WO2001000227A1 - Gonadotropin releasing hormone antagonist - Google Patents

Gonadotropin releasing hormone antagonist Download PDF

Info

Publication number
WO2001000227A1
WO2001000227A1 PCT/EP2000/005643 EP0005643W WO0100227A1 WO 2001000227 A1 WO2001000227 A1 WO 2001000227A1 EP 0005643 W EP0005643 W EP 0005643W WO 0100227 A1 WO0100227 A1 WO 0100227A1
Authority
WO
WIPO (PCT)
Prior art keywords
antagonist
gnrh
gnrh antagonist
amount
ganirelix
Prior art date
Application number
PCT/EP2000/005643
Other languages
French (fr)
Inventor
Henrik Johan Matthieu Maria De Greef
Bernadette Maria Julia Louise Mannaerts
Everardus Otto Maria Orlemans
Gerrit Voortman
Original Assignee
Akzo Nobel N.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Akzo Nobel N.V. filed Critical Akzo Nobel N.V.
Priority to JP2001505935A priority Critical patent/JP2003503358A/en
Priority to CA002341974A priority patent/CA2341974A1/en
Priority to AU62638/00A priority patent/AU6263800A/en
Priority to EP00949193A priority patent/EP1140132A1/en
Publication of WO2001000227A1 publication Critical patent/WO2001000227A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • A61K38/09Luteinising hormone-releasing hormone [LHRH], i.e. Gonadotropin-releasing hormone [GnRH]; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/02Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin
    • A61P5/04Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin for decreasing, blocking or antagonising the activity of the hypothalamic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/32Antioestrogens

Definitions

  • the present invention relates to the use of GnRH antagonists in controlled ovarian hyperstimulation (COH) as well as to a method to prevent premature LH surge. It also relates to a cartridge comprising said antagonist and a kit comprising said cartridge and FSH.
  • COH controlled ovarian hyperstimulation
  • LH Luteinizing Hormone
  • FSH Follicle Stimulating Hormone
  • LH surge is a consequence of the raise in estrogen levels brought about by the endogenous secretion of LH and FSH.
  • the estrogen is part of a positive feedback mechanism resulting in the elevated LH level.
  • GnRH analogues are useful for a variety of disorders in which immediate reversible suppression of the pituitary-gonadal axis is desired. This can in principle be achieved with GnRH agonists as well as with GnRH antagonists. In comparison to GnRH agonists, GnRH antagonists have the advantage of not inducing an initial release of gonadotropins (flare-up) and steroids before suppression.
  • GnRH agonists are clinically applied for the prevention of endogenous LH-surges during controlled ovarian hyperstimulation for Assisted Reproduction Techniques (ART).
  • Specific disadvantages of GnRH agonists are the initial flare-up and the rather long period until pituitary suppression becomes effective.
  • patients undergoing COH start only treatment with (recombinant) FSH after 2 to 3 weeks pretreatment with GnRH agonists.
  • GnRH antagonists by GnRH receptor competition provide an immediate inhibition of gonadotropin secretion, especially of LH.
  • GnRH antagonist treatment is only required during the few days when there is an increased risk for a premature LH surge.
  • the GnRH antagonist dosage range is critical: too low a GnRH antagonist dosage leading to prmature LH rises, while too high a GnRH antagonist dosage hampered follicular maturation.
  • the antagonist ganirelix for example a fixed amount being at least 0.125 mg but less than 1 mg and preferably about 0.25 mg was suggested (W098/58657).
  • the invention therefore relates to a pharmaceutical preparation comprising GnRH antagonist, while applying a dosage adjusted for body weight sufficient to prevent a premature LH surge and ensuring successful treatment outcome.
  • Such preparation is useful in the treatment of women undergoing COH.
  • a preferred antagonist according to the present invention is ganirelix which has the following chemical name:
  • the abbreviated structure is [N- Ac-D-Na(2) 1 ,D-pCIPhe ,D-Pal(3) 3 ,D-hArg(Et 2 ) 6 ,L-hArg(Et 2 ) 8 ,D-Ala 10 -GnRH.
  • the GnRH antagonist ganirelix is disclosed in US patent No. 4,801 ,577 for nonapeptide and decapeptide analogs of LHRH useful as LHRH antagonists. This patent, which is fully incorporated herein by reference, describes the method for the preparation of these compounds. It is indicated that the compounds described therein can be used for the prevention of ovarian hyperstimulation.
  • a daily range is suggested for administration of the active ingredient between 0.001 and 5 mg/kg body weight, preferably between 0.01 and 1 mg/kg.
  • the preparation is administered together with FSH during the days of ovarian stimulation when a premature LH rise may easily occur e.g. from day 5 of FSH administration onwards.
  • the preparation in its proposed dosage range has the advantage of providing an immediate effect that prevents an LH surge and at the same time maximizes the chances of establishing pregnancy.
  • Administration is usually stopped when sufficient follicles have matured and exogenous hCG/LH is given for induction of ovulation.
  • the amount of hCG/LH usually amounts 5000-10000 IU.
  • induction of ovulation can be performed by administration of a GnRH agonist.
  • the agonist instead of hCG/LH is usually given on the same day in an amount sufficient to trigger ovulation.
  • Suitable range is 10-1000 ⁇ g.
  • Suitable agonists are e.g. buserelin, triptorelin and luprorelin.
  • the exact regimen for administration might depend on the individual response and is finally to be decided by the clinician who treats the subject. For this reason the duration of initial ovarian stimulation with FSH alone as well as the duration of combined treatment with FSH/GnRH antagonist treatment may vary.
  • FSH treatment usually starts at menses day 1 , 2 or 3.
  • Ovarian stimulation with FSH alone may be continued up to 5 days in an amount of e.g. 150-225 IU.
  • FSH is administered preferably as a recombinant protein.
  • Treatment with GnRH antagonist may be started at the first day of FSH, but preferably such treatment starts at FSH treatment day 4 or 5.
  • the GnRH antagonist is administered in the previously determined amount according to the invention in combination with FSH in amounts between 50 - 600 IU, preferably between 100 - 300 IU.
  • GnRH antagonist treatment may last 2 - 14 days i.e. up to the moment whereupon the patient is treated with exogenous LH/hCG or a GnRH or GnRH agonist for ovulation induction.
  • ganirelix in an amount according to Formula I is used for the manufacture of a medicament to prevent a premature LH surge in women undergoing controlled ovarian hyperstimulation.
  • the pharmaceutical preparations for use according to the invention can be prepared in accordance with standard techniques such as for example are described in the standard reference, Gennaro et al. (Ed.), Remmington's Pharmaceutical Sciences, (18 th ed. Mack Publishing Company, 1990, e.g. Part 8: Pharmaceutical Preparations And Their Manufacture).
  • the active substance is mixed with or dissolved in a pharmaceutical acceptable carrier. Any conventional pharmaceutical carrier that does not interfere with performance of the active ingredient can be used in the preparations according to the present invention.
  • Formulations may contain as common excipients sterile water or saline, alkylene glycols such as propylene glycol, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphtalenes and the like.
  • the pharmaceutical preparation of the antagonist may be administered parenterally. Preferably it is administered subcutaneously, particularly in the form of liquid solutions or suspensions.
  • a typical formulation is a solution containing, in addition to the active substance in an amount as indicated above, glacial acetic acid, mannitol, and water adjusted to pH 5 with sodium hydroxide and / or hydrochloric acid.
  • preservations such as e.g. methyl- and propylparaben or benzylalcohol can be added.
  • the solutions can be packaged e.g. in glass vials, cartridges or in syringes.
  • a cartridge containing a sterile liquid formulation of ganirelix.
  • a cartridge means a closed container, such as an ampoule, a vial, a bottle or a bag comprising an amount of GnRH antagonist so as to administer accurately and preferably repeatedly to a patient a dosage of GnRH antagonist according to Formula I.
  • a cartridge may contain an amount of the liquid antagonist formulation corresponding to one or more therapeutic dosages of the antagonist. Preferably these dosages are to be applied in a single regimen.
  • the cartridges contain an amount of GnRH antagonist sufficient for 5 administrations.
  • the cartridges are preferably used in combination with a device making it possible to deliver adjustable dosages needed in the regimen.
  • a device for administration comprising a cartridge containing a sterile liquid formulation according to the invention.
  • a preferred device for administration is a pen-type injector, which comprise means for easy adjustment of the amount of a formulation that is to be injected.
  • pen type injectors are known per se, such as for instance the well known B-D Pen (a trademark of Becton Dickinson and Company), an insulin-injection system.
  • Adjustable cartridges according to the invention have the advantage of accurate self-administration thereby increasing the convenience for the patients.
  • kits for use in controlled ovarian hyperstimulation in female patients Such a kit comprises a GnRH antagonist in a dosage form and quantity so as to accurately administer to a patient in an amount according to Formula I, in a frequency effective to prevent a premature LH surge.
  • the kit comprises FSH in a dosage form and quantity suitable for administering in an amount and frequency effective to stimulate growth of follicles.
  • the kit may comprise also hCG/LH or GnRH agonists in a dosage form and quantity suitable for administering in an amount and frequency effective to induce ovulation.
  • the GnRH antagonist preferably is packaged in a cartridge. This cartridge preferably is to be used in combination with a device for administration such as a pen type injector allowing an adjustable and accurate administration of GnRH antagonist.
  • the kit might also comprise a pen type injector system.
  • FIG. 1 LH levels were measured at the start (just before the first injection of ganirelix) and end of ganirelix treatment.
  • the graph shows the pregnancies in relation to the various levels. LH levels are indicated in IU/L.
  • Figure 2 Area under the curve of ganirelix versus body weight of subjects in three pharmacokinetic studies. Circles mean protocol A; triangles mean protocol B and squares mean protocol C.
  • Figure 3 Chance of pregnancy versus body weight; results from Phase III efficacy study. Dots in the top of the graph correspond to pregnant subjects
  • Recombinant FSH (recFSH) treatment was started on day 2 or 3 of the menstrual cycle by a once daily SC injection. Just prior to the first injection of recFSH an hCG test was performed to exclude pregnancy, a blood sample for hormone analysis was taken and an ultrasonography (USS) was performed. During recFSH treatment day 1 through 5, the daily dose of recFSH was fixed to 150 international units (IU). On day 6 of recFSH treatment ganirelix treatment was started by daily SC administration until and including the day before the day of hCG.
  • the dose of recFSH was adjusted depending on the individual ovarian response as assessed by USS. From the first day of ganirelix i.e. from recFSH treatment day 6 onwards up to and including the day of hCG, a blood sample for hormone analysis was taken prior to drug administration. And an USS was performed, at least every two days.
  • LH levels were assessed by a standard LH specific assay at the Central Laboratory of the Analytisch Biochemisch Laboratorium (Assen, The Netherlands). From the data on LH levels, a plot was constructed in order to investigate the possible role of LH on pregnancy.
  • LH levels were measured at the start (just before the first injection of ganirelix) and end of ganirelix treatment.
  • Figure 1 shows the pregnancies in relation to the various levels.
  • Protocol A an open-label two-way crossover study to assess the absolute bioavailablity of 0.25 mg ganirelix after single injection.
  • Protocol B an open-label randomized, multiple dose parallel-design study to assess the dose- proportionality and the pharmacokinetic properties of ganirelix (0.125 , 0.25 and 0.5 mg) after repeated subcutaneous administration.
  • Protocol C an open, randomized, two-way crossover study to establish the local tolerance and bioavailability of ganirelix after multiple subcutaneous administration (2 mg).
  • Pharmaco-statistical models have been set up to describe the influence of body weight on the effectiveness of ganirelix for both the prevention of LH- rises and pregnancy outcome.
  • Spline functions have been applied to give the best and assumptionless mathematical description of the available data. Using these models optimal doses with respect to the prevention of LH-rises and pregnancy have been determined for different body weights.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Endocrinology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Diabetes (AREA)
  • Reproductive Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a method to prevent a premature LH surge. The method employs the administration of the gonadotropin releasing hormone antagonist ganirelix in an amount dependent on the body weight of the patient. The method can be used in combination with administration of exogenous FSH in the treatment of women undergoing controlled ovarian superovulation.

Description

Gonadotropin releasing hormone antagonist
The present invention relates to the use of GnRH antagonists in controlled ovarian hyperstimulation (COH) as well as to a method to prevent premature LH surge. It also relates to a cartridge comprising said antagonist and a kit comprising said cartridge and FSH.
The glycoprotein hormones Luteinizing Hormone (LH) and Follicle Stimulating Hormone (FSH) are released from the pituitary gland under control of Gonadotropin Releasing Hormone (GnRH). They act on the ovary to stimulate steroid synthesis and secretion and thus play a central role in the reproductive cycle.
In the normal cycle, there is a mid-cycle surge in LH concentration which is followed by ovulation. The LH surge is a consequence of the raise in estrogen levels brought about by the endogenous secretion of LH and FSH. The estrogen is part of a positive feedback mechanism resulting in the elevated LH level.
GnRH analogues are useful for a variety of disorders in which immediate reversible suppression of the pituitary-gonadal axis is desired. This can in principle be achieved with GnRH agonists as well as with GnRH antagonists. In comparison to GnRH agonists, GnRH antagonists have the advantage of not inducing an initial release of gonadotropins (flare-up) and steroids before suppression.
Currently, GnRH agonists are clinically applied for the prevention of endogenous LH-surges during controlled ovarian hyperstimulation for Assisted Reproduction Techniques (ART). Specific disadvantages of GnRH agonists are the initial flare-up and the rather long period until pituitary suppression becomes effective. Usually, patients undergoing COH start only treatment with (recombinant) FSH after 2 to 3 weeks pretreatment with GnRH agonists.
Women treated for this purpose without GnRH analogues, all show attenuated LH rises irrespective of the treatment schedule used. Usually these rises occur prematurely due to a positive feedback of rising estradiol (E2) produced by a cohort of relative small follicles. The exposure of non-mature follicles to high levels of LH leads to premature luteinisation of granulosa cells and hence to increased production of progesterone and decreased synthesis of E2. These changes lead to disrupted maturation and decreased fertilization and implantation rates. Success rates of COH cycles in which premature LH rises are detected, are reported to be low and often these cycles are canceled because the number and/or size of follicles is still too small.
GnRH antagonists by GnRH receptor competition provide an immediate inhibition of gonadotropin secretion, especially of LH. Thus, during COH by FSH, GnRH antagonist treatment is only required during the few days when there is an increased risk for a premature LH surge. It has been found that the GnRH antagonist dosage range is critical: too low a GnRH antagonist dosage leading to prmature LH rises, while too high a GnRH antagonist dosage hampered follicular maturation. For the antagonist ganirelix for example a fixed amount being at least 0.125 mg but less than 1 mg and preferably about 0.25 mg was suggested (W098/58657).
Surprisingly, however, it has now been found that there is no relationship between the implantation rate and level of LH (AUC), whereas there does exist a relationship between the GnRH antagonist levels (AUC) and the implantation rate. It has now been found that antagonist is to be administered in an amount depending on the body weight (BW).
The invention therefore relates to a pharmaceutical preparation comprising GnRH antagonist, while applying a dosage adjusted for body weight sufficient to prevent a premature LH surge and ensuring successful treatment outcome. Such preparation is useful in the treatment of women undergoing COH.
A preferred antagonist according to the present invention is ganirelix which has the following chemical name:
N-Acetyl-3-(2-naphtyl)-D-alanyl-4-chloro-D-phenylalanyl-3-(3-pyridyl)-D- alanyl-L-seryl-L-N9,N10-diethyl-D-homoarginyl-L-leucin-N9,N10-diethyl-L- homoarginyl-L-propyl-D-alanylamide acetate. The abbreviated structure is [N- Ac-D-Na(2)1,D-pCIPhe ,D-Pal(3)3,D-hArg(Et2)6,L-hArg(Et2)8,D-Ala10-GnRH. The GnRH antagonist ganirelix is disclosed in US patent No. 4,801 ,577 for nonapeptide and decapeptide analogs of LHRH useful as LHRH antagonists. This patent, which is fully incorporated herein by reference, describes the method for the preparation of these compounds. It is indicated that the compounds described therein can be used for the prevention of ovarian hyperstimulation. For human therapy a daily range is suggested for administration of the active ingredient between 0.001 and 5 mg/kg body weight, preferably between 0.01 and 1 mg/kg.
It has now been found that the optimal relationship between body weight and GnRH antagonist dosage can be defined (in micrograms) by the Formula: (5.5*BW - 166) ± 7% (Formula I) wherein BW represents the body weight of the patient in kg.
The preparation is administered together with FSH during the days of ovarian stimulation when a premature LH rise may easily occur e.g. from day 5 of FSH administration onwards. The preparation in its proposed dosage range has the advantage of providing an immediate effect that prevents an LH surge and at the same time maximizes the chances of establishing pregnancy. Administration is usually stopped when sufficient follicles have matured and exogenous hCG/LH is given for induction of ovulation. The amount of hCG/LH usually amounts 5000-10000 IU. Alternatively, induction of ovulation can be performed by administration of a GnRH agonist. The agonist instead of hCG/LH is usually given on the same day in an amount sufficient to trigger ovulation. A suitable range is 10-1000 μg. Suitable agonists are e.g. buserelin, triptorelin and luprorelin. The exact regimen for administration might depend on the individual response and is finally to be decided by the clinician who treats the subject. For this reason the duration of initial ovarian stimulation with FSH alone as well as the duration of combined treatment with FSH/GnRH antagonist treatment may vary. FSH treatment usually starts at menses day 1 , 2 or 3. Ovarian stimulation with FSH alone may be continued up to 5 days in an amount of e.g. 150-225 IU. FSH is administered preferably as a recombinant protein. Treatment with GnRH antagonist may be started at the first day of FSH, but preferably such treatment starts at FSH treatment day 4 or 5. The GnRH antagonist is administered in the previously determined amount according to the invention in combination with FSH in amounts between 50 - 600 IU, preferably between 100 - 300 IU. GnRH antagonist treatment may last 2 - 14 days i.e. up to the moment whereupon the patient is treated with exogenous LH/hCG or a GnRH or GnRH agonist for ovulation induction.
According to another aspect of the invention ganirelix in an amount according to Formula I is used for the manufacture of a medicament to prevent a premature LH surge in women undergoing controlled ovarian hyperstimulation.
The pharmaceutical preparations for use according to the invention can be prepared in accordance with standard techniques such as for example are described in the standard reference, Gennaro et al. (Ed.), Remmington's Pharmaceutical Sciences, (18th ed. Mack Publishing Company, 1990, e.g. Part 8: Pharmaceutical Preparations And Their Manufacture). For the purpose of making the pharmaceutical preparations according to the invention, the active substance is mixed with or dissolved in a pharmaceutical acceptable carrier. Any conventional pharmaceutical carrier that does not interfere with performance of the active ingredient can be used in the preparations according to the present invention. Formulations may contain as common excipients sterile water or saline, alkylene glycols such as propylene glycol, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphtalenes and the like.
The pharmaceutical preparation of the antagonist may be administered parenterally. Preferably it is administered subcutaneously, particularly in the form of liquid solutions or suspensions. A typical formulation is a solution containing, in addition to the active substance in an amount as indicated above, glacial acetic acid, mannitol, and water adjusted to pH 5 with sodium hydroxide and / or hydrochloric acid. Optionally preservations such as e.g. methyl- and propylparaben or benzylalcohol can be added. The solutions can be packaged e.g. in glass vials, cartridges or in syringes.
In a further aspect of the invention there is provided a cartridge containing a sterile liquid formulation of ganirelix. As used herein a cartridge means a closed container, such as an ampoule, a vial, a bottle or a bag comprising an amount of GnRH antagonist so as to administer accurately and preferably repeatedly to a patient a dosage of GnRH antagonist according to Formula I. Thus, a cartridge may contain an amount of the liquid antagonist formulation corresponding to one or more therapeutic dosages of the antagonist. Preferably these dosages are to be applied in a single regimen. Preferably the cartridges contain an amount of GnRH antagonist sufficient for 5 administrations. The cartridges are preferably used in combination with a device making it possible to deliver adjustable dosages needed in the regimen.
In another aspect of the invention there is provided a device for administration comprising a cartridge containing a sterile liquid formulation according to the invention. A preferred device for administration is a pen-type injector, which comprise means for easy adjustment of the amount of a formulation that is to be injected. Such pen type injectors are known per se, such as for instance the well known B-D Pen (a trademark of Becton Dickinson and Company), an insulin-injection system.
Adjustable cartridges according to the invention have the advantage of accurate self-administration thereby increasing the convenience for the patients.
In yet another aspect of the invention there is provided a kit for use in controlled ovarian hyperstimulation in female patients. Such a kit comprises a GnRH antagonist in a dosage form and quantity so as to accurately administer to a patient in an amount according to Formula I, in a frequency effective to prevent a premature LH surge. In addition the kit comprises FSH in a dosage form and quantity suitable for administering in an amount and frequency effective to stimulate growth of follicles. Optionally the kit may comprise also hCG/LH or GnRH agonists in a dosage form and quantity suitable for administering in an amount and frequency effective to induce ovulation. The GnRH antagonist preferably is packaged in a cartridge. This cartridge preferably is to be used in combination with a device for administration such as a pen type injector allowing an adjustable and accurate administration of GnRH antagonist. Thus, the kit might also comprise a pen type injector system.
The invention is further explained by reference to the following Examples. Legends to the Figures
Figure 1 : LH levels were measured at the start (just before the first injection of ganirelix) and end of ganirelix treatment. The graph shows the pregnancies in relation to the various levels. LH levels are indicated in IU/L.
Figure 2: Area under the curve of ganirelix versus body weight of subjects in three pharmacokinetic studies. Circles mean protocol A; triangles mean protocol B and squares mean protocol C.
Figure 3: Chance of pregnancy versus body weight; results from Phase III efficacy study. Dots in the top of the graph correspond to pregnant subjects
(100%), while for subjects not pregnant (0%) these dots are displayed at the bottom of the graph. The spline function (chance of pregnancy vs body weight) is the resulting curve of these observations.
Examples
Example 1
LH serum levels versus pregnancy
Recombinant FSH (recFSH) treatment was started on day 2 or 3 of the menstrual cycle by a once daily SC injection. Just prior to the first injection of recFSH an hCG test was performed to exclude pregnancy, a blood sample for hormone analysis was taken and an ultrasonography (USS) was performed. During recFSH treatment day 1 through 5, the daily dose of recFSH was fixed to 150 international units (IU). On day 6 of recFSH treatment ganirelix treatment was started by daily SC administration until and including the day before the day of hCG.
During ganirelix treatment, the dose of recFSH was adjusted depending on the individual ovarian response as assessed by USS. From the first day of ganirelix i.e. from recFSH treatment day 6 onwards up to and including the day of hCG, a blood sample for hormone analysis was taken prior to drug administration. And an USS was performed, at least every two days.
LH levels were assessed by a standard LH specific assay at the Central Laboratory of the Analytisch Biochemisch Laboratorium (Assen, The Netherlands). From the data on LH levels, a plot was constructed in order to investigate the possible role of LH on pregnancy.
LH levels were measured at the start (just before the first injection of ganirelix) and end of ganirelix treatment. Figure 1 shows the pregnancies in relation to the various levels.
Clearly, from this graph, no relationship between LH serum levels and pregnancy outcome was found.
Example 2
Body Weight versus pregnancy For the body weight parameter the treated groups of example 1 were divided into categories of a 10 kg range and the pregnancy rates found for these separate categories were investigated. The results are indicated in Table 1
Figure imgf000009_0001
Table 1 : Relationship between pregnancy and body weight
From this table a relationship between the parameter body weight and clinical outcome (i.e. pregnancy) was observed.
Example 3
Body weight vs AUC
Several pharmacokinetic studies were carried out with ganirelix. Protocol A: an open-label two-way crossover study to assess the absolute bioavailablity of 0.25 mg ganirelix after single injection. Protocol B: an open-label randomized, multiple dose parallel-design study to assess the dose- proportionality and the pharmacokinetic properties of ganirelix (0.125 , 0.25 and 0.5 mg) after repeated subcutaneous administration. Protocol C: an open, randomized, two-way crossover study to establish the local tolerance and bioavailability of ganirelix after multiple subcutaneous administration (2 mg).
In all these trials A, B and C, blood samples were taken at regular intervals and the amount of ganirelix present in the blood was determined. Plots were prepared showing the amount of ganirelix as a function of time. This allowed the determination of the AUC. Next, the AUC was related to the body weight of the subjects. Results are indicated in Figure 2.
A pooled analysis of the three pharmacokinetic studies has demonstrated that the clearance of ganirelix is positively related to body weight. This is expressed in a lower area under the curve (AUC) for subjects with a higher body weight (Figure 2). Thus at the same dose level, individuals with a relatively high body weight will be exposed to lower levels of ganirelix, and individuals with a low body weight to relatively high levels. As body weight is related to ganirelix levels it can be expected that body weight influences the clinical outcome.
This hypothesis is supported by the results of a large Phase III efficacy study. At lower body weights a significant decrease in pregnancy rate was observed (see example 1 and Figure 3). These subjects have been exposed to relatively higher levels of ganirelix. The pregnancy rate therefore could be optimized in these individuals by adjustment of the dose, according to their body weights.
Example 4
Dose finding
Pharmaco-statistical models have been set up to describe the influence of body weight on the effectiveness of ganirelix for both the prevention of LH- rises and pregnancy outcome. Spline functions have been applied to give the best and assumptionless mathematical description of the available data. Using these models optimal doses with respect to the prevention of LH-rises and pregnancy have been determined for different body weights.
Figure imgf000010_0001
Table 2: Relationship between body weight and optimal dose
Results are indicated in Table 2. Linear regression shows that these dosages can be given by the formula: (5.5*BW -166) ± 7% wherein BW is the body weight in kg.

Claims

Claims
1. In the treatment of female patients undergoing controlled ovarian hyperstimulation comprising administration of exogenous FSH and a
GnRH antagonist, the improvement comprising administering said antagonist in an amount depending on the body weight of the patient wherein the relationship between body weight (BW in kg) and GnRH antagonist dosage (in micrograms) is defined by the Formula: (5.5*BW -166) ± 7% (Formula I).
2. Treatment according to claim 1 wherein said GnRH antagonist is ganirelix.
3. A cartridge containing a sterile liquid GnRH antagonist containing formulation in an amount so as to accurately administer to a patient an amount according to Formula 1 of claim 1 or 2.
4. The cartridge of claim 3 wherein the GnRH antagonist is ganirelix.
5. A device for administration of an adjustable sterile liquid GnRH antagonist comprising a cartridge according to claim 3 or 4.
6. Use of GnRH antagonist for the manufacture of a medicament to prevent a premature LH surge in female patients undergoing controlled ovarian hyperstimulation, the method of controlling ovarian hyperstimulation comprising administering to the patient a GnRH antagonist in an amount depending on the body weight of the patient wherein the relationship between body weight (BW in kg) and GnRH antagonist dosage (in micrograms) is defined by the Formula:
(5.5*BW -166) ± 7%.
7. Use of GnRH antagonist according to claim 6 wherein said GnRH antagonist is ganirelix.
8. A kit for use in controlled ovarian hyperstimulation in female patients comprising a GnRH antagonist in a dosage form and quantity so as to administer accurately in an amount according to Formula I of claim 1 and a frequency effective to prevent a premature LH surge; FSH in a dosage form and quantity so as to administer in an amount and frequency effective to stimulate growth of follicles; and LH/hCG or a GnRH agonist in an dosage form and quantity so as to administer in an amount and frequency effective to induce ovulation.
9. Kit according to claim 8 wherein the antagonist is ganirelix.
10. Kit according to claims 8 or 9 wherein the antagonist is packaged in a cartridge according to claim 4.
11. Kit according to claims 9-10 comprising a device for administration of an adjustable sterile liquid amount of the antagonist.
PCT/EP2000/005643 1999-06-23 2000-06-19 Gonadotropin releasing hormone antagonist WO2001000227A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2001505935A JP2003503358A (en) 1999-06-23 2000-06-19 Gonadotropin-releasing hormone antagonists
CA002341974A CA2341974A1 (en) 1999-06-23 2000-06-19 Gonadotropin releasing hormone antagonist
AU62638/00A AU6263800A (en) 1999-06-23 2000-06-19 Gonadotropin releasing hormone antagonist
EP00949193A EP1140132A1 (en) 1999-06-23 2000-06-19 Gonadotropin releasing hormone antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP99202027 1999-06-23
EP99202027.1 1999-06-23

Publications (1)

Publication Number Publication Date
WO2001000227A1 true WO2001000227A1 (en) 2001-01-04

Family

ID=8240349

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2000/005643 WO2001000227A1 (en) 1999-06-23 2000-06-19 Gonadotropin releasing hormone antagonist

Country Status (6)

Country Link
EP (1) EP1140132A1 (en)
JP (1) JP2003503358A (en)
AR (1) AR024438A1 (en)
AU (1) AU6263800A (en)
CA (1) CA2341974A1 (en)
WO (1) WO2001000227A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003103770A2 (en) * 2002-06-07 2003-12-18 Pantarhei Bioscience B.V. Method of controlled ovarian hyperstimulation and pharmaceutical kit for use in such method
WO2012042381A1 (en) * 2010-09-29 2012-04-05 Ferring B.V. Composition for controlled ovarian stimulation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998055470A1 (en) * 1997-06-05 1998-12-10 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
WO1998058657A1 (en) * 1997-06-20 1998-12-30 Akzo Nobel N.V. Gonadotropin releasing hormone antagonist

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998055470A1 (en) * 1997-06-05 1998-12-10 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
WO1998058657A1 (en) * 1997-06-20 1998-12-30 Akzo Nobel N.V. Gonadotropin releasing hormone antagonist

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
OBERYÉ J ET AL: "Local tolerance, pharmacokinetics, and dynamics of ganirelix (Orgalutran) administration by Medi-Jector compared to conventional needle injections", HUMAN REPRODUCTION, vol. 15, no. 2, February 2000 (2000-02-01), pages 245 - 249, XP000876537 *
PAULSON R J ET AL: "Addition of a gonadotropin releasing hormone ( GnRH ) antagonist and exogenous gonadotropins to unstimulated in vitro fertilization (IVF) cycles: physiologic observations and preliminary experience.", JOURNAL OF ASSISTED REPRODUCTION AND GENETICS, (1994 JAN) 11 (1) 28-32., XP000876515 *
THE GANIRELIX DOSE-FINDING STUDY GROUP: "A double-blind, randomized, dose-finding study to assess the efficacy of the gonadotrophin-releasing hormone antagonist ganirelix (Org 37462) to prevent premature luteinizing hormone surges in women undergoing ovarian stimulation with recombinant follicle stimulating hormone (Puregon)", HUMAN REPRODUCTION, vol. 13, no. 11, 1998, pages 3023 - 3031, XP000876538 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003103770A2 (en) * 2002-06-07 2003-12-18 Pantarhei Bioscience B.V. Method of controlled ovarian hyperstimulation and pharmaceutical kit for use in such method
WO2003103770A3 (en) * 2002-06-07 2004-04-22 Ares Trading Sa Method of controlled ovarian hyperstimulation and pharmaceutical kit for use in such method
EA009371B1 (en) * 2002-06-07 2007-12-28 Арес Трейдинг С.А. Method of controlled ovarian hyperstimulation pharmaceutical kit for use in such method
US7815912B2 (en) 2002-06-07 2010-10-19 Ares Trading, S.A. Method of controlled ovarian hyperstimulation and pharmaceutical kit for use in such method
WO2012042381A1 (en) * 2010-09-29 2012-04-05 Ferring B.V. Composition for controlled ovarian stimulation
EP2842567A1 (en) * 2010-09-29 2015-03-04 Ferring B.V. Composition for controlled ovarian stimulation
EP2621517B1 (en) 2010-09-29 2015-06-17 Ferring B.V. Composition for use in treating infertility
US9320778B2 (en) 2010-09-29 2016-04-26 Ferring B.V. Method for controlled ovarian stimulation with combined FSH and hCG
US10064920B2 (en) 2010-09-29 2018-09-04 Ferring B.V. Method for controlled ovarian stimulation using FSH and hCG

Also Published As

Publication number Publication date
AU6263800A (en) 2001-01-31
JP2003503358A (en) 2003-01-28
CA2341974A1 (en) 2001-01-04
AR024438A1 (en) 2002-10-02
EP1140132A1 (en) 2001-10-10

Similar Documents

Publication Publication Date Title
ZELEZNIK et al. Ovarian responses in macaques to pulsatile infusion of follicle-stimulating hormone (FSH) and luteinizing hormone: increased sensitivity of the maturing follicle to FSH
de Ziegler et al. Suppression of the ovary using a gonadotropin releasing-hormone agonist prior to stimulation for oocyte retrieval
US9107871B2 (en) LHRH—antagonists in the treatment of fertility disorders
EP0994718B1 (en) Gonadotropin releasing hormone antagonist
JP2005501918A (en) Use of hCG in controlled superovulation stimulation
Wallach et al. Ovulation induction with pulsatile gonadotropin-releasing hormone: technical modalities and clinical perspectives
Bremner et al. Gonadotropin-releasing hormone antagonist plus testosterone: a potential male contraceptive
JPH05507493A (en) Combination therapy with GnRH antagonists and GnRH to modulate gonadotropin levels in mammals
Balasch et al. Pituitary-ovarian suppression by the standard and half-doses of D-Trp-6-luteinizing hormone-releasing hormone depot
JP2005501919A (en) Use of hCG and LH in controlled superovulation stimulation
US20030092628A1 (en) Gonadotropin releasing hormone antagonist
US20050049199A1 (en) Use of lh in controlled ovarian hyperstimulation
EP1140132A1 (en) Gonadotropin releasing hormone antagonist
AU2002331939A1 (en) Use of LH in controlled ovarian hyperstimulation
Lemay Clinical appreciation of LHRH analogue formulations
Bider et al. Pituitary and ovarian suppression rate after high dosage of gonadotropin-releasing hormone agonist
Neves-e-Castro et al. Use of luteinizing hormone-releasing hormone analogs in non-neoplastic gynecologic conditions
CA2200541C (en) Lhrh-antagonists in the treatment of fertility disorders
Chaussain et al. Treatment of precocious puberty with a long-acting preparation of D-Trp6-LHRH
Van der Spuy et al. Early clinical studies with LHRH antagonists in women
Bhathena Optimising Protocols for Induction of Ovulation
Baird Manipulation of the ovarian cycle
Kuehl et al. Incorporation of a GnRH agonist, leuprolide acetate, into regimens with exogenous gonadotropins to produce ovarian stimulation and ovulation in the nonpregnant squirrel monkey
Engel et al. Luteinizing hormone-releasing hormone antagonists in gynecology
CN1199642A (en) LHRH-antagonists in treatment of fertility disorders

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AU BA BB BG BR CA CN CU CZ EE GE HR HU ID IL IN IS JP KP KR LC LK LR LT LV MG MK MN MX NO NZ PL RO RU SG SI SK SL TR TT UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

ENP Entry into the national phase

Ref document number: 2341974

Country of ref document: CA

Ref country code: CA

Ref document number: 2341974

Kind code of ref document: A

Format of ref document f/p: F

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 62638/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2000949193

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09763434

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 2000949193

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2000949193

Country of ref document: EP