WO2009134713A1 - Compositions and methods for male contraception - Google Patents

Compositions and methods for male contraception Download PDF

Info

Publication number
WO2009134713A1
WO2009134713A1 PCT/US2009/041795 US2009041795W WO2009134713A1 WO 2009134713 A1 WO2009134713 A1 WO 2009134713A1 US 2009041795 W US2009041795 W US 2009041795W WO 2009134713 A1 WO2009134713 A1 WO 2009134713A1
Authority
WO
WIPO (PCT)
Prior art keywords
cdb
androgen receptor
receptor agonist
administered
composition
Prior art date
Application number
PCT/US2009/041795
Other languages
French (fr)
Inventor
Joseph S. Podolski
Ronald D. Wiehle
Original Assignee
Repros Therapeutics Inc.
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 Repros Therapeutics Inc. filed Critical Repros Therapeutics Inc.
Publication of WO2009134713A1 publication Critical patent/WO2009134713A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • A61K31/5685Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone having an oxo group in position 17, e.g. androsterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/16Masculine contraceptives

Definitions

  • the present invention relates to compositions and methods for male contraception. More specifically, the present invention relates to compositions comprising one or more progesterone antagonists for inhibiting spermatogenesis.
  • the instant invention provides methods for inhibiting spermatogenesis comprising administering to a male mammal, an androgen receptor agonist and a composition comprising one or more progesterone antagonists in a collective amount effective to inhibit spermatogenesis.
  • the methods further comprise the administration of a progestin and/or a gonadotropin-releasing hormone (GnRH) agonist or antagonist.
  • GnRH gonadotropin-releasing hormone
  • the male mammal may be a human male.
  • the progesterone antagonist may be a pure antiprogestin or a selective progesterone receptor modulator (SPRM).
  • SPRM selective progesterone receptor modulator
  • the progesterone antagonist has low affinity for glucocorticoid receptor.
  • the instant invention provides methods of contraception comprising administering to a male mammal, an androgen receptor agonist and a composition comprising one or more progesterone antagonists in a collective amount effective to achieve oligospermia or, preferably, azoospermia in the male mammal.
  • the methods further comprise the administration of a progestin and/or a gonadotropin-releasing hormone (GnRH) agonist or antagonist.
  • the male mammal may be a human male.
  • the progesterone antagonist may be a pure antiprogestin or a selective progesterone receptor modulator (SPRM).
  • the progesterone antagonist has low affinity for glucocorticoid receptor.
  • administration of the progesterone antagonist is for a period of time effective to inhibit spermatogensis in the male mammal.
  • the term "effective dosage” means an amount of the composition's active component sufficient to achieve the desired effect which may be, e.g., inhibition of spermatogenesis.
  • selective progesterone receptor modulators means compounds that affect functions of progesterone receptor in a tissue-specific manner.
  • the compounds act as progesterone receptor antagonists in some tissues (for example, in the uterus) and as progesterone receptor agonists in other tissues.
  • treat or “treatment” refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • progesterone agonist means a compound that binds to a progesterone receptor and mimics the action of the natural hormone.
  • progesterone antagonist means a compound that binds to a progesterone receptor and inhibits the effect of progesterone.
  • the present invention relates to the use of progesterone antagonists at doses effective to inhibit spermatogenesis in a male mammal when administered simultaneously, separately or sequentially with an androgen receptor agonist.
  • compositions of the invention make them surprisingly useful for inhibiting spermatogenesis.
  • a male mammal with a need or desire to inhibit spermatogenesis is administered a composition comprising a progesterone antagonist in an amount effective to inhibit spermatogenesis.
  • the male mammal may be a human male.
  • the present invention provides a method of inhibiting spermatogenesis comprising administering to a male mammal an androgen receptor agonist and a composition comprising at least one progesterone antagonist, wherein the androgen receptor agonist and progesterone antagonist are administered in a collective amount effective to inhibit spermatogenesis.
  • the progesterone antagonist may be administered simultaneously, separately or sequentially with the administration of the androgen receptor agonist. Administration of exogenous androgens is known to inhibit spermatogenesis.
  • Androgen receptor agonists may be natural or synthetic and include, without limitation: androgen hormones such as testosterone, dihydrotestosterone and 5 ⁇ -androstanediol; testosterone undecanoate, testosterone enanthate, testosterone esters, testosterone proprionate, mesterolone, danazol and gestrinone.
  • a preferred androgen receptor agonist is testosterone undecanoate.
  • Selective androgen receptor agonists may also be used for their agonist activity.
  • the dosage of the androgen receptor agonist may be any dosage known in the art to inhibit spermatogenesis.
  • IM intramuscular
  • a composition comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist relative to that known in the art to inhibit spermatogenesis.
  • a "suboptimal" dose of androgen receptor agonist is any such reduced dosage of androgen receptor agonist lower than would result in optimal inhibition of spermatogenesis. Reducing the dose of androgen receptor agonist is expected to reduce androgenic side effects and should therefore result in greater patient acceptability and compliance.
  • the present invention provides a method for inhibiting spermatogenesis comprising the administration of a composition comprising one or more progesterone antagonists to a male in an amount effective to inhibit spermatogenesis when combined with a suboptimal dose of androgen receptor agonist, preferably testosterone undecanoate.
  • the composition may be administered simultaneously, separately or sequentially with the androgen receptor agonist.
  • the present invention provides methods of enhancing the inhibition of spermatogenesis in a male mammal, comprising administering to the mammal a composition comprising at least one progesterone antagonist and an androgen receptor agonist.
  • the instant invention provides methods of contraception comprising administering to a male mammal an androgen receptor agonist and a composition comprising at least one progesterone antagonist, wherein the androgen receptor agonist and progesterone antagonist are administered in a collective amount effective to achieve oligospermia or, preferably, azoospermia in the male mammal.
  • the progesterone antagonist may be administered simultaneously, separately or sequentially with the administration of the androgen receptor agonist.
  • Normal sperm counts in human males are generally from about 20 million to about 120 million sperm per milliliter of semen.
  • compositions of the invention are of sufficient dosage and for a sufficient administration period to achieve oligospermia or preferably, azoospermia.
  • Oligospermia is defined herein as a concentration of sperm less than 20 million per milliliter of semen.
  • Azoospermia is defined herein as a lack of measurable level of sperm in the semen.
  • administering decreases the concentration of sperm below about 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, and preferably below about 1 million sperm per milliliter of semen in the male mammal.
  • concentration of sperm is decreased below 1 million per milliliter of semen, contraceptive protection is provided to a Pearl index rating of 1.4 per 100 couple- years.
  • Methods of the invention may comprise administering a composition comprising an effective amount of a progesterone antagonist for an administration period of least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or more days.
  • the composition may also be administered for an administration period of least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more months.
  • the composition may also be administered for an administration period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years.
  • the composition may be administered daily or periodically such as every other day, every other month, and the like.
  • the composition may also be administered intermittently.
  • the composition may be administered for an administration period of 1, 2, 3, 4, 5 or more months, followed by a period of discontinuance, followed by an administration period of 1, 2, 3, 4, 5 or more months, and so on.
  • intermittent administration it is meant a period of administration of a therapeutically effective dose of progesterone antagonist, followed by a time period of discontinuance, which is then followed by another administration period and so forth.
  • periodic of discontinuance or “discontinuance period” it is meant a discontinuing of the daily, weekly, monthly or therebetween administration of progesterone antagonist.
  • the time period of discontinuance may be longer or shorter than the administration period but is always longer than the dosing interval during the administration period.
  • the discontinuance period is at least 2 days, at least 8 days or at least 32 days, respectively.
  • the discontinuance period may be at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32 or more days.
  • combination therapy comprising the administration of a composition comprising at least one progesterone anatagonist and one or more androgen receptor agonists further comprises the administration of one or more anti-gonadotrophic agents.
  • the antigonadotrophic agents are progestins.
  • Administration of exogenous progestin is known to reduce the amount of exogenous androgen receptor agonist necessary to inhibit spermatogenesis.
  • Progestins are defined herein as natural or synthetic progestational substances that mimic some or all of the actions of progesterone.
  • Nonlimiting examples of progestins include derivatives of 19-nortestosterone, such as oestranes and gonanes, derivatives of 17 ⁇ - acetoxyprogesterone (pregnanes), and 17 ⁇ -hydroxyprogesterone derivatives such as medroxyprogesterone acetate (MPA).
  • Examples of oestranes include norethindrone and its acetates, and ethynodiol diacetate.
  • Examples of gonanes include norgestrel, levonorgestrel and derivatives of levonorgestrel such as desogestrel, norgestimate and gestodene.
  • a preferred progestin is levonorgestrel.
  • the dosages of progestin and androgen receptor agonist may be any dosage, the combination of which is known in the art to inhibit spermatogenesis.
  • medroxyprogesterone 300 mg
  • IM injections of 1000 mg testosterone undecanoate every eight weeks
  • 150 and 300 mg medroxyprogesterone to IM injections of 1000 mg testosterone undecanoate every eight weeks increased the incidence of azoospermia and severe oligospermia during the 24-week treatment period from 80% to 100%.
  • compositions comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist and/or progestin relative to that known in the art to inhibit spermatogenesis, to achieve oligospermia or preferably to achieve azoospermia.
  • the antigonadotrophic agents are gonadotropin- releasing hormone (GnRH) agonists or antagonists.
  • GnRH gonadotropin- releasing hormone
  • Administration of exogenous GnRH agonists/antagonists is known to reduce the amount of exogenous androgen receptor agonist necessary to inhibit spermatogenesis.
  • GnRH agonists include nafarelin, buserelin, leuprolide, triptorelin, goserelin, [DLys 6 ]GnRH, [DAIa 6 ] GnRH and the like.
  • GnRH antagonists include histrelin, abarelix and those found in U.S. Patent Nos.
  • a preferred GnRH agonist is leuprolide.
  • the dosages of GnRH agonist/antagonist and androgen receptor agonist may be any dosage, the combination of which is known in the art to inhibit spermatogenesis.
  • a GnRH antagonist coadministered every 2 weeks with IM testosterone enanthate resulted in azoospermia in 88% of men by week 10.
  • compositions comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist and/or GnRH agonist/antagonist relative to that known in the art to inhibit spermatogenesis, to achieve oligospermia or preferably to achieve azoospermia.
  • the progesterone antagonist exhibits reduced affinity for the glucocorticoid receptor. More preferably, the binding affinity of the progesterone antagonist for the progesterone receptor is at least 1.5 times greater than the binding affinity of the progesterone antagonist for the glucocorticoid receptor.
  • any known progesterone antagonist with characteristics of the compounds described above can be used by an artisan practicing the instant invention.
  • Particularly useful compounds include those disclosed in U.S. Patent No. 6,900,193, hereby incorporated by reference in its entirety, as well as those disclosed in U.S. Patent No. 6,861,415, hereby incorporated by reference in its entirety, that are 21- substituted 19-norpregnanes with a general formula:
  • X may be, for example alkyl, alkenyl, alkynyl, hydrogen, halo, monoalkylamino or dialkylamino, such as N,N-dimethylamino;
  • Ri may be, for example O, NOH or NO-methyl
  • R 2 may be, for example hydrogen or acetyl
  • R3 may be, for example methyloxy, formyloxy, acetoxy, acyloxy, S-alkoxy, acetyltheonyl, glycimate, vinyl ether, acethyloxymethyl, methyl carbonate, halogens, methyl, hydroxy, and ethyloxy.
  • 21 -substituted 19-norpregnanes include, but are not limited to, the following 24 compounds disclosed below.
  • CDB-4247 (21-propio[[l]]nyloxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4361 (21 -vinyl ether- 17 ⁇ -acetoxy- 1 l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4059 (21-acetoxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4124 (21-methoxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4031 (21 -bromine- 17 ⁇ -acetoxy- 11 ⁇ -(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-3876 (21 -chlorine- 17 ⁇ -acetoxy- 1 l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4058 (21-flourine-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4030 (21 -methyl- 17 ⁇ -acetoxy- 1 l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4152 21 -hydroxy- 17 ⁇ -acetoxy- 1 l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione with the following structural formula:
  • CDB-4167 21-ethyloxy-17a-acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione with the following structural formula:
  • CDB-4101 (21 -methoxythio- 17 ⁇ -acetoxy- 11 ⁇ -(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4110 (21-acetonide-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4111 (21 -BMD- 17 ⁇ -acetoxy- 11 ⁇ -(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4125 (21-(Cyp*-hydroxy)-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4205 (3 -hydroxy amino-21 -methoxy- 17 ⁇ -acetoxy- 11 ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4206 (3-hydroxyamino-21-acetoxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4226 (3-hydroxyamino-21-ethyloxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4262 S-methoxyamino ⁇ l-ethyloxy-lVa-acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione with the following structural formula:
  • CDB-4223 (21-methylthio-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4119 (4-benzoin-21 -acetylthio- 17 ⁇ -acetoxy- 11 ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4239 (4-benzoin-21-methoxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4306 (21-glycinate-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4352 (ll-cyanothio-Ha-acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)-19 norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • CDB-4362 (21-methoxyacetyl-17 ⁇ -acetoxy-l l ⁇ -(4 N, N-dimethylaminophenyl)- 19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
  • any progesterone antagonist may be used in the practice of the present invention.
  • the progesterone antagonist has one or more of the following characteristics: low antiglucocorticoid activity, minimal estrogenic and anti-estrogenic activities, and does not substantially elevate progesterone levels.
  • Antiprogestins which may be useful in the invention include, without limitation, asoprisnil (benzaldehyde, 4-[(l l ⁇ ,17 ⁇ )-17-methoxy-17-(methoxymethyl)- 3-oxoestra-4,9-dien-l l-yl]-l-(E)-oxim; J867), its metabolite J912 (4-[17 ⁇ -Hydroxy- 17 ⁇ -(methoxymethyl)-3-oxoestra-4,9-dien-l l ⁇ -yl]benzaldehyd-(lE)-oxim), and other compounds described in DE 43 32 283 and DE 43 32 284; CDB-2914 (17 ⁇ - acetoxy- 11 ⁇ -(4-N,N-dimethylaminophenyl)- 19-norpregna-4,9-dien-3 ,20-dione) and other compounds described in Stratton et al, 2000, Hu.
  • antiprogestins that may be useful in the invention include, without limitation, (6 ⁇ ,l l ⁇ ,17 ⁇ )-l l-(4-dimethylaminophenyl)-6-methyl-4',5'- dihydrospiro[estra-4,9-diene-17,2'(3'H)-furan]-3-one (ORG-31710) and other compounds described in U.S. Patent No.
  • antiprogestins that may be useful in the invention include, without limitation, mifepristone (l l ⁇ -[p-(Dimethylamino)phenyl]-17 ⁇ -hydroxy-17- (l-propynyl)estra-4,9-dien-3-one; RU 486) and other compounds described in U.S.
  • the 11 ⁇ -substituted 19-norsteroids such as 1 l ⁇ -(4-Methoxyphenyl)-17 ⁇ -hydroxy-17 ⁇ -ethynyl-4,9-estradien-3-one described in Belagner et al., 1981, Steroids 37:361-382; the 11 ⁇ -aryl-4-estrenes such as (Z)-I l ⁇ - [(4-Dimethylamino)phenyl)] - 17 ⁇ -hy droxy- 17 ⁇ -(3 -hydroxy- 1 -propeny l)estr-4-en-3 - one described in U.S. Patent No.
  • Patent No. 5,292,878 (Z)- 1 lbeta, 19-[4-(3-Pyridinyl)-o-phenylene]- 17beta-hydroxy-l 7 ⁇ -[3-hydroxy- 1 - propenyl]-4-androsten-3-one and other compounds described in U.S. Patent No. 5,439,913; the 13-alkyl-l l-beta-phenyl gonanes such as l lbeta-[4-(l- methylethenyl)phenyl]- 17 ⁇ -hydroxy- 17beta-(3-hydroxypropyl)- 13 ⁇ -estra-4,9-dien- 3-one described in U.S. Patent No.
  • Patent No. 5,244,886 the 19,11 -beta-bridged steroids described in U.S. Patent Nos: 5,095,129, 5,446,178, 5,478,956 and 5,232,915; the 1-arylsulphonyl, arylcarbonyl and l-arylphosphonyl-3-phenyl-l,4,5,6-tetrahydropyridazines described in U.S. Patnet No. 5,684,151; the 1-arylsulphonyl, arylcarbonyl and arylthiocarbonyl pyridazino derivatives described in U.S. Patent No.
  • the progesterone antagonist is the antiprogestin/SPRM CDB-4124 (21-methoxy-17 ⁇ -acetoxy-l l ⁇ -(4 N, N- dimethylaminophenyl)- 19-norpregna-4,9-diene-3 ,20-dione).
  • compositions of the instant invention are preferably suitable for prolonged administration.
  • the progesterone antagonist preferably has only low glucocorticoid receptor binding activity and therefore, does not substantially interfere with functions of glucocorticoid receptor.
  • administration of the compositions may have reduced side effects, such as mood swings, fatigue and weight loss, typically found when antiprogestins with a high affinity for glucocorticoid receptor are used.
  • the instant invention teaches methods that can be used for identifying compounds that possess selective progesterone receptor binding activity. These methods include receptor binding and in vivo bioassays such as anti- McGinty, anti-Clauberg, glucocorticoid, estrogenic, androgenic, anti-glucocorticoid (AG), anti-estrogen, and anti-androgen activities as well as post-coital and anti- ovulatory activities where in the leading compounds of the instant invention are used as a reference.
  • receptor binding and in vivo bioassays such as anti- McGinty, anti-Clauberg, glucocorticoid, estrogenic, androgenic, anti-glucocorticoid (AG), anti-estrogen, and anti-androgen activities as well as post-coital and anti- ovulatory activities where in the leading compounds of the instant invention are used as a reference.
  • the instant invention teaches that the potential SPRMs can be also analyzed for their effect on transcriptional activity in human cells.
  • this analysis can furnish information about (1) SPRM 's interaction with receptor, (2) interaction of the activated receptor with other transcription factors, (3) activation of a transcriptional complex at a progesterone response element (PRE); and ultimately its effect on gene expression.
  • plasmid expressing the hPR-B can be cotransfected with any reporter known to a person skilled in the relevant art under the PRE-dependent promoter into HeLa, HepG2 or T47D cells.
  • the reporters may include, but are not limited to, luciferase, beta-galactosidase, green fluorescent protein, red fluorescent protein or yellow fluorescent protein.
  • the cells are treated with either a test compound or one of the disclosed in this application SPRMs that serves as a positive control. Following treatment, cells are assayed for reporter expression.
  • the instant invention teaches that prospective SPRMs can be tested for their ability to oppose dexamethasone-induced cell death in human lymphocytic cell line CEM-7 and compared to effects of SPRMs disclosed in the instant specification.
  • dexamethasone can be added at a concentration that results in cell death.
  • the cells are then treated with either RU486, one of SPRMs of the instant invention or a test compound at concentrations between 10 ⁇ 6 and l0 ⁇ 8 M.
  • Progesterone antagonist compounds that may be used in accordance with the present invention can be synthesized using synthetic chemistry techniques known in the art such as those disclosed in U.S. Patent No. 6,861,415.
  • compositions of the invention comprise one or more progesterone antagonists or pharmaceutically acceptable salts thereof.
  • the salt compound obtained may be either in neutral or salt form. Salt forms include hydrates and other solvates and also crystalline polymorphs. Both the free base and the salts of these end products may be used in accordance with the invention.
  • Acid addition salts may in a manner known per se be transformed into the free base using basic agents such as alkali or by ion exchange.
  • the free base obtained may also form salts with organic or inorganic acids.
  • acids which form suitably pharmaceutically acceptable salts.
  • examples of such acids are hydrochloric acid, sulfuric acid, phosphoric acid, nitric acid, aliphatic acid, alicyclic carboxylic or sulfonic acids, such as formic acid, acetic acid, propionic acid, succinic acid, glycolic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, glucuronic acid, fumaric acid, maleic acid, hydroxymaleic acid, pyruvic acid, aspartic acid, glutamic acid, p-hydroxybenzoic acid, embonic acid, ethanesulfonic acid, hydroxyethanesulfonic acid, phenylacetic acid, mandelic acid, alogenbensenesulfonic acid, toluenesulfonic acid, galactaric acid, galacturonic acid or naphthalenesulfonic acid.
  • Base addition salts may also be used in accordance with the invention and may be prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkali earth metals or organic amines. Examples of metals used as cations are sodium, potassium, calcium, magnesium and the like.
  • Suitable amines are amino acids such as lysine, choline, diethanolamine, ethylenediamine, N-methylglucamine and the like [00047]
  • the compounds of the instant invention can be administered to a patient via any conventional route where the progesterone antagonist is active.
  • a progesterone antagonist of the instant invention can be administered orally, parenterally, sublingually, transdermally, rectally, transmucosally, topically, via inhalation, via buccal administration, or combinations thereof.
  • Parenteral administration includes, but is not limited to, intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, intrathecal, intraarticular, intracisternal and intraventricular.
  • the administration form can be a tablet, capsule, pill, nasal mist, aerosol, pellet, implant (or other depot) and the like.
  • a therapeutically effective amount of the composition required for use in therapy may vary depending on the particular compound employed, the mode of administration, the severity of the condition being treated, the length of time that activity is desired, among other factors, and is ultimately determined by the attendant physician.
  • doses employed for human treatment typically are in the range of about 0.001 mg/kg to about 500 mg/kg per day, for example about 1 ⁇ g/kg to about 1 mg/kg per day or about 1 ⁇ g/kg to about 100 ⁇ g/kg per day.
  • the total daily dosage is from about 1 to 100 mg, preferably from about 2 to 80 mg.
  • the dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the desired dose may be conveniently administered in a single dose, or as multiple doses administered at appropriate intervals, for example as two, three, four or more subdoses per day.
  • a composition of the invention may be administered to a subject to provide the subject with a progesterone antagonist in an amount of about 1 ⁇ g/kg to about 1 mg/kg body weight, for example about 1 ⁇ g/kg, about 25 ⁇ g/kg, about 50 ⁇ g/kg, about 75 ⁇ g/kg, about 100 ⁇ g/kg, about 125 ⁇ g/kg, about 150 ⁇ g/kg, about 175 ⁇ g/kg, about 200 ⁇ g/kg, about 225 ⁇ g/kg, about 250 ⁇ g/kg, about 275 ⁇ g/kg, about 300 ⁇ g/kg, about 325 ⁇ g/kg, about 350 ⁇ g/kg, about 375 ⁇ g/kg, about 400 ⁇ g/kg, about 425 ⁇ g/kg, about 450 ⁇ g/kg, about 475 ⁇ g/kg, about 500 ⁇ g/kg, about 525 ⁇ g/kg, about 550 ⁇ g/kg, about 575
  • Solid carriers may include starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while liquid carriers may include sterile water, polyethylene glycols, non-ionic surfactants and edible oils such as corn, peanut and sesame oils, as are appropriate to the nature of the active ingredient and the particular form of administration desired.
  • Flavoring agents, coloring agents, preserving agents, and antioxidants, for example, vitamin E and ascorbic acid may be included in preparations as well. Under ordinary conditions of storage and use, the preparations may contain a preservative to prevent the growth of microorganisms.
  • compositions of the instant invention can be formulated into tablets in a tablet press by using techniques well-known to an artisan skilled in the relevant field.
  • the active ingredients according to the invention can also be pressed separately into two-layer tablets.
  • tablets may include androgen receptor agonists, GnRH agonists, GnRH antagonists, progestins or any combination thereof as one of the active ingredients.
  • Compositions of the instant invention can also be formulated as an oily solution.
  • Patients undergoing treatments with the compositions of the instant invention should be monitored routinely for their glucocorticoid levels.
  • the following non-limiting examples are provided to aid in understanding the teachings of the instant invention.
  • All patents, patent applications and publications referenced herein are hereby incorporated by reference herein to the fullest extent allowed under the law.
  • Example 1 Formulations of The Instant Invention Can Be Prepared As Tablets.
  • Example 2 Compounds of the Instant Invention May Have Only Weak Antiglucocorticoid Receptor Binding Activity.
  • rbPR rabbit progesterone receptor
  • rbGR glucocorticoid receptor
  • TEGMD buffer 10 mM Tris, pH 7.2, 1.5 mM EDTA, 0.2 mM sodium molybdate, 10% glycerol, 1 mM DTT
  • the cytosol was incubated with 6 nM l,2-[ 3 H]progesterone (50.0 Ci/mmole) and competitors were added at concentrations from 2 to 100 nM.
  • the cytosol was incubated with 6 nM 6,7-[ 3 H]-dexamethasone (40 Ci/mmol) and test compounds were added at concentrations from 20 to 100 nM. After overnight incubation at 4 C, bound and unbound [ 3 H] steroids were separated by addition of dextran-coated charcoal and centrifugation at 2100 x g for 15 min at 4 C.
  • the standards for the PR and GR assays were unlabeled progesterone and dexamethasone, respectively.
  • the results of these experiments are summarized in Table 1, as a ratio of the relative binding affinities of each compound for the rbPR and rbGR receptors (rbPR/rbGR). This differential reflects the relative activity of a compound in a cell or tissue that possesses the two receptors and the requisite transcriptional co factors.
  • mice underwent sterile abdominal surgery to ligate a 3-4 cm segment of both uterine horns.
  • the test compound in appropriate solvent was injected intraluminally into the ligated segment of one uterine horn and vehicle alone into the other.
  • a stimulating dose of progesterone (267 ⁇ g/day) was administered subcutaneously to each rabbit daily for the next three days to induce endometrial proliferation. All animals were sacrificed at day 10 for removal of the uterus where a segment central to the ligatures was removed and fixed in 10% neutral buffered formalin and submitted for histological processing. Five micron sections stained with hematoxylin and cosin were evaluated microscopically for the degree of endometrial glandular proliferation.
  • the percent inhibition of endometrial proliferation for each rabbit was calculated and the mean of the group of five animals recorded.
  • immature female rabbits received a subcutaneous injection of 5 ⁇ g estradiol in 10% ethanol/sesame oil daily for 6 consecutive days.
  • animals received progesterone by subcutaneous injection (160 ⁇ g/day) and the experimental compound in appropriate vehicle orally or subcutaneously for five consecutive days.
  • progesterone only. Twenty-four hours after the last dose, all animals were sacrificed for removal of the uterus which was cleaned of all fat and connective tissue, weighed to the nearest 0.2 mg and placed in 10% neutral buffered formalin for subsequent histological processing.
  • CDB-4124 and CDB-4059 have strong antiprogestin activity in the rabbit uterus in comparison to RU 486 and CDB-2914. Both compounds lack estrogenic, androgenic, anti- estrogenic, and anti-androgenic activities. Both compounds possess minimal anti-glucocorticoid receptor activity, a feature that distinguishes them from RU 486 and CDB-2914 which are moderately active in glucocorticoid receptor binding. In these assays, CDB-4124 performed slightly better than CDB-4059
  • SPRM rbPR/rbGR rabbit Biol SPRM rbPR/rbGR rabbit Biol.
  • rats treated with RU 486 at 10 mg/kg showed no significant difference in the levels of Cortisol.
  • CDB-4059 at the same dose levels had significantly higher levels of serum Cortisol than rats from a control group.
  • Cortisol levels can be explained by assuming that after 21 days of chronic dosing, a rat liver was able to metabolize RU 486 better than either of the two CDB compounds.
  • Corticosterone is the most abundant glucocorticoid in rats.
  • the effects of the SPRMs on Cortisol may be secondary to strong effects on corticosterone.
  • the levels of corticosterone were measured in groups, which showed the strongest changes in Cortisol levels, such as groups treated with CDB-4124 at 20 mg/kg or 10mg/kg.
  • the following groups were also assayed: a group that received 20 mg/kg CDB-4124 plus 10 mg/kg progesterone, a group that received 10 mg/kg CDB-4124 plus 10 mg/kg progesterone, a group that received 10 mg/kg RU 486, a group that received 10 mg/kg of progesterone alone, a control group.
  • the levels of corticosterone were 10-40 times higher than the levels of Cortisol. However, almost no difference between groups with respect to mean corticosterone levels was observed.
  • CDB-4124 Although no strong effect of CDB-4124 on the primary glucocorticoid of the rat was found, nevertheless, for safety reasons, patients given CDB-4124 or CDB- 4059 in Phase I clinical trials should be monitored for possible anti-glucocorticoid effects including a possible increase in serum Cortisol, corticosterone, or ACTH.
  • testicular parenchyma are used for determining the number of advanced (steps 17-19) spermatids by the homogenization technique as described, for example in Lue et al., Endocrinology, Vol. 141, No. 4, pp. 1414-1424 (2000), the contents of which are incorporated herein by reference. Briefly, testicular parenchyma are weighed and then homogenized in the same volume (equivalent to testicular parenchyma weight) of 0.01 M PBS (pH 7.4). An aliquot, after appropriate dilution is counted in a hemocytometer.
  • Each square of the hemocytometer with covers lip in place represents a total volume of 10 "4 cm 3 .
  • the number of spermatids per ml or per gram testis is obtained, then multiplied by the testis volume (equivalent to testicular weight) to yield the number of spermatids per testis.
  • a successful antiprogestin/SPRM reduces the number of spermatids per testis relative to testosterone alone.
  • the contralateral testes are then fixed by vascular perfusion with 5% gluteraldehyde in 0.05 M cacodylate buffer (pH 7.4) for 30 minutes preceded by a brief saline wash.
  • the testes are removed, cut into small (about 0.2 cm) transverse slices and placed into the same fixative overnight and processed for routine paraffin embedding for in situ detection of apoptosis.
  • In situ detection of cells with DNA strand breaks is performed by the terminal deoxynuleotidyl transferase (TdT)- mediated deoxy-UTP nick end labeling (TUNEL) technique.
  • TdT terminal deoxynuleotidyl transferase
  • TUNEL deoxy-UTP nick end labeling
  • tissue sections are incubated with proteinase K for 15 minutes at room temperature, washed in distilled water, and then treated with 2% hydrogen peroxide in PBS for 5 minutes at room temperature to quench endogenous peroxidase activity. Sections are then incubated with a mixture containing digoxigenin-conjugated nucleotides and TdT in a humidified chamber at 37 degrees C for 1 hour and subsequently treated with antidigoxigenin-peroxidase for 30 minutes at room temperature. To detect immunoreactive cells, the sections are incubated with a mixture of 0.05% diaminobenzidine and 0.01% hydrogen peroxide for 6 minutes.
  • Sections are counterstained with 0.5% methyl green, dehydrated in 100% butanol, cleared in xylene and mounted with Permount (Fisher Scientific, Fairlawn, NJ).
  • the apoptotic index (apoptotic germ cells per 100 Sertoli cells) at various seminiferous epithelial stages (preferably stages VII-VIII) is then calculated using a microscope with a XlOO oil immersion objective.
  • An antiprogestin/SPRM candidate that is pro- apoptotic in the testis increases the apoptotic index relative to testosterone alone.
  • Urinary estrogen levels were measured for each animal of Example 6 for one month during the middle of the study (weeks 14-17) and for the last month of the study (weeks 33-36).
  • the follicular phase results are based on 35 baseline ovulating cycles. The results are presented below:
  • Example 8 CDB-4124 and Lupron® but not RU 486 Suppress Proliferation in Cynomolgus Monkey Endometrial Epithelia.
  • Apoptosis was assessed in tissue from the same animals on slides by the terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling (TUNEL) technique.
  • TUNEL terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling
  • CDB-4124 Suppresses Proliferation in Human Endometrial Epithelia in a Dose-dependent Manner
  • CDB-4124 a GnRH agonist, commonly used for the treatment of endometriosis (also known as Lupron®).
  • CDB-4124 and RU 486 are Apoptotic in Rat Breast Tissue
  • Sprague-Dawley female rats were given 10 mg/kg body weight of DMBA at 50 days of age.
  • One group of 14 rats (Group 2) received sesame oil at 50 days of age instead of DMBA to serve as the no-DMBA controls.
  • Group 1 received daily subcutaneous (s.c.) injections of vehicle (10% ethanol in sesame oil).
  • Group 2 no DMBA control group - no tumors expected received vehicle on a schedule decided beforehand to simulate initiation of treatment over a three-month time period.
  • Groups 3 and 4 received daily s.c. injections of RU 486 or micronized progesterone at 10 mg/kg body weight, respectively.
  • Groups 5 through 9 received 20 mg/kg, 10 mg/kg, 2 mg/kg, 1 mg/kg and 0.1 mg/kg of CDB-4124, respectively.
  • Groups 10 through 14 mirrored the treatment given 5 through 9 except that 10 mg/kg of micronized progesterone was also added as a component to compositions for injections.
  • the ability of CDB-4124 to decrease proliferation appears to be important for the tumor suppressor activity of CDB-4124 because one of the major differences between CDB-4124 and RU 486 is that CDB-4124 reduced proliferation much more efficiently than RU 486.
  • An interruption or suppression of a strong proliferative effect of progesterone is a plausible mechanism by which CDB-4124 may reduce proliferation.

Abstract

The subject matter of the instant invention is pertinent to the field of male contraception. More specifically, the subject matter of the instant invention concerns methods of inhibiting spermatogenesis in males. The instant invention is also relevant to methods of male contraception. Compositions for practicing the methods, comprising progesterone antagonists are also disclosed. Embodiments of the instant invention also disclose methods for identifying new selective progesterone receptor modulators for practicing disclosed methods of treatment.

Description

COMPOSITIONS AND METHODS FOR MALE CONTRACEPTION
CROSS-REFERENCE TO RELATED APPLICATIONS
[00001] This application claims the benefit of U.S. Provisional Application No. 61/048,444, filed April 28, 2008, the contents of which are incorporated herein by reference.
FIELD OF THE INVENTION
[00002] The present invention relates to compositions and methods for male contraception. More specifically, the present invention relates to compositions comprising one or more progesterone antagonists for inhibiting spermatogenesis.
BACKGROUND OF THE INVENTION
[00003] Unplanned pregnancy is one of the most common medical problems faced by sexually active women despite the wide variety of contraceptive methods and devices available. Current contraceptive methods and devices are overwhelmingly targeted at the female and include birth control pills, intrauterine devices, spermicidal gels or creams and intravaginal barriers such as sponges or diaphragms. In contrast, only a few male birth control options (e.g. condoms or vasectomy) are available. [00004] Each of the male contraceptives currently available has certain drawbacks. For example, vasectomy is a surgical procedure which, while nearly 100% effective, requires additional surgery to reverse, and even then is only sometimes reversible. Condoms are less effective than birth control pills and are subject to leakage due to tearing. Additionally, use of condoms requires action by the male immediately prior to intercourse and results in a reported loss of sensation. Thus, noncompliance dilutes the effectiveness of this contraceptive. [00005] Several studies have shown suppression of spermatogenesis in human males by treatment with testosterone. Clinical trials have demonstrated that administration of exogenous testosterone alone can provide nearly universal azoospermia (lack of any measurable level of sperm in the semen) in Asian populations, although success has been more modest in Caucasian populations. Although achievement of azoospermia is ideal in order to confer optimal contraceptive protection, various degrees of sperm suppression have been shown to provide acceptable contraceptive protection. Normal sperm counts in human males are generally from about 20 million to about 120 million sperms per milliliter of semen.
[00006] Transient exposure of the scrota to heat has been shown to enhance testostereone-mediated suppression of spermatogenesis, due to accelerated apoptosis. A recent study demonstrates that treatment with the progestin levonorgestrel enhances testosterone-induced spermatogenesis suppression in human males allowing the use of lower concentrations of testosterone to achieve the same effect.
SUMMARY OF THE INVENTION
[00007] The instant invention provides methods for inhibiting spermatogenesis comprising administering to a male mammal, an androgen receptor agonist and a composition comprising one or more progesterone antagonists in a collective amount effective to inhibit spermatogenesis. Optionally, the methods further comprise the administration of a progestin and/or a gonadotropin-releasing hormone (GnRH) agonist or antagonist. The male mammal may be a human male. The progesterone antagonist may be a pure antiprogestin or a selective progesterone receptor modulator (SPRM). In a preferred embodiment, the progesterone antagonist has low affinity for glucocorticoid receptor. In another preferred embodiment, administration of the progesterone antagonist is for a period of time effective to prevent conception. [00008] In another aspect, the instant invention provides methods of contraception comprising administering to a male mammal, an androgen receptor agonist and a composition comprising one or more progesterone antagonists in a collective amount effective to achieve oligospermia or, preferably, azoospermia in the male mammal. Optionally, the methods further comprise the administration of a progestin and/or a gonadotropin-releasing hormone (GnRH) agonist or antagonist. The male mammal may be a human male. The progesterone antagonist may be a pure antiprogestin or a selective progesterone receptor modulator (SPRM). In a preferred embodiment, the progesterone antagonist has low affinity for glucocorticoid receptor. In another preferred embodiment, administration of the progesterone antagonist is for a period of time effective to inhibit spermatogensis in the male mammal.
DETAILED DESCRIPTION OF THE INVENTION
[00009] The term "effective dosage" means an amount of the composition's active component sufficient to achieve the desired effect which may be, e.g., inhibition of spermatogenesis.
[00010] The term "selective progesterone receptor modulators" means compounds that affect functions of progesterone receptor in a tissue-specific manner. The compounds act as progesterone receptor antagonists in some tissues (for example, in the uterus) and as progesterone receptor agonists in other tissues.
[00011] The terms "treat" or "treatment" refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down
(lessen) an undesired physiological change or disorder. For purposes of the present invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented. [00012] The term "progesterone agonist" means a compound that binds to a progesterone receptor and mimics the action of the natural hormone. [00013] The term "progesterone antagonist" means a compound that binds to a progesterone receptor and inhibits the effect of progesterone. [00014] The present invention relates to the use of progesterone antagonists at doses effective to inhibit spermatogenesis in a male mammal when administered simultaneously, separately or sequentially with an androgen receptor agonist. It has been surprisingly discovered that administration of certain antiprogestins induces apoptosis in several mammalian tissues including endometrial tissue and breast tissue and exhibits a dose-dependent inhibition of the LH surge that is the signal for ovulation. The pro-apoptotic and apparent LH lowering characteristics of compositions of the invention make them surprisingly useful for inhibiting spermatogenesis.
[00015] Administration of testosterone is known to suppress the secretion of luteinizing hormone (LH) and follicle stimulating hormone (FSH) in the pituitary and decrease intratesticular testosterone. This in turn results in the reversible inhibition of spermatogenesis to azoospermia or severe oligospermia in men. This is consistent with earlier studies demonstrating that mice lacking the LH receptor are infertile and that human patients with LH-β subunit mutations have severely impaired spermatogenesis.
[00016] Reversible inhibition of spermatogenesis with exogenous testosterone has been shown to be greatly enhanced with addition of progestins such as levongestrel or gonadotropin-releasing hormone (GnRH) agonists/antagonists, allowing the use of lower concentrations of testosterone to achieve azoospermia or severe oligospermia. [00017] In adult mammals, germ cell death is observed during spermatogenesis and is a pivotal factor in sperm output. Several studies have implicated increased apoptosis of germ cells as the mechanism by which exogenous testosterone inhibits spermatogenesis. Moreover, a recent study conducted on human males has demonstrated that transient testicular warming enhances and hastens testosterone- mediated inhibition of spermatogenesis and that the enhanced effect is due to an increase in the number of apoptotic germ cells.
[00018] In view of the ability of progesterone agonists such as levonorgestrel to enhance the testosterone-mediated inhibition of spermatogenesis, the use of progesterone antagonists in inhibiting spermatogenesis is unexpected. [00019] In one embodiment, a male mammal with a need or desire to inhibit spermatogenesis is administered a composition comprising a progesterone antagonist in an amount effective to inhibit spermatogenesis. The male mammal may be a human male.
[00020] In a related embodiment, the present invention provides a method of inhibiting spermatogenesis comprising administering to a male mammal an androgen receptor agonist and a composition comprising at least one progesterone antagonist, wherein the androgen receptor agonist and progesterone antagonist are administered in a collective amount effective to inhibit spermatogenesis. The progesterone antagonist may be administered simultaneously, separately or sequentially with the administration of the androgen receptor agonist. Administration of exogenous androgens is known to inhibit spermatogenesis. Androgen receptor agonists may be natural or synthetic and include, without limitation: androgen hormones such as testosterone, dihydrotestosterone and 5α-androstanediol; testosterone undecanoate, testosterone enanthate, testosterone esters, testosterone proprionate, mesterolone, danazol and gestrinone. A preferred androgen receptor agonist is testosterone undecanoate. Selective androgen receptor agonists may also be used for their agonist activity. The dosage of the androgen receptor agonist may be any dosage known in the art to inhibit spermatogenesis. For example, it has been shown that weekly intramuscular (IM) injections of 200 mg of testosterone enanthate for six months is sufficient to induce azoospermia in about 65-74% of males and is sufficient to induce oligospermia in nearly all males. Monthly IM injections of 500 mg of testosterone undecanoate, following a 1000 mg loading dose, has been shown to be sufficient to induce azoospermia in nearly all males. Parental testosterone preparations and testosterone implants have been demonstrated to provide equivalent or greater efficacy to IM testosterone enanthate. For example, implants of testosterone pellets containing up to 800 mg (5.2 mg/day release rate) achieved azoospermia in about 40% of men during a 12-month period. In specific embodiments it is contemplated that administration of a composition comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist relative to that known in the art to inhibit spermatogenesis. For purposes of this application, a "suboptimal" dose of androgen receptor agonist is any such reduced dosage of androgen receptor agonist lower than would result in optimal inhibition of spermatogenesis. Reducing the dose of androgen receptor agonist is expected to reduce androgenic side effects and should therefore result in greater patient acceptability and compliance. Thus, in one embodiment, the present invention provides a method for inhibiting spermatogenesis comprising the administration of a composition comprising one or more progesterone antagonists to a male in an amount effective to inhibit spermatogenesis when combined with a suboptimal dose of androgen receptor agonist, preferably testosterone undecanoate. The composition may be administered simultaneously, separately or sequentially with the androgen receptor agonist. In other embodiments, the present invention provides methods of enhancing the inhibition of spermatogenesis in a male mammal, comprising administering to the mammal a composition comprising at least one progesterone antagonist and an androgen receptor agonist.
[00021] In another embodiment, the instant invention provides methods of contraception comprising administering to a male mammal an androgen receptor agonist and a composition comprising at least one progesterone antagonist, wherein the androgen receptor agonist and progesterone antagonist are administered in a collective amount effective to achieve oligospermia or, preferably, azoospermia in the male mammal. The progesterone antagonist may be administered simultaneously, separately or sequentially with the administration of the androgen receptor agonist. [00022] Normal sperm counts in human males are generally from about 20 million to about 120 million sperm per milliliter of semen. Where the intended effect is contraception, administration of compositions of the invention is of sufficient dosage and for a sufficient administration period to achieve oligospermia or preferably, azoospermia. "Oligospermia" is defined herein as a concentration of sperm less than 20 million per milliliter of semen. "Azoospermia" is defined herein as a lack of measurable level of sperm in the semen. Thus, when contraception is the desired effect, administration of an androgen receptor agonist and a composition comprising at least one progesterone antagonist to male mammal decreases the concentration of sperm below about 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, and preferably below about 1 million sperm per milliliter of semen in the male mammal. When the concentration of sperm is decreased below 1 million per milliliter of semen, contraceptive protection is provided to a Pearl index rating of 1.4 per 100 couple- years.
[00023] Long-term administration of the compositions may be preferable where the desired effect is contraception. Methods of the invention may comprise administering a composition comprising an effective amount of a progesterone antagonist for an administration period of least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or more days. The composition may also be administered for an administration period of least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more months. The composition may also be administered for an administration period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years. During the administration period, the composition may be administered daily or periodically such as every other day, every other month, and the like. The composition may also be administered intermittently. For example, the composition may be administered for an administration period of 1, 2, 3, 4, 5 or more months, followed by a period of discontinuance, followed by an administration period of 1, 2, 3, 4, 5 or more months, and so on.
[00024] By "intermittent administration" it is meant a period of administration of a therapeutically effective dose of progesterone antagonist, followed by a time period of discontinuance, which is then followed by another administration period and so forth. [00025] By "period of discontinuance" or "discontinuance period" it is meant a discontinuing of the daily, weekly, monthly or therebetween administration of progesterone antagonist. The time period of discontinuance may be longer or shorter than the administration period but is always longer than the dosing interval during the administration period. For example, where the administration period comprises daily, weekly, or monthly dosing, the discontinuance period is at least 2 days, at least 8 days or at least 32 days, respectively. Thus, the discontinuance period may be at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32 or more days.
[00026] In certain aspects of the invention, combination therapy comprising the administration of a composition comprising at least one progesterone anatagonist and one or more androgen receptor agonists further comprises the administration of one or more anti-gonadotrophic agents.
[00027] In one embodiment, the antigonadotrophic agents are progestins. Administration of exogenous progestin is known to reduce the amount of exogenous androgen receptor agonist necessary to inhibit spermatogenesis. Progestins are defined herein as natural or synthetic progestational substances that mimic some or all of the actions of progesterone. Nonlimiting examples of progestins include derivatives of 19-nortestosterone, such as oestranes and gonanes, derivatives of 17α- acetoxyprogesterone (pregnanes), and 17α-hydroxyprogesterone derivatives such as medroxyprogesterone acetate (MPA). Examples of oestranes include norethindrone and its acetates, and ethynodiol diacetate. Examples of gonanes include norgestrel, levonorgestrel and derivatives of levonorgestrel such as desogestrel, norgestimate and gestodene. A preferred progestin is levonorgestrel. The dosages of progestin and androgen receptor agonist may be any dosage, the combination of which is known in the art to inhibit spermatogenesis. For example, the addition of a single IM injection of medroxyprogesterone (300 mg) hastened and enhanced the inhibition of spermatogenesis achieved with a regimen of weekly IM injections of 200 mg testosterone enanthate. In another study, the addition of 150 and 300 mg medroxyprogesterone to IM injections of 1000 mg testosterone undecanoate every eight weeks increased the incidence of azoospermia and severe oligospermia during the 24-week treatment period from 80% to 100%. Another study comparing 100 mg IM weekly testosterone enanthate with and without daily levonorgestrel (125 meg, 250 meg or 500 meg orally) for six months reported a higher percentage of men achieving oligospermia in the androgen/progestin groups (89%, 89%, 78%) than in androgen only groups (56%). In specific embodiments it is contemplated that administration of a composition comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist and/or progestin relative to that known in the art to inhibit spermatogenesis, to achieve oligospermia or preferably to achieve azoospermia.
[00028] In another embodiment, the antigonadotrophic agents are gonadotropin- releasing hormone (GnRH) agonists or antagonists. Administration of exogenous GnRH agonists/antagonists is known to reduce the amount of exogenous androgen receptor agonist necessary to inhibit spermatogenesis. Non-limiting examples of GnRH agonists include nafarelin, buserelin, leuprolide, triptorelin, goserelin, [DLys6]GnRH, [DAIa6] GnRH and the like. Non-limiting examples of GnRH antagonists include histrelin, abarelix and those found in U.S. Patent Nos. 4,409,208, 4,547,370, 4,565,804, 4,569,927 and 4,619,914, incorporated herein in by reference in their entirety. A preferred GnRH agonist is leuprolide. The dosages of GnRH agonist/antagonist and androgen receptor agonist may be any dosage, the combination of which is known in the art to inhibit spermatogenesis. For example, a GnRH antagonist coadministered every 2 weeks with IM testosterone enanthate resulted in azoospermia in 88% of men by week 10. In specific embodiments it is contemplated that administration of a composition comprising a progesterone antagonist will allow a reduction in the amount of androgen receptor agonist and/or GnRH agonist/antagonist relative to that known in the art to inhibit spermatogenesis, to achieve oligospermia or preferably to achieve azoospermia. [00029] To achieve the appropriate therapeutic outcome in the combination therapies contemplated herein, i.e., to achieve inhibition of spermatogenesis in a male mammal, to achieve oligospermia or preferably to achieve azoospermia, one would administer to the male mammal a composition comprising a progesterone antagonist and at least one androgen receptor agonist and optionally at least one progestin and/or GnRH agonist/antagonist in a combined amount effective to produce the desired therapeutic outcome.
[00030] In a preferred embodiment of each method of the invention, the progesterone antagonist exhibits reduced affinity for the glucocorticoid receptor. More preferably, the binding affinity of the progesterone antagonist for the progesterone receptor is at least 1.5 times greater than the binding affinity of the progesterone antagonist for the glucocorticoid receptor.
[00031] Any known progesterone antagonist with characteristics of the compounds described above can be used by an artisan practicing the instant invention. Particularly useful compounds include those disclosed in U.S. Patent No. 6,900,193, hereby incorporated by reference in its entirety, as well as those disclosed in U.S. Patent No. 6,861,415, hereby incorporated by reference in its entirety, that are 21- substituted 19-norpregnanes with a general formula:
Figure imgf000013_0001
wherein:
X may be, for example alkyl, alkenyl, alkynyl, hydrogen, halo, monoalkylamino or dialkylamino, such as N,N-dimethylamino;
Ri may be, for example O, NOH or NO-methyl;
R2 may be, for example hydrogen or acetyl; and
R3 may be, for example methyloxy, formyloxy, acetoxy, acyloxy, S-alkoxy, acetyltheonyl, glycimate, vinyl ether, acethyloxymethyl, methyl carbonate, halogens, methyl, hydroxy, and ethyloxy.
The examples of 21 -substituted 19-norpregnanes include, but are not limited to, the following 24 compounds disclosed below.
1. CDB-4247 (21-propio[[l]]nyloxy-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000014_0001
2. CDB-4361 (21 -vinyl ether- 17α-acetoxy- 1 lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000014_0002
3. CDB-4059 (21-acetoxy-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000015_0001
4. CDB-4124 (21-methoxy-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000016_0001
5. CDB-4031 (21 -bromine- 17α-acetoxy- 11 β-(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000016_0002
6. CDB-3876 (21 -chlorine- 17α-acetoxy- 1 lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000017_0001
7. CDB-4058 (21-flourine-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000017_0002
8. CDB-4030 (21 -methyl- 17α-acetoxy- 1 lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000018_0001
9. CDB-4152 (21 -hydroxy- 17α-acetoxy- 1 lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000018_0002
10. CDB-4167 (21-ethyloxy-17a-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000019_0001
11. CDB-4101 (21 -methoxythio- 17α-acetoxy- 11 β-(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000019_0002
12. CDB-4110 (21-acetonide-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000020_0001
13. CDB-4111 (21 -BMD- 17α-acetoxy- 11 β-(4 N, N-dimethylaminophenyl)- 19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000020_0002
14. CDB-4125 (21-(Cyp*-hydroxy)-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000021_0001
*Cyp = 3-Cyclopentylpropionyloxy-
15. CDB-4205 (3 -hydroxy amino-21 -methoxy- 17α-acetoxy- 11 β-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000021_0002
16. CDB-4206 (3-hydroxyamino-21-acetoxy-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000022_0001
17. CDB-4226 (3-hydroxyamino-21-ethyloxy-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000022_0002
18. CDB-4262 (S-methoxyamino^l-ethyloxy-lVa-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000023_0001
19. CDB-4223 (21-methylthio-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000023_0002
20. CDB-4119 (4-benzoin-21 -acetylthio- 17α-acetoxy- 11 β-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000024_0001
21. CDB-4239 (4-benzoin-21-methoxy-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000025_0001
22. CDB-4306 (21-glycinate-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19- norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000025_0002
23. CDB-4352 (ll-cyanothio-Ha-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)-19 norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000026_0001
24. CDB-4362 (21-methoxyacetyl-17α-acetoxy-l lβ-(4 N, N-dimethylaminophenyl)- 19-norpregna-4,9-diene-3,20-dione) with the following structural formula:
Figure imgf000026_0002
[00032] 11 β-monodemethylated derivatives of the 24 compounds disclosed above (i.e., those in which X is N-methylamino) are also particularly useful in practicing the instant invention. In this regard, CDB-4453 (21-methoxy-17α-acetoxy-l lβ-(4-N- methylaminophenyl)- 19-norpregna-4,9-diene-3 ,20-dione), a monodemethylated derivative of CDB-4124, has been demonstrated to possess even lower anti- glucocorticoid activity than its parent. Attardi et al, 2002, MoI. Cell. Endocrin. 188:111-123, the contents of which are incorporated herein by reference. [00033] Although compounds of the general formula above and their monodemethylated derivatives are preferred, any progesterone antagonist may be used in the practice of the present invention. Preferably, the progesterone antagonist has one or more of the following characteristics: low antiglucocorticoid activity, minimal estrogenic and anti-estrogenic activities, and does not substantially elevate progesterone levels.
[00034] Antiprogestins which may be useful in the invention include, without limitation, asoprisnil (benzaldehyde, 4-[(l lβ,17β)-17-methoxy-17-(methoxymethyl)- 3-oxoestra-4,9-dien-l l-yl]-l-(E)-oxim; J867), its metabolite J912 (4-[17β-Hydroxy- 17α-(methoxymethyl)-3-oxoestra-4,9-dien-l lβ-yl]benzaldehyd-(lE)-oxim), and other compounds described in DE 43 32 283 and DE 43 32 284; CDB-2914 (17α- acetoxy- 11 β-(4-N,N-dimethylaminophenyl)- 19-norpregna-4,9-dien-3 ,20-dione) and other compounds described in Stratton et al, 2000, Hu. Reprod. 15:1092-1099; JNJ- 1250132 and other compounds described in Allan et al, 2006, Steroids 71 :949-954; 5-Aryl-l,2-dihydrochromeno[3,4-f]quinolines described in Zhi et al, 1998, J. Med. Chem. 41 :291-302; l,4-dihydro-benzo[d][l,3]oxazin-2-ones described in U.S. Patent Nos.: 6,509,334, 6,566,358 and 6,713,478 to Zhang et al; l,3-dihydro-indol-2-ones described in U.S. Patent No. 6,391,907 to Fensome et al.; 2,3-dihydro-lH-indoles described in U.S. Patent No. 6,417,214 to Ulrich et al.; benzimidazolones and analogues thereof described in U.S. Patent No. 6,380,235 to Zhang et al.; 2,1- benzisothiazoline 2,2-dioxides described in U.S. Patent No. 6,339,098 to Collins et al.; cyclocarbamates and cyclo-amides described in U.S. Patent Nos.: 6,306,851 and 6,441,019 to Santilli et al.; cyclic urea and cyclic amide derivatives described in U.S. Patent No. 6,369,056 to Zhang et al.; and quinazolinone and benzoxazine derivatives described in U.S. Patent No. 6,358,948 to Zhang et al.
[00035] Other antiprogestins that may be useful in the invention include, without limitation, (6α,l lβ,17β)-l l-(4-dimethylaminophenyl)-6-methyl-4',5'- dihydrospiro[estra-4,9-diene-17,2'(3'H)-furan]-3-one (ORG-31710) and other compounds described in U.S. Patent No. 4,871,724; (1 lβ,17α)-l l-(4-acetylphenyl)- 17,23-epoxy-19,24-dinorchola-4,9,20-trien-3-one (ORG-33628); (7β,l lβ,17β)-l l-(4- dimethylaminophenyl-7-methyl]-4',5'-dihydrospiro[estra-4,9-diene-17,2'(3'H)- furan]-3-one (ORG-31806) and other compounds described in U.S. Patent No. 4,921,845; ZK-112993 and other compounds described in Michna et al., 1992, J. Steroid Biochem. Molec. Biol. 41 :339-348; ORG-31376; ORG-33245; ORG-31167; ORG-31343; RU-2992; RU-1479; RU-25056; RU-49295; RU-46556; RU-26819; LGl 127; LG120753; LG120830; LG1447; LG121046; CGP-19984A; RTI-3021-012; RTI-3021-022; RTI-3021-020; RWJ-25333; ZK-136796; ZK-114043; ZK-230211; ZK-136798; ZK-98229; ZK-98734; and ZK-137316. [00036] Still other antiprogestins that may be useful in the invention include, without limitation, mifepristone (l lβ-[p-(Dimethylamino)phenyl]-17β-hydroxy-17- (l-propynyl)estra-4,9-dien-3-one; RU 486) and other compounds described in U.S. Patent Nos.: 4,386,085, 4,447,424, 4,519,946 and 4,634,695; the phosphorus- containing 17β-side chain mifepristone analogues described in Jiang et ah, 2006, Steroids 71 :949-954; onapristone (l lβ-[p-(dimethylamino)phenyl]-17α-hydroxy-17- (3-hydroxypropyl)-13α-estra-4,9-dien-3-one) and other compounds described in U.S. Patent No. 4,780,461; lilopristone (((Z)-I lβ-[(4-dimethylamino)phenyl]-17-β- hydroxy-17α-(3-hydroxy-l-propenyl)estra-4,9-dien-3-one) and other compounds described in U.S. Patent No. 4,609,651; the 11 β-substituted 19-norsteroids, such as 1 lβ-(4-Methoxyphenyl)-17β-hydroxy-17α-ethynyl-4,9-estradien-3-one described in Belagner et al., 1981, Steroids 37:361-382; the 11 β-aryl-4-estrenes such as (Z)-I lβ- [(4-Dimethylamino)phenyl)] - 17β-hy droxy- 17α-(3 -hydroxy- 1 -propeny l)estr-4-en-3 - one described in U.S. Patent No. 5,728,689; the l lβ-aryl-estrene derivatives described in U.S. Patent Nos.: 5,843,933 and 5,843,931; the 11-benzaldoxime-estra- diene derivatives such as 4-[17β-Methoxy-17α-(methoxymethyl)-3-oxoestra-4,9- dien-1 lβ-yl]benzaldehyde-l-(E)-oxime described in U.S. Patent No. 5,693,628; the l l-benzaldoxime-17β-methoxy-17α-methoxymethyl-estradiene derivatives such as 4-[l 7β-Methoxy- 17α-(methoxymethyl)-3-oxoestra-4,9-dien- 11 β-yl]benzaldehyde- 1 - (E)-[O-(ethylamino)carbonyl]oxime described in U.S. Patent No. 5,576,310; the S- substituted 1 lβ-benzadoxime-estra-4,9-diene-carbonic acid thiolesters such as 4- [ 17β-Methoxy- 17α-(methoxymethyl)-3-oxoestra-4,9-dien- 11 β-yl]benzaldehyde- 1 - (E)-[O-(ethylthio)carbonyl]oxime, described in WO 99/45023; the steroid esters such as (Z)-6 ' -(4-cy anophenyl)-9, 11 α-dihydro- 17β-hydroxy- 17α- [4-( 1 -oxo-3 - methylbutoxy)-l-butenyl]4'H-naphtho[3',2',l ';10,9,l l]estr-4-en-3-one described in DE 19652408, DE 4434488, DE 4216003, DE 4216004 and WO 98/24803; the fluorinated 17α-alkyl chain steroids such as l lβ-(4-acetylphenyl)-17β-hydroxy-17α- (l,l,2,2,2-pentafluoroethyl)estra-4,9-dien-3-one described in WO 98/34947; the 17- spirofuran-3'-ylidene steroids such as l lbeta-(4-Acetylphenyl)-19,24-dinor- 17,23- epoxy-17alpha-chola-4,9,20-trien-3-one described in U.S. Patent No. 5,292,878; (Z)- 1 lbeta, 19-[4-(3-Pyridinyl)-o-phenylene]- 17beta-hydroxy-l 7α-[3-hydroxy- 1 - propenyl]-4-androsten-3-one and other compounds described in U.S. Patent No. 5,439,913; the 13-alkyl-l l-beta-phenyl gonanes such as l lbeta-[4-(l- methylethenyl)phenyl]- 17α-hydroxy- 17beta-(3-hydroxypropyl)- 13 α-estra-4,9-dien- 3-one described in U.S. Patent No. 5,446,036; the 11-arylsteroids such as 4',5'- Dihydro-l lbeta-[4-(dimethylamino)phenyl]-6beta-methylspiro[estra-4,9-dien- 17beta,2'(3'H)-furan]-3-one described in U.S. Patent No. 4,921,845; the 11-beta-aryl- estradienes described in U.S. Patent Nos.: 4,829,060, 4,814,327 and 5,089,488; the 1 l-beta-aryl-4,9 gonadiens and 1 l-beta-aryl-13-alkyl-4,9-gonadiens described in U.S. Patent Nos.: 5,739,125, 5,407,928 and 5,273,971; the l l-beta-aryl-6-alkyl (or alkenyl or alkinyl) steroids described in EP 289073; the 10-beta,l 1 -beta-bridged steroids described in U.S. Patent No. 5,093,507; the 1 l-beta-aryl-14-beta- steroids described in U.S. Patent No. 5,244,886; the 19,11 -beta-bridged steroids described in U.S. Patent Nos: 5,095,129, 5,446,178, 5,478,956 and 5,232,915; the 1-arylsulphonyl, arylcarbonyl and l-arylphosphonyl-3-phenyl-l,4,5,6-tetrahydropyridazines described in U.S. Patnet No. 5,684,151; the 1-arylsulphonyl, arylcarbonyl and arylthiocarbonyl pyridazino derivatives described in U.S. Patent No. 5,753,655; the l,2-dihydro-[l,2- g]quinoline derivatives and l,2-dihydro-chromeno-[3,4-f]quinoline derivatives described in U.S. Patent Nos: 5,688,808,. 5,693,646, 5,693,647, 5,696,127, 5,696,130 and 5,696,133; the oxa-steroids 6 derived from (8S, 13S, 14R)-7-oxa-estra-4,9-diene- 3,17-dione 1 described in Kang et al, 2007, Bioorg. Med. Chem. Lett. 15:907-910; and the 7-oxa-steroids 4 described in Kang et al., 2007, Bioorg. Med. Chem. Lett. 17:2531-2534.
[00037] In the preferred embodiment, the progesterone antagonist is the antiprogestin/SPRM CDB-4124 (21-methoxy-17α-acetoxy-l lβ-(4 N, N- dimethylaminophenyl)- 19-norpregna-4,9-diene-3 ,20-dione). [00038] Where contraception is the intended effect, compositions of the instant invention are preferably suitable for prolonged administration. In this regard, the progesterone antagonist preferably has only low glucocorticoid receptor binding activity and therefore, does not substantially interfere with functions of glucocorticoid receptor. Thus, administration of the compositions may have reduced side effects, such as mood swings, fatigue and weight loss, typically found when antiprogestins with a high affinity for glucocorticoid receptor are used.
[00039] In another embodiment the instant invention teaches methods that can be used for identifying compounds that possess selective progesterone receptor binding activity. These methods include receptor binding and in vivo bioassays such as anti- McGinty, anti-Clauberg, glucocorticoid, estrogenic, androgenic, anti-glucocorticoid (AG), anti-estrogen, and anti-androgen activities as well as post-coital and anti- ovulatory activities where in the leading compounds of the instant invention are used as a reference.
[00040] In another embodiment, the instant invention teaches that the potential SPRMs can be also analyzed for their effect on transcriptional activity in human cells. When SPRMs disclosed in the instant invention are used as a reference, this analysis can furnish information about (1) SPRM 's interaction with receptor, (2) interaction of the activated receptor with other transcription factors, (3) activation of a transcriptional complex at a progesterone response element (PRE); and ultimately its effect on gene expression. In these experiments, plasmid expressing the hPR-B can be cotransfected with any reporter known to a person skilled in the relevant art under the PRE-dependent promoter into HeLa, HepG2 or T47D cells. The reporters may include, but are not limited to, luciferase, beta-galactosidase, green fluorescent protein, red fluorescent protein or yellow fluorescent protein. After transfection, the cells are treated with either a test compound or one of the disclosed in this application SPRMs that serves as a positive control. Following treatment, cells are assayed for reporter expression.
[00041] In another embodiment, the instant invention teaches that prospective SPRMs can be tested for their ability to oppose dexamethasone-induced cell death in human lymphocytic cell line CEM-7 and compared to effects of SPRMs disclosed in the instant specification. In these experiments, dexamethasone can be added at a concentration that results in cell death. The cells are then treated with either RU486, one of SPRMs of the instant invention or a test compound at concentrations between 10~6 and l0~8 M. [00042] Progesterone antagonist compounds that may be used in accordance with the present invention can be synthesized using synthetic chemistry techniques known in the art such as those disclosed in U.S. Patent No. 6,861,415. It is to be understood that certain functional groups may interfere with other reactants or reagents under the reaction conditions and therefore may need temporary protection. The use of protecting groups is described in 'Protective Groups in Organic Synthesis', 2nd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1991). [00043] In one embodiment, compositions of the invention comprise one or more progesterone antagonists or pharmaceutically acceptable salts thereof. Depending on the process conditions the salt compound obtained may be either in neutral or salt form. Salt forms include hydrates and other solvates and also crystalline polymorphs. Both the free base and the salts of these end products may be used in accordance with the invention.
[00044] Acid addition salts may in a manner known per se be transformed into the free base using basic agents such as alkali or by ion exchange. The free base obtained may also form salts with organic or inorganic acids.
[00045] In the preparation of acid addition salts, preferably such acids are used which form suitably pharmaceutically acceptable salts. Examples of such acids are hydrochloric acid, sulfuric acid, phosphoric acid, nitric acid, aliphatic acid, alicyclic carboxylic or sulfonic acids, such as formic acid, acetic acid, propionic acid, succinic acid, glycolic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, glucuronic acid, fumaric acid, maleic acid, hydroxymaleic acid, pyruvic acid, aspartic acid, glutamic acid, p-hydroxybenzoic acid, embonic acid, ethanesulfonic acid, hydroxyethanesulfonic acid, phenylacetic acid, mandelic acid, alogenbensenesulfonic acid, toluenesulfonic acid, galactaric acid, galacturonic acid or naphthalenesulfonic acid. All crystalline form polymorphs may be used in accordance with the invention. [00046] Base addition salts may also be used in accordance with the invention and may be prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner. Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkali earth metals or organic amines. Examples of metals used as cations are sodium, potassium, calcium, magnesium and the like. Examples of suitable amines are amino acids such as lysine, choline, diethanolamine, ethylenediamine, N-methylglucamine and the like [00047] For the aforementioned purposes, the compounds of the instant invention can be administered to a patient via any conventional route where the progesterone antagonist is active. For instance, a progesterone antagonist of the instant invention can be administered orally, parenterally, sublingually, transdermally, rectally, transmucosally, topically, via inhalation, via buccal administration, or combinations thereof. Parenteral administration includes, but is not limited to, intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, intrathecal, intraarticular, intracisternal and intraventricular. The administration form can be a tablet, capsule, pill, nasal mist, aerosol, pellet, implant (or other depot) and the like. [00048] A therapeutically effective amount of the composition required for use in therapy may vary depending on the particular compound employed, the mode of administration, the severity of the condition being treated, the length of time that activity is desired, among other factors, and is ultimately determined by the attendant physician. However, in general, doses employed for human treatment typically are in the range of about 0.001 mg/kg to about 500 mg/kg per day, for example about 1 μg/kg to about 1 mg/kg per day or about 1 μg/kg to about 100 μg/kg per day. For most large mammals, the total daily dosage is from about 1 to 100 mg, preferably from about 2 to 80 mg. The dosage regimen may be adjusted to provide the optimal therapeutic response. The desired dose may be conveniently administered in a single dose, or as multiple doses administered at appropriate intervals, for example as two, three, four or more subdoses per day.
[00049] Illustratively, a composition of the invention may be administered to a subject to provide the subject with a progesterone antagonist in an amount of about 1 μg/kg to about 1 mg/kg body weight, for example about 1 μg/kg, about 25 μg/kg, about 50 μg/kg, about 75 μg/kg, about 100 μg/kg, about 125 μg/kg, about 150 μg/kg, about 175 μg/kg, about 200 μg/kg, about 225 μg/kg, about 250 μg/kg, about 275 μg/kg, about 300 μg/kg, about 325 μg/kg, about 350 μg/kg, about 375 μg/kg, about 400 μg/kg, about 425 μg/kg, about 450 μg/kg, about 475 μg/kg, about 500 μg/kg, about 525 μg/kg, about 550 μg/kg, about 575 μg/kg, about 600 μg/kg, about 625 μg/kg, about 650 μg/kg, about 675 μg/kg, about 700 μg/kg, about 725 μg/kg, about 750 μg/kg, about 775 μg/kg, about 800 μg/kg, about 825 μg/kg, about 850 μg/kg, about 875 μg/kg, about 900 μg/kg, about 925 μg/kg, about 950 μg/kg, about 975 μg/kg or about 1 mg/kg body weight. [00050] The compositions of the instant invention may contain from about 25 to about 90% of the active ingredient in combination with the carrier, more usually between about 5% and 60% by weight.
[00051] Solid carriers may include starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while liquid carriers may include sterile water, polyethylene glycols, non-ionic surfactants and edible oils such as corn, peanut and sesame oils, as are appropriate to the nature of the active ingredient and the particular form of administration desired. Flavoring agents, coloring agents, preserving agents, and antioxidants, for example, vitamin E and ascorbic acid, may be included in preparations as well. Under ordinary conditions of storage and use, the preparations may contain a preservative to prevent the growth of microorganisms. [00052] The compositions of the instant invention can be formulated into tablets in a tablet press by using techniques well-known to an artisan skilled in the relevant field. Optionally, the active ingredients according to the invention can also be pressed separately into two-layer tablets. According to the instant invention, tablets may include androgen receptor agonists, GnRH agonists, GnRH antagonists, progestins or any combination thereof as one of the active ingredients. Compositions of the instant invention can also be formulated as an oily solution. [00053] Patients undergoing treatments with the compositions of the instant invention should be monitored routinely for their glucocorticoid levels. [00054] The following non-limiting examples are provided to aid in understanding the teachings of the instant invention. [00055] All patents, patent applications and publications referenced herein are hereby incorporated by reference herein to the fullest extent allowed under the law.
Example 1. Formulations of The Instant Invention Can Be Prepared As Tablets.
[00056] To obtain tablets for practicing the instant invention, the following ingredients can be pressed together in a tablet press:
50.0 mg of CDB-4124
140.5 mg of lactose
69.5 mg of corn starch
2.5 mg of poly-N-vinylpyrrolidone
2.0 mg of aerosil
0.5 mg of magnesium stearate
[00057] To obtain tablets comprising progestins for practicing the instant invention, the following ingredients can be pressed together in a tablet press:
250.0 μg of desogestrel
50.0 mg of CDB-4124
125.0 mg of lactose
40.0 mg of corn starch
2.5 mg of poly-N-vinylpyrrolidone 25
2.0 mg of aerosil
0.5 mg of magnesium stearate [00058] To obtain oily preparations for practicing the instant invention, for example the following ingredients can be mixed together and loaded into ampoules:
100.0 mg of CDB-4124 343.4 mg of castor oil 608.6 mg of benzyl benzoate
Example 2. Compounds of the Instant Invention May Have Only Weak Antiglucocorticoid Receptor Binding Activity.
[00059] Certain antiprogestins were tested in receptor-binding assays for their ability to bind rabbit progesterone receptor (rbPR) and glucocorticoid receptor (rbGR). Briefly, cytosol containing PR or GR were prepared in TEGMD buffer (10 mM Tris, pH 7.2, 1.5 mM EDTA, 0.2 mM sodium molybdate, 10% glycerol, 1 mM DTT) from uterus or thymus, respectively, of estradiol-primed immature rabbits. For PR binding, the cytosol was incubated with 6 nM l,2-[3H]progesterone (50.0 Ci/mmole) and competitors were added at concentrations from 2 to 100 nM. For binding to GR, the cytosol was incubated with 6 nM 6,7-[3H]-dexamethasone (40 Ci/mmol) and test compounds were added at concentrations from 20 to 100 nM. After overnight incubation at 4 C, bound and unbound [3H] steroids were separated by addition of dextran-coated charcoal and centrifugation at 2100 x g for 15 min at 4 C. Supernatants containing the [3H]-steroid receptor complexes were decanted into vials containing 4 ml Optifluor (Packard Instrument Co.), vortexed, equilibrated in a liquid scintillation counter for 30 minutes and then counted for 2 minutes. The EC50 (Effective Concentration) for each standard curve and each of the compound curves was determined by entering the counting data into a four parameter sigmoidal computer program (RiaSmart® Immunoassay Data Reduction Program, Packard Instrument Co., Meriden, Conn.). Relative binding affinity (RBA) for each compound was calculated using the following equation: EC50 of standard/ECso of test compound x 100. The standards for the PR and GR assays were unlabeled progesterone and dexamethasone, respectively. The results of these experiments are summarized in Table 1, as a ratio of the relative binding affinities of each compound for the rbPR and rbGR receptors (rbPR/rbGR). This differential reflects the relative activity of a compound in a cell or tissue that possesses the two receptors and the requisite transcriptional co factors.
[00060] Also given in Table 1 are the relative biological activities of the same compounds in the rabbit uterus by the anti-McGinty and anti-Clauberg assays. Compound CDB-2914 (listed at the end of the Table) was used as the control or reference compound (rabbit Biological Activity = 1.00) for these experiments because results of experiments using CDB-2914 have been published before (Hild-Petito et al, 1996; Passaro et al, 1997; Reel et al, 1998; Lamer et al, 2000). For the anti- McGinty test, immature female rabbits received a subcutaneous injection of 5 μg estradiol in 10% ethanol/sesame oil daily for 6 consecutive days. On day 7, animals underwent sterile abdominal surgery to ligate a 3-4 cm segment of both uterine horns. The test compound in appropriate solvent was injected intraluminally into the ligated segment of one uterine horn and vehicle alone into the other. A stimulating dose of progesterone (267 μg/day) was administered subcutaneously to each rabbit daily for the next three days to induce endometrial proliferation. All animals were sacrificed at day 10 for removal of the uterus where a segment central to the ligatures was removed and fixed in 10% neutral buffered formalin and submitted for histological processing. Five micron sections stained with hematoxylin and cosin were evaluated microscopically for the degree of endometrial glandular proliferation. The percent inhibition of endometrial proliferation for each rabbit was calculated and the mean of the group of five animals recorded. For the Anti-Clauberg test, immature female rabbits received a subcutaneous injection of 5 μg estradiol in 10% ethanol/sesame oil daily for 6 consecutive days. On day 7, animals received progesterone by subcutaneous injection (160 μg/day) and the experimental compound in appropriate vehicle orally or subcutaneously for five consecutive days. One group of rabbits received progesterone only. Twenty-four hours after the last dose, all animals were sacrificed for removal of the uterus which was cleaned of all fat and connective tissue, weighed to the nearest 0.2 mg and placed in 10% neutral buffered formalin for subsequent histological processing. Five micron sections stained with hematoxylin and eosin were evaluated microscopically for the degree of endometrial glandular proliferation. The percent inhibition of endometrial proliferation at each dose level of the test compound was derived by comparison with progesterone-stimulated animals alone. The data presented in Table 1 (rabbit Biol. Act.) reflects the average of the results obtained for each compound by the anti-McGinty and anti-Clauberg assays relative to CDB-2914. [00061] The tested antiprogestins were ranked on the basis of the selectivity of each compound for the rabbit PR over the rabbit GR, as listed in Table 1. The antiprogestins were also ranked on the basis of the biological activity in the rabbit uterus. Data presented in Table 1 show that the affinity of leading compounds for progesterone receptor was at least 1.5 times greater than their affinity for glucocorticoid receptor.
[00062] The results of these studies also show that the two leading compounds CDB-4124 and CDB-4059 have strong antiprogestin activity in the rabbit uterus in comparison to RU 486 and CDB-2914. Both compounds lack estrogenic, androgenic, anti- estrogenic, and anti-androgenic activities. Both compounds possess minimal anti-glucocorticoid receptor activity, a feature that distinguishes them from RU 486 and CDB-2914 which are moderately active in glucocorticoid receptor binding. In these assays, CDB-4124 performed slightly better than CDB-4059
TABLE 1 -RECEPTOR BINDING AND BIOLOGICAL ACTIVITIES OF SPRMS
SPRM rbPR/rbGR rabbit Biol. SPRM rbPR/rbGR rabbit Biol.
Act. Act.
4239 14.80 0.60 4416 1.33 0.77
4241 9.10 0.34 4417 1.31 0.70
4361 7.20 3.03 4111 1.30 0.36
4306 5.90 0.95 4125 1.19 1.55
4363 5.75 2.53 4223 1.17 not given
3875 5.11 1.40 4398 1.16 0.99
4362 4.74 1.25 4058 1.08 0.90
4352 4.21 0.57 4418 1.03 0.25
4176 3.83 0.20 4177 1.03 0.00
4243 2.90 0.00 4030 0.96 0.30
4119 2.60 0.10 4374 0.95 2.25
4324 2.16 1.10 4399 0.93 0.35
4247 2.06 1.70 4152 0.82 1.40
4205 1.99 1.00 4110 0.70 0.10
4059 1.89 2.90 4031 0.69 0.70 4400 1.76 2.29 4101 0.61 0.65
3247 1.74 0.10 4248 0.42 0.00
4167 1.69 1.50 4227 0.38 0.00
4124 1.58 3.60 4393 0.35 0.00
4226 1.51 0.54 4396 0.18 not given
4206 1.44 0.68 2914 1.07 1.00
Example 3. Measuring Cortisol.
[00063] Several different experimental systems support a conclusion that RU 486 increases Cortisol because RU 486 has strong anti-glucocorticoid properties in humans and primates.
[00064] However, rats treated with RU 486 at 10 mg/kg showed no significant difference in the levels of Cortisol. In contrast, rats treated with either CDB-4124 or
CDB-4059 at the same dose levels had significantly higher levels of serum Cortisol than rats from a control group.
[00065] These higher levels were in the range of 3-4 ug/dl (30-40 ng/ml). The effects were dose-dependent in that increasing doses of CDB-4124 led to increased
Cortisol.
[00066] This difference in effects of RU 486 versus CDB-4124 or CDB-4059 on
Cortisol levels can be explained by assuming that after 21 days of chronic dosing, a rat liver was able to metabolize RU 486 better than either of the two CDB compounds.
Example 4. Measuring Corticosterone.
[00067] Corticosterone is the most abundant glucocorticoid in rats. The effects of the SPRMs on Cortisol may be secondary to strong effects on corticosterone. To better explore this phenomenon, the levels of corticosterone were measured in groups, which showed the strongest changes in Cortisol levels, such as groups treated with CDB-4124 at 20 mg/kg or 10mg/kg. For comparison, the following groups were also assayed: a group that received 20 mg/kg CDB-4124 plus 10 mg/kg progesterone, a group that received 10 mg/kg CDB-4124 plus 10 mg/kg progesterone, a group that received 10 mg/kg RU 486, a group that received 10 mg/kg of progesterone alone, a control group. The levels of corticosterone were 10-40 times higher than the levels of Cortisol. However, almost no difference between groups with respect to mean corticosterone levels was observed. There were no differences among the groups before treatment (p = 0.43, Kruskal-Wallis test), after 21 days of treatment (p = 0.57, Kruskal-Wallis test), or after 28 days of treatment and at sacrifice (p = 0.061, Kruskal-Wallis test.
[00068] To measure effects of exogenous progesterone on serum corticosterone, the levels of corticosterone were compared in 3 paired groups that differed in whether they received exogenous progesterone (e.g., comparisons of control versus progesterone or CDB-4124 at 20 mg/kg versus CDB-4124 at 20 mg/kg plus progesterone, or CDB-4124 at 10 mg/kg versus CDB-4124 at 10 mg/kg plus progesterone). There was a statistically significant difference detected: the levels of corticosterone were lowered in animals treated with progesterone after 21 days of treatment (p = 0.029, Mann- Whitney Wilcoxon test, two-tailed). This effect was not verified in sera taken at sacrifice. No differences in serum corticosterone were found between the progesterone and the CDB-4124 groups, the progesterone and the RU- 486 groups, or the RU-486 group and the CDB-4124 groups. [00069] The relationship between serum Cortisol and serum corticosterone in each group was also examined. There was a strong positive linear correlation between the two for CDB-4124 at 20 mg/kg (r2 = 0.78), for CDB-4124 at 10 mg/kg (r2 = 0.82), and for RU 486 (r2 = 0.85). Adding progesterone to the first two CDB-4124 groups made the relationship far less strong (r2 = 0.34 for Group 10 and r2 = 0.37 for Group 11, respectively). Progesterone itself showed no such positive relationship (r2 = -1.0). The control group demonstrated no relationship between the two glucocorticoids (r2 = 0.064). Thus, increased levels of Cortisol in groups receiving CDB-4124 are correlated to levels of corticosterone, due perhaps to conversion from corticosterone that is somehow enhanced. This is consistent with an effect of CDB-4124 seen above: an effect on metabolic enzymes responsible for levels of progesterone and Cortisol.
[00070] Although no strong effect of CDB-4124 on the primary glucocorticoid of the rat was found, nevertheless, for safety reasons, patients given CDB-4124 or CDB- 4059 in Phase I clinical trials should be monitored for possible anti-glucocorticoid effects including a possible increase in serum Cortisol, corticosterone, or ACTH.
Example 5. Testing Antiprogestins/SPRMs for the Ability to Inhibit Spermatogenesis and for Pro-apoptotic Effects on Testicular Germ Cells In Vivo
[00071] Adult (e.g. 60-day-old) male Sprague Dawley rats are divided into two groups. Both groups are given subdermal testosterone implants with a release rate of about 30 μg/cm-day along the rats' dorsal surface while under anesthesia for a treatment period of six weeks. One group is additionally given a range of concentrations of a candidate antiprogestin/SPRM during the treatment period. At the end of six weeks, all animals are injected with heparin (130 IU/100 grams body weight) 15 minutes prior to administration of a lethal injection of sodium phentobarbital (100 mg/kg BW IP). One testis from each rat is then removed and weighed, and after decapsulation, testicular parenchyma are used for determining the number of advanced (steps 17-19) spermatids by the homogenization technique as described, for example in Lue et al., Endocrinology, Vol. 141, No. 4, pp. 1414-1424 (2000), the contents of which are incorporated herein by reference. Briefly, testicular parenchyma are weighed and then homogenized in the same volume (equivalent to testicular parenchyma weight) of 0.01 M PBS (pH 7.4). An aliquot, after appropriate dilution is counted in a hemocytometer. Each square of the hemocytometer with covers lip in place represents a total volume of 10"4 cm3. The number of spermatids per ml or per gram testis is obtained, then multiplied by the testis volume (equivalent to testicular weight) to yield the number of spermatids per testis. A successful antiprogestin/SPRM reduces the number of spermatids per testis relative to testosterone alone.
[00072] The contralateral testes are then fixed by vascular perfusion with 5% gluteraldehyde in 0.05 M cacodylate buffer (pH 7.4) for 30 minutes preceded by a brief saline wash. The testes are removed, cut into small (about 0.2 cm) transverse slices and placed into the same fixative overnight and processed for routine paraffin embedding for in situ detection of apoptosis. In situ detection of cells with DNA strand breaks is performed by the terminal deoxynuleotidyl transferase (TdT)- mediated deoxy-UTP nick end labeling (TUNEL) technique. Briefly, after deparaffϊnization and rehydration, tissue sections are incubated with proteinase K for 15 minutes at room temperature, washed in distilled water, and then treated with 2% hydrogen peroxide in PBS for 5 minutes at room temperature to quench endogenous peroxidase activity. Sections are then incubated with a mixture containing digoxigenin-conjugated nucleotides and TdT in a humidified chamber at 37 degrees C for 1 hour and subsequently treated with antidigoxigenin-peroxidase for 30 minutes at room temperature. To detect immunoreactive cells, the sections are incubated with a mixture of 0.05% diaminobenzidine and 0.01% hydrogen peroxide for 6 minutes. Sections are counterstained with 0.5% methyl green, dehydrated in 100% butanol, cleared in xylene and mounted with Permount (Fisher Scientific, Fairlawn, NJ). The apoptotic index (apoptotic germ cells per 100 Sertoli cells) at various seminiferous epithelial stages (preferably stages VII-VIII) is then calculated using a microscope with a XlOO oil immersion objective. An antiprogestin/SPRM candidate that is pro- apoptotic in the testis increases the apoptotic index relative to testosterone alone.
Example 6. CDB-4124 Lowers Luteal Phase Progesterone in Cynomolgus Monkeys
[00073] Cynomolgus monkeys {Macaca fascicularis) (n=14) were treated orally for 36 weeks with CDB-4124 or RU-486 at 1.0 mg/kg/day or with placebo (control). Another group (n=14) received Lupron® IM once per month. Urinary progesterone levels were measured for each animal for one month during the middle of the study (weeks 14-17) and for the last month of the study (weeks 33-36). The results are presented below:
Figure imgf000047_0001
Example 7. CDB-4124 Does Not Lower Follicular Phase Estrogen in Cynomolgus
Monkeys
[00074] Urinary estrogen levels were measured for each animal of Example 6 for one month during the middle of the study (weeks 14-17) and for the last month of the study (weeks 33-36). The follicular phase results are based on 35 baseline ovulating cycles. The results are presented below:
Figure imgf000047_0002
Example 8. CDB-4124 and Lupron® but not RU 486 Suppress Proliferation in Cynomolgus Monkey Endometrial Epithelia.
[00075] At week 36, three animals from each group of Example 6 were injected within 24 hours of sacrifice with the thymidine analog bromodeoxyuridine (BrdU), a marker of proliferating cells and their progeny, to assess tissue proliferation. Full thickness uterine sections were stained and examined microscopically for evidence of proliferation in terms of the % cells positive for incorporation of BrdU:
Figure imgf000048_0001
Example 9. CDB-4124 and RU 486 but not Lupron® Enhance Apoptosis in Cynomolgus Monkey Endometrial Epithelium
[00076] Apoptosis was assessed in tissue from the same animals on slides by the terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling (TUNEL) technique. The percent apoptotic cells is presented below:
Figure imgf000048_0002
Figure imgf000049_0001
Example 10. CDB-4124 Suppresses Proliferation in Human Endometrial Epithelia in a Dose-dependent Manner
[00077] Thirty-nine pre-menopausal adult women diagnosed with endometriosis were the subject of a six month study of Proellex™ (CDB-4124) in the treatment of endometriosis. The study included three dose levels of CDB-4124 as well as a positive control arm. The positive control was Lucrin®, a GnRH agonist, commonly used for the treatment of endometriosis (also known as Lupron®). CDB-4124 was administered in a double blinded fashion as a daily oral capsule at dosages of 12.5mg/day (n=2), 25mg/day (n=3) and 50mg/day (n=3). Another group (n=4) were injected with a slow release formulation of Lucrin® once per month as a positive control.
[00078] All doses of CDB-4124, as well as the Lucrin® dose, on average reduced distress related to pain over the course of the six month exposure to the drug, with the 50 mg CDB-4124 dose reducing both the duration and intensity of pain more effectively than the 12.5 mg or 25 mg doses and is significantly better (p = 0.0012) than Lucrin® in reducing the number of days of pain over the course of the study. Pain reduction also occurred more rapidly than with the active control, Lucrin®. The response of pain to treatment in this study was analyzed in two ways. Patients in the study maintained daily pain diaries to record the severity and frequency of pain. In addition, at each office visit, patients filled out endometriosis symptom surveys that included a questionnaire that evaluated intensity of pain on a bad day on a scale of 0- 10 with 10 being the greatest intensity. Daily pain diaries indicated that on average, women on Lucrin® experienced 19.4 days of pain over the first three months. Women on 50mg of CDB-4124 exhibited less than 1 day of pain over the same period. Women on 25mg and 12.5 mg of CDB-4124 exhibited more days of pain than that recorded by women receiving the highest dose of CDB-4124 or Lucrin®. There appeared to be a dose dependent effect on pain reduction. Over the 180 day treatment period, pain diaries indicated that women on the 50mg CDB-4124 dose had 170 or 96% pain free days (standard deviation = 8.86 days). This decrease in duration of pain was statistically better (p=0.0012) than the 117.8 (74%; standard deviation 51.4 days) pain free days achieved with Lucrin®. The 50 mg dose of CDB- 4124 was also statistically superior to both the 25mg and the 12.5mg doses with regard to pain free days. Patients on CDB-4124 12.5mg and 25mg doses had 115.9 (66%; standard deviation 69.2 days) and 133.6 (75%; standard deviation 27.4 days) pain free days, respectively. These results clearly support a dose response for CDB- 4124. The 25mg and 12.5mg doses of CDB-4124 were not statistically different from Lucrin®. At the end of the first month of therapy there was a statistically significant reduction in days of pain in the 50mg Proellex group (p=0.031) compared with baseline, but not in the three other treatment groups. The intensity of pain was assed by the question: "On a scale of 1-10, with 0 being no pain and 10 being extreme pain, how intense was your pain on a bad day?" The mean scores for intensity of pain at baseline were 6.3 for the CDB-4124 groups and 6.1 for the Lucrin® group. Statistically significant relief from pain was evident by the first month in the 25mg and 50mg Proellex groups. At month three all four active treatment groups had satistically significant reduction in pain compared with baseline, with the following scores: 3.7 (p = 0.03) for 12.5mg CDB-4124, 3.2 (p = 0.03) for 25 mg CDB-4124, 1.6 (p = 0.015) for 50mg CDB-4124 and 1.5 (p = 0.016) for Lucrin®. These dose related reductions continued until month six when the values for pain intensity were 2.0 (p = 0.008), 2.8 (p = 0.023), 0.6 (p = 0.004) and 0.7 (p = 0.016), respectively. Two months after stopping treatment pain returned and was of similar intensity in all four treatment groups.
[00079] Women receiving Lucrin® in the study, on average, experienced a reduction of estrogen to post-menopausal levels (<20 pg/ml) by month three and this was maintained through month six of treatment. This outcome was associated with a statistically significant increase (p = 0.023) in biomarkers of bone resorption compared with the baseline values at month three, and therefore an increased risk of bone loss. At month six as well as at the one-month follow up visit, this increase in markers of bone resporption was still present in women treated with Lucrin®. All doses of CDB-4124 maintained estrogen concentrations significantly above those seen with Lucrin® and remained in the low normal range (mean > 40 pg/ml). Importantly, there were no significant changes in biomarkers of bone resorption in any of the dose arms of CDB-4124 at three and six months of treatment. Women with post-menopausal levels of estrogen have been shown to be at greater risk for bone loss and other medical conditions. Lucrin®, therefore, is not indicated for treatment lasting longer than six months. [00080] Side effects of CDB-4124 were generally mild with no individual organ system being involved systematically. Although this was a small study and no definitive conclusions can be made from the safety data, there was no single signal of safety observed.
[00081] Women in the study were closely monitored for changes in the structure of the endometrium. Data from these examinations suggest an inverse dose dependent effect of CDB-4124 on endometrial thickness at the three month period. Comparisons were made to both baseline and visit one ultrasound measurements of endometrial thickness. After three months on treatment none of the women receiving the 50mg dose of CDB-4124 (n=3) exhibited thickened endometrium and actually exhibited a trend toward reduction in endometrial thickness compared to baseline. One woman receiving the 25mg dose of CDB-4124 (n=4) and two women receiving the 12.5mg dose of CDB-4124 (n=4) exhibited a thickened endometrium. The five women who received Lucrin® did not have a thickening of the endometrium due to a low estrogenic state. The results are presented below:
Figure imgf000052_0001
[00082] Unexpectedly, a dose-dependent inhibition of the LH surge, as measured by ovulation kits which indicate the presence of the LH surge about 30 hours prior to ovulation, was observed in women on CDB-4124, indicating that CDB-4124 may exert suppressive effects on pituitary hormones.
Example 9. CDB-4124 and RU 486 are Apoptotic in Rat Breast Tissue
[00083] To induce breast tumors, Sprague-Dawley female rats were given 10 mg/kg body weight of DMBA at 50 days of age. One group of 14 rats (Group 2) received sesame oil at 50 days of age instead of DMBA to serve as the no-DMBA controls. Animals were weighed and palpated weekly along the milk line for any sign of lesions or swellings. Tumor nodules were noted and measured weekly in two dimensions with calipers. When tumors grew to a size of 10-12 mm in any dimension, the individual animal was randomized into one of 14 groups. Tumors appeared as soon as 39 days after oral gavage and as late as 194 days (latter individual not included in study). The mean latency period for tumor appearance was 106 + 30 days. There were no differences between groups receiving DMBA in terms of latency (p = 0.545, Kruskal-Wallis test).
[00084] Animals were treated for 28 days on the following schedule. Group 1 received daily subcutaneous (s.c.) injections of vehicle (10% ethanol in sesame oil). Group 2 (no DMBA control group - no tumors expected) received vehicle on a schedule decided beforehand to simulate initiation of treatment over a three-month time period. Groups 3 and 4 received daily s.c. injections of RU 486 or micronized progesterone at 10 mg/kg body weight, respectively. Groups 5 through 9 received 20 mg/kg, 10 mg/kg, 2 mg/kg, 1 mg/kg and 0.1 mg/kg of CDB-4124, respectively. Groups 10 through 14 mirrored the treatment given 5 through 9 except that 10 mg/kg of micronized progesterone was also added as a component to compositions for injections.
[00085] Animals were sacrificed 3-5 days after the end of the 28-day treatment period, blood was drawn, and tumors were removed, weighed, measured, inspected, and portions frozen and/or placed in 10% phosphate buffered formalin for histopathology. The tissue samples were cut and stained with hematoxylin and eosin and were evaluated for histopathological classification. [00086] In order to assess the effects of progestins and anti-progestins on apoptosis, tissue sections derived from 46 individual rat tumors from treated and control animals were evaluated by an apoptosis hybridization kit (Oncor, Gaithersburg, MD). The cells in apoptosis were evaluated in the peripheral areas of the tumors and far from necrosis. At least 1,000 cells per tumor section were evaluated. A clear difference among the treatment groups relative to those of the control untreated animals as shown in Table 1 :
Table 1 : Apoptosis (% of cells in tumors in programmed cell death
Group Treatment % cells positive Compared to Controls t-test
1 Control tumors 0.81 + 0.31
3 RU 486 (10mg/kg) 3.34 + 2.57 p = 0.003
4 P4 (Progesterone) (10mg/kg) 1.28 + 0.51 p = 0.015
6 4124 (10 mg/kg) 3.84 + 3.10 p = 0.003
11 4124 + P4 3.78 + 4.93 P = 0.0496 ANOVA p = 0.003 [4124+P4 > Control, P4; RU486,
4124+P4>4124, Control] Post-analysis by the Multiple Range Test
[00087] A post-analysis by the Multiple Range Test indicated that CDB-4124 plus progesterone induced higher apoptosis than the control or progesterone -treated animals. Moreover, RU486, CDB-4124 and CDB4124 plus progesterone induced higher apoptotic cell death than observed in the control tumors. The effects of treatement with CDB-4124 were not different than RU486 (p=0.73, t-test). Similarly, the effects of CDB-4124 were the same as CDB-4124 + P4 (p=0.98, t-test). These results suggest that, in the presence of approximately equal amounts of progesterone, tumors respond to the anti-progestin CDB4124 with apoptosis. On the contrary, CDB-4124 leads to increased apoptosis compared to P4 (p=0.020, t-test). There is no apparent synergism between CDB-4124 and progesterone. The ability of CDB-4124 to decrease proliferation appears to be important for the tumor suppressor activity of CDB-4124 because one of the major differences between CDB-4124 and RU 486 is that CDB-4124 reduced proliferation much more efficiently than RU 486. An interruption or suppression of a strong proliferative effect of progesterone is a plausible mechanism by which CDB-4124 may reduce proliferation.

Claims

CLAIMSWE CLAIM:
1. A method for inhibiting spermatogenesis in a male comprising administering to a male an androgen receptor agonist and a composition comprising a compound of formula
(I):
Figure imgf000056_0001
(I)
or a pharmaceutically acceptable salt, hydrate or solvate thereof, wherein:
X represents an alkyl, alkenyl, alkynyl, hydrogen, halo, monoalkylamino or dialkylamino;
Ri represents O, NOH or NO-methyl;
R2 represents a hydrogen or acetyl; and
R3 represents methyloxy, formyloxy, acetoxy, acyloxy, S-alkoxy, acetyltheonyl, glycimate, vinyl ether, acethyloxymethyl, methyl carbonate, halogens, methyl, hydroxy, or ethyloxy,
whereby spermatogenesis is inhibited in said male.
2. The method of claim 1 , wherein the androgen receptor agonist is selected from the group consisting of testosterone, dihydrotestosterone, 5α-androstanediol, testosterone undecanoate, testosterone enanthate, testosterone proprionate, mesterolone, danazol and gestrinone.
3. The method of claim 2 wherein the androgen receptor agonist is testosterone undecanoate.
4. The method of claim 1 wherein said compound is CDB-4124.
5. The method of claim 3, wherein said composition and said androgen receptor agonist are administered sequentially.
6. The method of claim 3, wherein said composition and said androgen receptor agonist are administered simultaneously.
7. The method of claim 1 , wherein the dose of androgen receptor agonist is less than the dose required in the same patient in the absence of said composition in order to achieve the same result in terms of inhibiting spermatogenesis.
8. The method of claim 1 , further comprising the administration of at least one compound selected from the group consisting of a progestin, a gonadtropin releasing hormone (GnRH) agonist, and a GnRH antagonist.
9. The method of claim 1 , wherein the composition is administered for a period of time effective to prevent conception.
10. The method of claim 4, wherein said compound is administered at a dosage from 0.5mg/kg to 500mg/kg.
11. The method of claim 10, wherein said compound is administered at a dosage of 50mg per day.
12. The method of claim 1, wherein said androgen receptor agonist and said composition are administered for a period of at least from about one to about six months.
13. A method of contraception, comprising administering to a male an androgen receptor agonist and a composition comprising a compound of formula (I):
Figure imgf000058_0001
(I)
or a pharmaceutically acceptable salt, hydrate or solvate thereof, wherein:
X represents an alkyl, alkenyl, alkynyl, hydrogen, halo, monoalkylamino or dialkylamino;
Ri represents O, NOH or NO-methyl;
R2 represents a hydrogen or acetyl; and
R3 represents methyloxy, formyloxy, acetoxy, acyloxy, S-alkoxy, acetyltheonyl, glycimate, vinyl ether, acethyloxymethyl, methyl carbonate, halogens, methyl, hydroxy, or ethyloxy, whereby oligospermia or azoospermia is achieved in said male
14. The method of claim 13, wherein said compound is CDB-4124.
15. The method of claim 13, wherein the androgen receptor agonist is testosterone undecanoate.
16. The method of claim 13, wherein said composition and said androgen receptor agonist are administered simultaneously.
17. The method of claim 13, wherein said composition and said androgen receptor agonist are administered sequentially.
18. The method of claim 14, further comprising the administration of at least one compound selected from the group consisting of a progestin, a gonadtropin releasing hormone (GnRH) agonist, and a GnRH antagonist.
19. The method of claim 14, wherein said compound is administered at a dosage from 0.5mg/kg to 500mg/kg.
20. The method of claim 19, wherein said compound is administered at a dosage of 50mg/day.
PCT/US2009/041795 2008-04-28 2009-04-27 Compositions and methods for male contraception WO2009134713A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4844408P 2008-04-28 2008-04-28
US61/048,444 2008-04-28

Publications (1)

Publication Number Publication Date
WO2009134713A1 true WO2009134713A1 (en) 2009-11-05

Family

ID=40756947

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/041795 WO2009134713A1 (en) 2008-04-28 2009-04-27 Compositions and methods for male contraception

Country Status (4)

Country Link
AR (1) AR072781A1 (en)
CL (1) CL2009001009A1 (en)
TW (1) TW200950788A (en)
WO (1) WO2009134713A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016054197A1 (en) * 2014-09-30 2016-04-07 University Of Tennessee Research Foundation In situ gelling form for long-acting drug delivery

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007103510A2 (en) * 2006-03-08 2007-09-13 Danco Laboratories Llc Methods, dosing regimens & medications using anti-progestational agents for the treatment of disorders

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007103510A2 (en) * 2006-03-08 2007-09-13 Danco Laboratories Llc Methods, dosing regimens & medications using anti-progestational agents for the treatment of disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
FRANCIS Z ET AL: "Contraception of the future", REPRODUCTION HUMAINE ET HORMONES 200803 FR, vol. 21, no. 1, March 2008 (2008-03-01), pages 102 - 116, XP009118562, ISSN: 0994-3919 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016054197A1 (en) * 2014-09-30 2016-04-07 University Of Tennessee Research Foundation In situ gelling form for long-acting drug delivery

Also Published As

Publication number Publication date
CL2009001009A1 (en) 2009-06-19
TW200950788A (en) 2009-12-16
AR072781A1 (en) 2010-09-22

Similar Documents

Publication Publication Date Title
TWI477276B (en) Antiprogestin dosing regimens
JP6175364B2 (en) Compositions and methods for inhibiting endometrial proliferation
CZ292177B6 (en) Progesterone antagonists and gestagens for the treatment of endometriosis and leiomyomata uteri
SK284538B6 (en) Progesterone-antagonistic and anti-oestrogen active compounds for common use for female contraception
JP2011518883A (en) Compositions and methods for treating breast cancer
US20090118253A1 (en) Compositions and methods for treating dysfunctional uterine bleeding
WO2009134713A1 (en) Compositions and methods for male contraception
TW200822931A (en) Compositions and methods for suppressing endometrial proliferation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09739515

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09739515

Country of ref document: EP

Kind code of ref document: A1