCG were determined. Serum immunoactive HCG increased from 0.4-0.5 IU/l at baseline to mean peak concentrations, which were reached 20 h after injection of 156 IU/l with 5000 IU i.m., of 307 IU/l with 10,000 IU i.m. and of 339 IU/l with 10,000 IU s.c. Eight days after administration, < 10% of the maximum HCG activity was found for each regimen. The elimination half-life (t(1/2)) was on average 32-33 h, irrespective of the treatment regimen. Intramuscular and s.c. injections of 10,000 IU HCG were bioequivalent with respect to AUC(0-infinity). The Cmax and t(max) were also similar between the two administration routes but bioequivalence could not be proven due to intersubject variability. Intramuscular doses of 5000 IU and 10,000 IU HCG were dose-proportional. Since s.c. HCG is bioequivalent to i.m. HCG with respect to extent of absorption (its major pharmacokinetic variable) and is well tolerated, the s.c. administration route may be effectively and safely used in assisted reproduction. Moreover, since s.c. injection can be performed by the patients themselves, acceptability may be enhanced.


Subject(s)
Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacokinetics , Pituitary Gland/physiology , Administration, Oral , Adolescent , Adult , Contraceptives, Oral/administration & dosage , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Humans , Injections, Intramuscular , Pituitary Gland/drug effects
14.
Hum Reprod ; 13(3): 573-5, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9572414

ABSTRACT

This case report describes the first attempt to treat imminent ovarian hyperstimulation syndrome (OHSS) by using a gonadotrophin-releasing hormone (GnRH) antagonist. A 33 year old, normo-ovulatory woman undergoing in-vitro fertilization received daily subcutaneous injections of 150 IU of recombinant follicle-stimulating hormone (recFSH) from cycle day 2, together with GnRH antagonist (ganirelix) 0.125 mg from cycle day 7 onwards. On cycle day 10 the patient was found to have a serum oestradiol concentration of 16 500 pmol/l and, on ultrasound examination, four preovulatory (>16 mm) and nine intermediate sized (10-16 mm) follicles. RecFSH injections were discontinued, human chorionic gonadotrophin (HCG) withheld, whereas the ganirelix dose was increased to 2 mg/d. This regimen led to a rapid decrease in serum oestradiol concentrations and the decrease in ovarian size on ultrasound. Since GnRH antagonists will become clinically available for in-vitro fertilization programmes in the near future this suggested regimen might have a role in preventing severe OHSS.


Subject(s)
Fertilization in Vitro , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Ovarian Hyperstimulation Syndrome/prevention & control , Ovulation Induction/adverse effects , Adult , Estradiol/blood , Female , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Luteinizing Hormone/metabolism , Ovarian Follicle/diagnostic imaging , Ovarian Hyperstimulation Syndrome/etiology , Recombinant Proteins , Ultrasonography
15.
Fertil Steril ; 68(1): 138-42, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9207599

ABSTRACT

OBJECTIVE: To assess ongoing pregnancy rates (PRs) in IVF after treatment with recombinant FSH (follitropin beta, Puregon; NV Organon, Oss, The Netherlands) as compared with urinary gonadotropins. DESIGN: A combined analysis of three prospective, multicenter, randomized, comparative trials. SETTING: Twenty-five IVF centers in 13 countries. PATIENT(S): Six hundred ninety-seven infertile women receiving recombinant FSH and 463 women receiving hMG or urinary FSH and undergoing one cycle of controlled ovarian hyperstimulation and IVF-ET. INTERVENTION(S): A center-based and study-based analysis weighing the treatment differences in individual centers and studies, respectively. MAIN OUTCOME MEASURES(S): Pregnancy rate at least 12 weeks after ET per started cycle. RESULTS(S): In the center-based analysis, the ongoing PR was 22.9% for recombinant FSH and 17.9% for urinary gonadotropins. The 5.0% treatment difference (95% confidence interval [CI], 0.2% to 9.7%) was significant. When the results of the cryoprogram were included, the treatment difference increased to 6.4% (95% CI, 1.4% to 11.3%). Also in the study-based analysis, significantly higher PRs were seen after follitropin beta treatment. CONCLUSION(S): Follitropin beta (Puregon) used for controlled ovarian hyperstimulation in IVF yields significantly higher PRs compared with urinary gonadotropins.


Subject(s)
Fertilization in Vitro/methods , Follicle Stimulating Hormone/standards , Pregnancy Rate , Adult , Embryo Transfer , Female , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/urine , Follicle Stimulating Hormone, beta Subunit , Humans , Menotropins/administration & dosage , Prospective Studies , Recombinant Proteins/administration & dosage , Recombinant Proteins/standards
16.
Hum Reprod Update ; 2(2): 153-61, 1996.
Article in English | MEDLINE | ID: mdl-9079410

ABSTRACT

Single-dose and multiple-rising dose studies of recombinant follicle stimulating hormone (rFSH) in hypogonadotrophic male and female volunteers demonstrated that the rate of FSH absorption after i.m. injection is higher in men than in women. In the absence of endogenous FSH, a correlation between serum FSH and body weight became apparent. The elimination half-life of rFSH was not different between the sexes and was comparable with urinary FSH. However, the in-vitro bio:immuno ratio of serum FSH was significantly higher after the administration of rFSH than after urinary FSH. When rFSH was administered daily with a fixed dose, steady state levels were reached within 3-5 days. Serum FSH concentrations increased in a dose-dependent manner when the daily dose was increased weekly over 3 weeks from 75 to 225 IU. In hypogonadotrophic women, rFSH induced normal follicular growth whereas oestrogen synthesis was impaired. In women pituitary suppressed by a high-dose oral contraceptive, the daily administration of 150 IU rFSH for 1 week induced more and larger antral follicles than the same regimen with urinary FSH, whereas the serum immunoactive FSH concentrations measured 24 h after each dosing were similar. It is concluded that even though equal or lower serum immunoactive FSH concentrations were obtained following the administration of rFSH compared with urinary FSH, circulating bioactive FSH concentrations were higher. Therefore, the conventional idea that serum immunoreactive FSH correlates positively with the magnitude of the ovarian response should be reconsidered.


Subject(s)
Follicle Stimulating Hormone/pharmacokinetics , Clinical Trials as Topic , Contraceptives, Oral, Combined/administration & dosage , Drug Combinations , Female , Follicle Stimulating Hormone/pharmacology , Gonadotropins/deficiency , Half-Life , Humans , Lynestrenol/administration & dosage , Male , Mestranol/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Sex Factors
17.
Hum Reprod Update ; 2(2): 162-71, 1996.
Article in English | MEDLINE | ID: mdl-9079411

ABSTRACT

The clinical assessment of recombinant follicle stimulating hormone (rFSH; Puregon) in assisted reproduction technologies such as in-vitro fertilization (IVF) has probably been the most extensive clinical trial programme ever performed for the evaluation of a new fertility drug. It started with a pilot study to evaluate the potential of rFSH to stimulate the ovaries in the absence of luteinizing hormone (LH) using various gonadotrophin-releasing hormone (GnRH) agonists. After it became clear that FSH-induced steroidogenesis was not jeopardized even after severe pituitary suppression, comparisons between rFSH and urinary FSH or human menopausal gonadotrophins were made using different GnRH agonists or no agonists at all. In addition, the effects of the route of administration (s.c. or i.m.) were assessed. The study with the strongest statistical power to truly assess clinically relevant differences between rFSH and urinary FSH included approximately 1000 patient cycles. It indicated that after rFSH treatment, significantly more oocytes were retrieved, more embryos obtained and, as a result, more pregnancies achieved when the results of the cryoprogramme were included.


Subject(s)
Fertilization in Vitro , Follicle Stimulating Hormone/administration & dosage , Buserelin/administration & dosage , Clinical Trials as Topic , Embryo Transfer , Fertility Agents, Female/administration & dosage , Humans , Luteolytic Agents/administration & dosage , Menotropins/administration & dosage , Multicenter Studies as Topic , Recombinant Proteins/administration & dosage , Triptorelin Pamoate/administration & dosage
18.
Hum Reprod ; 10(10): 2534-40, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8567765

ABSTRACT

Urinary follicle stimulating hormone (FSH) is being used for the treatment of human infertility. Recently, FSH manufactured by means of recombinant DNA technology with a much higher purity (> 99%) has become available. A prospective, randomized, assessor-blind, multicentre (n = 18) study was conducted in infertile women undergoing in-vitro fertilization comparing recombinant FSH (Org 32489, Puregon) and urinary FSH (Metrodin). Eligible subjects were randomized (recombinant versus urinary FSH = 3:2) and pretreated with buserelin for pituitary suppression. FSH was given until three or more follicles with a diameter of at least 17 mm were seen. After oocyte retrieval, fertilization routines were applied according to local procedures. No more than three embryos were replaced. In all, 585 subjects received recombinant FSH and 396 urinary FSH. Significantly more oocytes were retrieved after recombinant FSH treatment (mean adjusted for centre 10.84 versus 8.95, P < 0.0001). Ongoing pregnancy rates per attempt and transfer in the recombinant FSH group were 22.17 and 25.97% respectively, and in the urinary FSH group, 18.22 and 22.02% respectively (not significant). Ongoing pregnancy rates including pregnancies resulting from frozen-thawed embryo cycles were 25.7% for recombinant and 20.4% for urinary FSH (P = 0.05). Compared to urinary FSH, the total dose of FSH was significantly lower with recombinant FSH (2138 versus 2385 IU, P < 0.0001) in a significantly shorter treatment period (10.7 versus 11.3 days, P < 0.0001). No clinically relevant differences between recombinant and urinary FSH were seen with respect to safety variables. It is concluded that recombinant FSH (Puregon) is more effective than urinary FSH in inducing multifollicular development and achieving an ongoing pregnancy.


Subject(s)
Follicle Stimulating Hormone/therapeutic use , Follicle Stimulating Hormone/urine , Infertility, Female/therapy , Recombinant Proteins/therapeutic use , Cell Nucleus/ultrastructure , Embryo Transfer , Female , Humans , Oocytes/ultrastructure , Pregnancy , Prospective Studies
19.
Hum Reprod ; 9(7): 1237-42, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7962424

ABSTRACT

Seven women suffering from hypogonadism due to previous hypophysectomy, isolated gonadotrophin deficiency, or Kallman's syndrome [median age 39 years (range 24-45)] volunteered to participate in a study to assess ovarian response following multiple-dose administration of recombinant human follicle-stimulating hormone (rhFSH; Org 32489). Baseline serum FSH and luteinizing hormone (LH) concentrations were 0.25 (< 0.05-1.15) IU/l and 0.06 (< 0.05-0.37) IU/l, respectively. Subjects received daily i.m. injections of rhFSH for 3 weeks (week 1: 75 IU/day, week 2: 150 IU/day, week 3: 225 IU/day). Blood sampling and sonographic investigations were performed on alternate days. Steady-state FSH concentrations were reached approximately 3-5 days after alterations of the doses administered. Maximum FSH concentrations were between 7.1 and 11.8 IU/l, whereas serum LH concentrations remained unchanged. Due to absent follicle development and lack of a rise in immunoreactive inhibin (INH) (response failure possibly due to early ovarian failure or resistant ovary syndrome) in two subjects, analysis of ovarian response was restricted to five volunteers. Serum androstenedione levels showed no significant changes during rhFSH administration. Although serum immunoreactive INH concentrations reached normal late follicular values [659 (388-993) IU/l], serum oestradiol revealed only a minor increase [77 (18-210) pmol/l]. Moreover, growth of (multiple) ovarian follicles was observed up to pre-ovulatory sizes (> 15 mm) in these patients.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Follicle Stimulating Hormone/therapeutic use , Gonadotropins, Pituitary/deficiency , Ovary/drug effects , Adult , Female , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/blood , Humans , Hypogonadism/drug therapy , Hypogonadism/etiology , Hypogonadism/physiopathology , Hypophysectomy/adverse effects , Hypopituitarism/drug therapy , Hypopituitarism/etiology , Hypopituitarism/physiopathology , Inhibins/blood , Kallmann Syndrome/drug therapy , Kallmann Syndrome/physiopathology , Luteinizing Hormone/blood , Middle Aged , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Ovary/physiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
20.
J Clin Endocrinol Metab ; 74(6): 1471-3, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1592896

ABSTRACT

To evaluate the importance of luteinizing hormone (LH) for normal estrogen production and subsequent development of ovarian follicles, a woman with isolated gonadotropin deficiency (LH; 0.37 IU/L, FSH 1.2 IU/L) was monitored during recombinant human follicle-stimulating hormone (hFSHrec) administration with respect to ovarian follicular growth and steroid production. During the first week (75 IU/day hFSHrec im) a significant rise in serum FSH (4.9 IU/L) was observed in the absence of changes in serum estradiol (E2) concentrations (36-76 pmol/L). During the following five days 150 IU/day hFSHrec was administered resulting in a further increase of serum FSH levels (maximum 8.5 IU/L). Development of multiple follicles--maximum diameter 22 mm as observed by transvaginal sonography--emerged together with a minor rise in E2 levels (from 76 to 236 pmol/L) and with a minimal increase in endometrial thickness (below 6 mm). Six days following the last injection of hFSHrec, aspiration of 3 follicles (13, 15 and 18 mm) was performed and low intrafollicular androstenedione (AD) (less than 675 nmol/L) and E2 (less than 9400 pmol/L) concentrations as compared to normal follicles were found. These first data on hFSHrec administration in the human suggest that; a) FSH alone can induce growth of preovulatory follicles, b) follicle growth does occur in the presence of subnormal E2 levels, c) LH is needed for adequate AD biosynthesis as substrate for aromatase activity. This indicates that growth and steroidogenic granulosa cell activity may be differentially regulated.


Subject(s)
Chorionic Gonadotropin/therapeutic use , Estradiol/biosynthesis , Follicle Stimulating Hormone/deficiency , Follicle Stimulating Hormone/toxicity , Luteinizing Hormone/deficiency , Ovarian Follicle/physiology , Adult , Amenorrhea/etiology , Drug Evaluation , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/therapeutic use , Humans , Luteinizing Hormone/blood , Ovarian Follicle/drug effects , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL