US20080108691A1 - Open-chain prolyl urea-related modulators of androgen receptor function - Google Patents

Open-chain prolyl urea-related modulators of androgen receptor function Download PDF

Info

Publication number
US20080108691A1
US20080108691A1 US11/931,498 US93149807A US2008108691A1 US 20080108691 A1 US20080108691 A1 US 20080108691A1 US 93149807 A US93149807 A US 93149807A US 2008108691 A1 US2008108691 A1 US 2008108691A1
Authority
US
United States
Prior art keywords
substituted
alkyl
group
agents
arylalkyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/931,498
Inventor
Lawrence Hamann
David Augeri
Mark Manfredi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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 Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US11/931,498 priority Critical patent/US20080108691A1/en
Publication of US20080108691A1 publication Critical patent/US20080108691A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/26Androgens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to open chain prolyl urea and thiourea related compounds, methods of using such compounds in the treatment of androgen receptor-associated conditions, such as age-related diseases, for example sarcopenia, and to pharmaceutical compositions containing such compounds.
  • Nuclear hormone receptors constitute a large super-family of structurally-related and sequence-specific gene regulators scientists have named “ligand-dependent transcription factors.” R. M. Evans, Science, 240:889 (1988).
  • the steroid binding NHR's form a recognized subset of the NHR's, including the progesterone receptor (PR), androgen receptor (AR), estrogen receptor (ER), glucocorticoid receptor (GR) and mineralocorticoid receptor (MR).
  • PR progesterone receptor
  • AR androgen receptor
  • ER estrogen receptor
  • GR glucocorticoid receptor
  • MR mineralocorticoid receptor
  • the conventional nuclear hormone receptors are generally transactivators in the presence of ligand, which selectively bind to the NHR in a way that effects gene transcription.
  • the AR is a ligand-activated transcriptional regulatory protein that mediates induction of male sexual development and function through its activity with endogenous androgens.
  • androgens are associated with male and female maintenance of muscle mass and strength, bone mass and erythropoiesis.
  • Androgens such as testosterone
  • Androgens also play an important role in many physiological processes, such as differentiation of male internal and external genitalia, development and maintenance of male secondary sexual characteristics (e.g., the development of prostate, seminal vesicles, penis, scrotum, skeletal muscle, redistribution of body fat, stimulation of long bone growth, closure of epiphyses, development of male hair growth pattern and enlargement of larynx), the maintenance of sexual behavior and function (e.g., libido and potency) and spermatogenesis (in man).
  • male secondary sexual characteristics e.g., the development of prostate, seminal vesicles, penis, scrotum, skeletal muscle, redistribution of body fat, stimulation of long bone growth, closure of epiphyses, development of male hair growth pattern and enlargement of larynx
  • the maintenance of sexual behavior and function e.g., libido and potency
  • spermatogenesis in man.
  • the serum androgen concentration in the body declines.
  • the age dependent decline in androgens is associated with changes in body composition for men and women, such as a lower percentage of muscle mass and an increase in body fat, e.g., sarcopenia.
  • modulation of the AR gene can have an impact on the physiological effects associated with androgen production.
  • the effectiveness of known modulators of steroid receptors is often tempered by their undesired side-effect profile, particularly during long-term administration.
  • the administration of synthetic androgens has been associated with liver damage, prostate cancer, adverse effects on male sexual function and adverse effects associated with cardiovascular and erythropoietic function.
  • RU486 is an example of a synthetic antagonist of the PR, which is utilized as a birth control agent (Vegeto et al., Cell 69: 703-713 (1992)).
  • Flutamide is an example of an antagonist of the AR, which is utilized for the treatment of prostate cancer (Neri et al, Endo. 91, 427-437 (1972)).
  • Tamoxifen is an example of a tissue-selective modulator of the ER function, that is used in the treatment of breast cancer (Smigel J. Natl. Cancer Inst. 90, 647-648 (1998)). Tamoxifen can function as an antagonist of the ER in breast tissue while acting as an agonist of the ER in bone (Grese et al., Proc. Natl. Acad. Sci. USA 94, 14105-14110 (1997)). Because of the tissue-selective effects seen for Tamoxifen, this agent, and agents like it, are referred to as tissue-selective estrogen receptor modulators.
  • non-endogenous ligands for NHR's can be obtained from food sources (Regal et al., Proc. Soc. Exp. Biol. Med. 223, 372-378 (2000) and Hempstock et al., J. Med. Food 2, 267-269 (1999)).
  • the flavanoid phytoestrogens are an example of an unnatural ligand for SB-NHR's that are readily obtained from a food source such as soy (Quella et al., J. Clin. Oncol. 18, 1068-1074 (2000) and Banz et al., J. Med. Food 2, 271-273 (1999)).
  • soy Quella et al., J. Clin. Oncol. 18, 1068-1074 (2000) and Banz et al., J. Med. Food 2, 271-273 (1999)
  • non-natural ligands can be synthetically engineered to serve as modulators of the function of NHR's.
  • engineering of an unnatural ligand can include the identification of a core structure which mimics the natural steroid core system. This can be achieved by random screening against several SB-NHR's, or through directed approaches using the available crystal structures of a variety of NHR ligand binding domains (Bourguet et al., Nature 375, 377-382 (1995), Brzozowski, et al., Nature 389, 753-758 (1997), Shiau et al., Cell 95, 927-937 (1998) and Tanenbaum et al., Proc. Natl. Acad. Sci.
  • Differential substitution about such a steroid mimic core can provide agents with selectivity for one receptor versus another. In addition, such modifications can be employed to obtain agents with agonist or antagonist activity for a particular SB-NHR. Differential substitution about the steroid mimic core can result in the formation of a series of high affinity agonists and antagonists with specificity for, for example, ER versus PR versus AR versus GR versus MR. Such an approach of differential substitution has been reported, for example, for quinoline based modulators of steroid NHR in J. Med. Chem., 41, 623 (1998); WO 9749709; U.S. Pat. No. 5,696,133; U.S. Pat. No.
  • identification of compounds which have good specificity for one or more steroid receptors, but which have reduced or no cross-reactivity for other steroid or intracellular receptors would be of significant value in the treatment of male and female hormone-responsive diseases.
  • selective modulators of the steroid binding nuclear hormone receptors particularly non-steroidal, non-toxic tissue selective androgen receptor modulators, which activate the androgen receptor in skeletal muscle while demonstrating limited or neutral effect on other androgen responsive (e.g., prostate) tissues.
  • compounds which are capable of modulating the function of a nuclear hormone receptor.
  • the compounds are selective androgen receptor modulators, and have the general formula I wherein
  • R 1 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl and CH 2 OR 4 ;
  • R 2 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo, heteroaryl or substituted heteroaryl and CH 2 OR 4 ;
  • R 3 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, CH 2 OR 4 , OR 2 , SR 2 , halo, NHR 2 , NHCOR 4 , NHCO 2 R 4 , NHCONR 4 R 4 ′ and NHSO 2 R 4 ;
  • R 4 and R 4 ′ for each occurrence are each independently selected from the group consisting of hydrogen(H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo and heteroaryl or substituted heteroaryl;
  • G is a mono- or polycyclic ring system selected from the group consisting of aryl, heterocyclo and heteroaryl, wherein said ring system may optionally substituted with one or more substituents selected from the group consisting of hydrogen, halo, CN, CF 3 , OR 4 , CO 2 R 4 , NR 4 R 4 ′, CONR 4 R 4 ′, CH 2 OR 4 , SR 4 , SOR 4 , SO 2 R 4 , NO 2 , alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
  • X is a linking group selected from the group consisting of NR 4 and CHR 4 ;
  • Y is selected from the group consisting of oxygen (O), NR 4 , NOR 4 and sulfur (S);
  • Z is oxygen (—O—) or NR 4 ;
  • n is an integer of 1 or 2.
  • the compounds of formula I modulate the function of the nuclear hormone receptors, particularly the androgen receptor, and include compounds which are, for example, selective agonists, partial agonists, antagonists or partial antagonists of the androgen receptor.
  • the compounds of formula I possess activity as agonists of the androgen receptor and may be used in the treatment of diseases or disorders associated with androgen receptor activity, such as maintenance of muscle strength and function (e.g., in the elderly); reversal or prevention of frailty or age-related functional decline (“ARFD”) in the elderly (e.g., sarcopenia); prevention of catabolic side effects of glucocorticoids; prevention and treatment of reduced bone density or growth (e.g., osteoporosis and osteopenia); treatment of chronic fatigue syndrome (CFS); chronic myalgia; treatment of acute fatigue syndrome and muscle loss following elective surgery (e.g., post-surgical rehabilitation); acceleration of wound healing; accelerating bone fracture repair (such as accelerating the recovery of hip fracture patients); treatment of wasting secondary
  • the present invention provides for compounds of formula I, pharmaceutical compositions employing such compounds and for methods of using such compounds.
  • the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I, alone or in combination with a pharmaceutically acceptable carrier.
  • a method for preventing, inhibiting or treating the progression or onset of diseases or disorders associated with nuclear hormone receptors, particularly, the androgen receptor, such as the diseases or disorders defined above and hereinafter, wherein a therapeutically effective amount of a compound of formula I is administered to a mammalian, i.e., human, patient in need of treatment.
  • the compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more other agent(s) active in the therapeutic areas described herein.
  • a method for preventing, inhibiting or treating the diseases as defined above and hereinafter, wherein a therapeutically effective amount of a combination of a compound of formula I and another type of therapeutic agent, is administered to a human patient in need of treatment.
  • R 1 is hydrogen (H) or alkyl
  • R 2 is hydrogen (H) or alkyl
  • R 3 is hydroxyl (OH);
  • X is NR 4 ;
  • Y is O
  • Z is O
  • n 1
  • R 8 , R 9 , R 10 and R 11 in each functional group are each independently selected from the group consisting of hydrogen (H), NO 2 , CN, CF 3 , OR 4 , CO 2 R 4 , NR 4 ′, CONR 4 R 4 ′, CH 2 OR 4 , alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
  • a to F is each independently selected from N or CR 1 ;
  • J, K, L, P and Q are each independently selected from NR 12 , O, S, SO, SO 2 or CR 12 R 12 ′;
  • R 12 and R 12 ′ in each functional group are each independently selected from a bond or R 1 ;
  • n is an integer of 0 or 1.
  • Chiralpak® Trademark of Chiral Technologies, Inc. Eaton, Pa.
  • YMC® trademark of YMC Co, Ltd., Kyoto, Japan
  • nM nanomolar
  • alkyl denotes branched or unbranched hydrocarbon chains, preferably having about 1 to about 8 carbons, such as, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, 2-methylpentyl pentyl, hexyl, isohexyl, heptyl, 4,4-dimethyl pentyl, octyl, 2,2,4-trimethylpentyl and the like.
  • Substituted alkyl includes an alkyl group optionally substituted with one or more functional groups which are commonly attached to such chains, such as, hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, amido, and the like to form alkyl groups such as trifluoro methyl, 3-hydroxyhexyl, 2-carboxypropyl, 2-fluoroethyl, carboxymethyl, cyanobutyl and the like.
  • one or more functional groups which are commonly attached to such chains, such as, hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbal
  • cycloalkyl as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl,
  • “Substituted cycloalkyl” includes a cycloalkyl group optionally substituted with 1 or more substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol and/or alkylthio and/or any of the substituents included in the definition of “substituted alkyl.”
  • substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol and/or alkylthio and/or any of
  • alkenyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbons in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12-tetradecatrienyl, and the like.
  • Substituted alkenyl includes an alkenyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • alkynyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons in the normal chain, which include one or more triple bonds in the normal chain, such as 2-propynyl, 3butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl,3-undecynyl, 4-dodecynyl and the like.
  • Substituted alkynyl includes an alkynyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • arylalkyl refers to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • Substituted arylalkyl includes arylalkyl groups wherein the aryl portion is optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • halogen or “halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
  • aryl or “Ar” as employed herein alone or as part of another group refers to monocyclic and polycyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2-naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings), for example
  • “Substituted aryl” includes an aryl group optionally substituted with one or more functional groups, such as halo, haloalkyl, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl-alkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino wherein
  • heteroaryl refers to a 5- or 7-membered aromatic ring which includes 1, 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocycloalkyl ring (e.g. benzothiophenyl, indolyl), and includes possible N-oxides.
  • “Substituted heteroaryl” includes a heteroaryl group optionally substituted with 1 to 4 substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • substituents such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • heteroaryl groups include the following: and the like.
  • heterocyclo represents an unsubstituted or substituted stable 5- to 7-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinyl,
  • the compounds of formula I can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of formula I have, for example, at least one basic center, they can form acid addition salts.
  • acetic acid such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (C 1 -C 4 ) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen,
  • Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center.
  • the compounds of formula I having at least one acid group can also form salts with bases.
  • Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as morpholine, thiomorpholine, piperidine, pyrrolidine, a mono, di or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine.
  • Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula
  • Preferred salts of the compounds of formula I which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate or nitrate.
  • Preferred salts of the compounds of formula I which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines.
  • modulator refers to a chemical compound with capacity to either enhance (e.g., “agonist” activity) or inhibit (e.g., “antagonist” activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • prodrug esters as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of formula I with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like.
  • any compound that can be converted in vivo to provide the bioactive agent i.e., the compound of formula I
  • prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in:
  • An administration of a therapeutic agent of the invention includes administration of a therapeutically effective amount of the agent of the invention.
  • therapeutically effective amount refers to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance.
  • All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form.
  • the compounds of the present invention can have asymmetric centers at any of the carbon atoms including any one of the R substituents. Consequently, compounds of formula I can exist in enantiomeric or diastereomeric forms or in mixtures thereof.
  • the processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When diastereomeric or enantiomeric products are prepared, they can be separated by conventional methods for example, chromatographic, chiral HPLC or fractional crystallization.
  • the compounds of formula I of the invention can be prepared as shown in the following reaction schemes and description thereof, as well as relevant published literature procedures that may be used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working Examples.
  • selected compounds of formula I can be prepared from suitably protected intermediates of formula III.
  • Intermediates of formula II an be obtained commercially, can be prepared by methods known in the literature or can be readily prepared by one skilled in the art.
  • Intermediates of formula III can be obtained commercially, can be prepared by methods known in the literature or can be readily prepared by one skilled in the art.
  • Treatment of an intermediate of formula II with suitable coupling reagents, such as EDAC and HOBT together with a suitable coupling partner such as a compound of the formula HZR 1 yields an intermediate of the formula III.
  • Treatment of III with an intermediate of formula G-NCY yields compounds of the formula I.
  • the intermediates of formula G-NCY can be obtained, for example, from commercially available isocyanates, thioisocyanates and carbodiimides or can be readily prepared by one skilled in the art.
  • a compound of formula Ia can subsequently be obtained by any one of various methods for amide alkylation, such as for example, treatment of a compound of formula I with a base such as NaH, followed by treatment with an electrophile such as R 4 —Br to give the alkylated product Ia.
  • Scheme II describes a method for preparing compounds of formula I wherein a suitably protected intermediate of formula III is treated with a phosgene like reagent of formula C1-E-Cl in the presence of a base, such as NaHCO 3 or triethylamine, to yield an intermediate of formula IV.
  • a base such as NaHCO 3 or triethylamine
  • the phosgene like intermediates of formula C1-E-Cl can be obtained from commercially available sources, can be prepared by methods known in the literature or can be readily be prepared by one skilled in the art.
  • Phosgene equivalents such as carbonyldiimidazoles, may alternatively be substituted for Cl-E-Cl.
  • the intermediate of formula IV can be reacted with an amine of formula H 2 N-G in the presence of a base, such as triethylamine, to give a compound of formula I.
  • a base such as triethylamine
  • the amine intermediates H 2 N-G can be obtained from commercially available sources, can be prepared by methods known in the literature or can be readily be prepared by one skilled in the art.
  • Scheme III describes a method to prepare isocyanates and isothiocyanates of general formula Y ⁇ C ⁇ N-G, wherein intermediates H 2 N-G are treated with phosgene, thiophosgene, or a (thio)phosgene-like reagent in the presence of an inorganic base such as sodium bicarbonate, or a organic base such as diisopropylethylamine in a solvent such as dichloromethane to afford an isocyanate or isothiocyanate of formula Y ⁇ C ⁇ N-G.
  • an inorganic base such as sodium bicarbonate
  • a organic base such as diisopropylethylamine
  • solvent such as dichloromethane
  • Scheme IV describes a method of preparing certain intermediate compounds of the general formula IX (G) for incorporation into compounds of the general formula I by the methods described in the preceeding schemes.
  • a suitable protecting group such as tert-butyloxycarbonyl (BOC) using tert-butyloxycarbonyl (BOC) chloride or tert-butyloxycarbonyl anhydride and a suitable base such as triethyl
  • a halogen group at the 4-position of the aryl ring of a compound of the formula VII can optionally be converted into, for example, a nitrile group to give a compound of the formula VIII by treatment with a suitable reagent, such as CuCN.
  • Removal of the protective group can be accomplished, for example in the case of a tert-butyloxycarbonyl (BOC) group, treatment of a compound of the formula VIII under suitable cleaving conditions such as HCl in EtOH or TFA in CH 2 Cl 2 to give a compound of the formula IX (G).
  • BOC tert-butyloxycarbonyl
  • Scheme V describes a method of preparing certain compounds of formula I. Beginning with a 4-cyano 1-alkylaromatic compound of formula X, such as 4-cyano-1-methylnaphthalene (compound of formula X, where R 4 ⁇ H and R 10 and R 11 are taken together to form the remainder of a naphthyl ring), treatment with a strong base such as lithium diisopropyl amide, followed by quenching with a suitable electrophile, such as methyl chloroformate, affords a compound of the formula XI.
  • a compound of the formula XI can be hydrolysed to the free acid by standard methods known to those skilled in the art, such as by treatment with LiOH in MeOH to provide a compound of the formula XII.
  • a compound of the formula XII can then be used as an intermediate to prepare certain compounds of the formula I (where X ⁇ CH 2 ) by the employment of, for example, standard peptide coupling conditions, such as EDAC and HOBT with triethylamine in a reaction with a suitable pyrrolidine or piperidinyl based coupling partner such as a compound of the formula III.
  • standard peptide coupling conditions such as EDAC and HOBT with triethylamine in a reaction with a suitable pyrrolidine or piperidinyl based coupling partner such as a compound of the formula III.
  • the compounds of the present invention modulate the function of the nuclear hormone receptors, particularly the androgen receptor, and include compounds which are, for example, selective agonists, partial agonists, antagonists or partial antagonists of the androgen receptor (AR).
  • AR androgen receptor
  • the present compounds are useful in the treatment of AR-associated conditions
  • An “AR-associated condition,” as used herein, denotes a condition or disorder which can be treated by modulating the function or activity of an AR in a subject, wherein treatment comprises prevention, partial alleviation or cure of the condition or disorder. Modulation may occur locally, for example, within certain tissues of the subject, or more extensively throughout a subject being treated for such a condition or disorder.
  • the compounds of the present invention can be administered to animals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to maintenance of muscle strength and function (e.g., in the elderly); reversal or prevention of frailty or age-related functional decline (“ARFD”) in the elderly (e.g., sarcopenia); treatment of catabolic side effects of glucocorticoids; prevention and/or treatment of reduced bone mass, density or growth (e.g., osteoporosis and osteopenia); treatment of chronic fatigue syndrome (CFS); chronic myalgia; treatment of acute fatigue syndrome and muscle loss following elective surgery (e.g., post-surgical rehabilitation); accelerating of wound healing; accelerating bone fracture repair (such as accelerating the recovery of hip fracture patients); accelerating healing of complicated fractures, e.g.
  • AFD age-related functional decline
  • distraction osteogenesis in joint replacement; prevention of post-surgical adhesion formation; acceleration of tooth repair or growth; maintenance of sensory function (e.g., hearing, sight, olefaction and taste); treatment of periodontal disease; treatment of wasting secondary to fractures and wasting in connection with chronic obstructive pulmonary disease (COPD), chronic liver disease, AIDS, weightlessness, cancer cachexia, burn and trauma recovery, chronic catabolic state (e.g., coma), eating disorders (e.g., anorexia) and chemotherapy; treatment of cardiomyopathy; treatment of thrombocytopenia; treatment of growth retardation in connection with Crohn's disease; treatment of short bowel syndrome; treatment of irritable bowel syndrome; treatment of inflammatory bowel disease; treatment of Crohn's disease and ulcerative colits; treatment of complications associated with transplantation; treatment of physiological short stature including growth hormone deficient children and short stature associated with chronic illness; treatment of obesity and growth retardation associated with obesity; treatment of anorexia (e.g., associated with cachexia
  • the present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of formula I, alone or in combination with a pharmaceutical carrier or diluent.
  • a pharmaceutical carrier or diluent e.g., a pharmaceutically acceptable carrier or diluent.
  • compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antibiotic or other pharmaceutically active material.
  • the compounds of the present invention may be combined with growth promoting agents, such as, but not limited to, TRH, diethylstilbesterol, theophylline, enkephalins, E series prostaglandins, compounds disclosed in U.S. Pat. No. 3,239,345, e.g., zeranol, and compounds disclosed in U.S. Pat. No. 4,036,979, e.g., sulbenox or peptides disclosed in U.S. Pat. No. 4,411,890.
  • growth promoting agents such as, but not limited to, TRH, diethylstilbesterol, theophylline, enkephalins, E series prostaglandins, compounds disclosed in U.S. Pat. No. 3,239,345, e.g., zeranol, and compounds disclosed in U.S. Pat. No. 4,036,979, e.g., sulbenox or peptides disclosed in U.S. Pat. No
  • the compounds of the invention may also be used in combination with growth hormone secretagogues such as GHRP-6, GHRP-1 (as described in U.S. Pat. No. 4,411,890 and publications WO 89/07110 and WO 89/07111), GHRP-2 (as described in WO 93/04081), NN703 (Novo Nordisk), LY444711 (Lilly), MK-677 (Merck), CP424391 (Pfizer) and B-HT920, or with growth hormone releasing factor and its analogs or growth hormone and its analogs or somatomedins including IGF-1 and IGF-2, or with alpha-adrenergic agonists, such as clonidine or serotinin 5-HT D agonists, such as sumatriptan, or agents which inhibit somatostatin or its release, such as physostigmine and pyridostigmine.
  • growth hormone secretagogues such as GHRP-6, GHRP-1 (as described in U
  • a still further use of the compounds of the invention is in combination with estrogen, testosterone, a selective estrogen receptor modulator, such as tamoxifen or raloxifene, or other androgen receptor modulators, such as those disclosed in Edwards, J. P. et. al., Bio. Med. Chem. Let., 9, 1003-1008 (1999) and Hamann, L. G. et. al., J. Med. Chem., 42, 210-212 (1999).
  • a selective estrogen receptor modulator such as tamoxifen or raloxifene
  • other androgen receptor modulators such as those disclosed in Edwards, J. P. et. al., Bio. Med. Chem. Let., 9, 1003-1008 (1999) and Hamann, L. G. et. al., J. Med. Chem., 42, 210-212 (1999).
  • a further use of the compounds of this invention is in combination with progesterone receptor agonists (“PRA”), such as levonorgestrel, medroxyprogesterone acetate (MPA).
  • PRA progesterone receptor agonists
  • MPA medroxyprogesterone acetate
  • the compounds of the present invention may be employed alone or in combination with each other and/or other modulators of nuclear hormone receptors or other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-diabetic agents; anti-osteoporosis agents; anti-obesity agents; anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; anti-platelet agents; anti-thrombotic and thrombolytic agents; cardiac glycosides; cholesterol/lipid lowering agents; mineralocorticoid receptor antagonists; phosphodiesterase inhibitors; protein tyrosine kinase inhibitors; thyroid mimetics (including thyroid receptor agonists); anabolic agents; HIV or AIDS therapies; therapies useful in the treatment of Alzheimer's disease and other cognitive disorders; therapies useful in the treatment of sleeping disorders; anti-proliferative agents; and anti-tumor agents.
  • suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-diabetic agents; anti-o
  • Suitable anti-diabetic agents for use in combination with the compounds of the present invention include biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins (including insulin secretagogues or insulin sensitizers), meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide and glipizide), biguanide/glyburide combinations (e.g., Glucovance®), thiazolidinediones (e.g., troglitazone, rosiglitazone and pioglitazone), PPAR-alpha agonists, PPAR-gamma agonists, PPAR alpha/gamma dual agonists, SGLT2 inhibitors, glycogen phosphorylase inhibitors, inhibitors of fatty acid binding protein (aP2) such as those disclosed in U.
  • Suitable anti-osteoporosis agents for use in combination with the compounds of the present invention include alendronate, risedronate, PTH, PTH fragment, raloxifene, calcitonins, steroidal or non-steroidal progesterone receptor agonists, RANK ligand antagonists, calcium sensing receptor antagonists, TRAP inhibitors, selective estrogen receptor modulators (SERM's), estrogen and AP-1 inhibitors.
  • Suitable anti-obesity agents for use in combination with the compounds of the present invention include aP2 inhibitors, such as those disclosed in U.S. Ser. No. 09/519,079 filed Mar. 6, 2000, PPAR gamma antagonists, PPAR delta agonists, beta 3 adrenergic agonists, such as AJ9677 (Takeda/Dainippon), L750355 (Merck), or CP331648 (Pfizer) or other known beta ⁇ agonists as disclosed in U.S. Pat. Nos.
  • a lipase inhibitor such as orlistat or ATL-962 (Alizyme)
  • a serotonin (and dopamine) reuptake inhibitor such as sibutramine, topiramate (Johnson & Johnson) or axokine (Regeneron)
  • a thyroid receptor beta drug such as a thyroid receptor ligand as disclosed in WO 97/21993 (U.
  • anorectic agent such as dexamphetamine, phentermine, phenylpropanolamine or mazindol.
  • Suitable anti-inflammatory agents for use in combination with the compounds of the present invention include prednisone, dexamethasone, Enbrel®, cyclooxygenase inhibitors (i.e., COX-1 and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, Naproxen®, Celebrex®, Vioxx®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CellCept®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-1, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR1384), prostaglandin synthesis inhibitors, budesonide, clofazimine, CNI-1493, CD4 antagonists (e.g., prilix
  • Suitable anti-anxiety agents for use in combination with the compounds of the present invention include diazepam, lorazepam, buspirone, oxazepam, and hydroxyzine pamoate.
  • Suitable anti-depressants for use in combination with the compounds of the present invention include citalopram, fluoxetine, nefazodone, sertraline, and paroxetine.
  • Suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and T-type; e.g. diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenopril, fosino
  • Dual ET/AII antagonist e.g., compounds disclosed in WO 00/01389
  • neutral endopeptidase (NEP) inhibitors neutral endopeptidase (NEP) inhibitors
  • vasopepsidase inhibitors dual NEP-ACE inhibitors
  • omapatrilat and gemopatrilat e.g., omapatrilat and gemopatrilat
  • Suitable anti-platelet agents for use in combination with the compounds of the present invention include GPIIb/IIIa blockers (e.g., abciximab, eptifibatide, tirofiban), P2Y12 antagonists (e.g., clopidogrel, ticlopidine, CS-747), thromboxane receptor antagonists (e.g., ifetroban), aspirin, and PDE-III inhibitors (e.g., dipyridamole) with or without aspirin.
  • GPIIb/IIIa blockers e.g., abciximab, eptifibatide, tirofiban
  • P2Y12 antagonists e.g., clopidogrel, ticlopidine, CS-747
  • thromboxane receptor antagonists e.g., ifetroban
  • aspirin e.g., ifetroban
  • PDE-III inhibitors e.g., dipyridamole
  • Suitable cholesterol/lipid lowering agents for use in combination with the compounds of the present invention include HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin) and ZD-4522 (a.k.a.
  • HMG-CoA reductase inhibitors e.g., pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin) and ZD-4522 (a.k.a.
  • squalene synthetase inhibitors include rosuvastatin, or atavastatin or visastatin (squalene synthetase inhibitors), fibrates, bile acid sequestrants, ACAT inhibitors, MTP inhibitors, lipooxygenase inhibitors, cholesterol absorption inhibitors, and cholesterol ester transfer protein inhibitors (e.g., CP-529414).
  • mineralocorticoid receptor antagonists for use in combination with the compounds of the present invention include spironolactone and eplerinone.
  • PDE phosphodiesterase
  • PDE-3 inhibitors such as cilostazol
  • PDE-5 inhibitors such as sildenafil
  • thyroid mimetics examples include thyrotropin, polythyroid, KB-130015, and dronedarone.
  • Suitable therapies for treatment of Alzheimer's disease and cognitive disorders for use in combination with the compounds of the present invention include donepezil, tacrine, revastigmine, 5HT6, gamma secretase inhibitors, beta secretase inhibitors, SK channel blockers, Maxi-K blockers, and KCNQs blockers.
  • Suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, MLIB agonists, and GABA/NMDA receptor antagonists.
  • Suitable anti-proliferative agents for use in combination with the compounds of the present invention include cyclosporin A, paclitaxel, FK-506, and adriamycin.
  • Suitable anti-tumor agents for use in combination with the compounds of the present invention include paclitaxel, adriamycin, epothilones, cisplatin and carboplatin.
  • Compounds of the present invention may further be used in combination with nutritional supplements such as those described in U.S. Pat. No. 5,179,080, especially in combination with whey protein or casein, amino acids (such as leucine, branched amino acids and hydroxymethylbutyrate), triglycerides, vitamins (e.g., A, B6, B12, folate, C, D and E), minerals (e.g., selenium, magnesium, zinc, chromium, calcium and potassium), carnitine, lipoic acid, creatinine, B-hyroxy-B-methylbutyriate (Juven) and coenzyme Q-10.
  • nutritional supplements such as those described in U.S. Pat. No. 5,179,080, especially in combination with whey protein or casein, amino acids (such as leucine, branched amino acids and hydroxymethylbutyrate), triglycerides, vitamins (e.g., A, B6, B12, folate, C, D and E), minerals (
  • compounds of the present invention may be used in combination with therapeutic agents used in the treatment of sexual dysfunction, including but not limited to PDE-5 inhibitors, such as sildenafil or IC-351.
  • Compounds of the present invention may further be used in combination with antiresorptive agents, hormone replacement therapies, vitamin D analogues, elemental calcium and calcium supplements, cathepsin K inhibitors, MMP inhibitors, vitronectin receptor antagonists, Src SH 2 antagonists, vacular—H + -ATPase inhibitors, ipriflavone, fluoride, Tibolone, prostanoids, 17-beta hydroxysteroid dehydrogenase inhibitors and Src kinase inhibitors.
  • Compounds of the present invention may be used in combination with male contraceptives, such as nonoxynol 9 or therapeutic agents for the treatment of hair loss, such as minoxidil and finasteride or chemotherapeutic agents, such as with LHRH agonists.
  • male contraceptives such as nonoxynol 9
  • therapeutic agents for the treatment of hair loss such as minoxidil and finasteride
  • chemotherapeutic agents such as with LHRH agonists.
  • the compounds of the present invention may be used in combination with anti-cancer and cytotoxic agents, including but not limited to alkylating agents such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes such as L-asparaginase; farnesyl-protein transferase inhibitors; 5 ⁇ -reductase inhibitors; inhibitors of 17 ⁇ -hydroxysteroid dehydrogenase type 3; hormonal agents such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-d
  • anti-cancer and cytotoxic agents include but are not limited to mechlorethamine hydrochloride, cyclophosphamide, chlorambucil, melphalan, ifosfamide, busulfan, carmustin, lomustine, semustine, streptozocin, thiotepa, dacarbazine, methotrexate, thioguanine, mercaptopurine, fludarabine, pentastatin, cladribin, cytarabine, fluorouracil, doxorubicin hydrochloride, daunorubicin, idarubicin, bleomycin sulfate, mitomycin C, actinomycin D, safracins, saframycins, quinocarcins, discodermolides, vincristine, vinblastine, vinorelbine tartrate, etoposide, etoposide phosphate, teni
  • Preferred member of these classes include, but are not limited to, paclitaxel, cisplatin, carboplatin, doxorubicin, caminomycin, daunorubicin, aminopterin, methotrexate, methopterin, mitomycin C, ecteinascidin 743, or porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podophyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine and leurosine.
  • anticancer and other cytotoxic agents include the following: epothilone derivatives as found in German Patent No. 4138042.8; WO 97/19086, WO 98/22461, WO 98/25929, WO 98/38192, WO 99/01124, WO 99/02224, WO 99/02514, WO 99/03848, WO 99/07692, WO 99/27890, WO 99/28324, WO 99/43653, WO 99/54330, WO 99/54318, WO 99/54319, WO 99/65913, WO 99/67252, WO 99/67253 and WO 00/00485; cyclin dependent kinase inhibitors as found in WO 99/24416 (see also U.S.
  • the compounds of the formula I can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.
  • the present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds can also be administered liposomally.
  • compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the compounds of formula I can also be delivered through the oral cavity by sublingual and/or buccal administration.
  • Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG).
  • Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934).
  • HPC hydroxy propyl cellulose
  • HPMC hydroxy propyl methyl cellulose
  • SCMC sodium carboxy methyl cellulose
  • maleic anhydride copolymer e.g., Gantrez
  • agents to control release such as polyacrylic copolymer (e.g. Carbopol 934).
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug.
  • compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • the effective amount of a compound of the present invention can be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human of from about 0.01 to 2000 mg of active compound per day, which can be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to NHR-associated conditions.
  • transactivation assays of a transfected reporter construct and using the endogenous androgen receptor of the host cells.
  • the transactivation assay provides a method for identifying functional agonists and partial agonists that mimic, or antagonists that inhibit, the effect of native hormones, in this case, dihydrotestosterone (DHT).
  • DHT dihydrotestosterone
  • This assay can be used to predict in vivo activity as there is a good correlation in both series of data. See, e.g. T. Berger et al., J. Steroid Biochem. Molec. Biol. 773 (1992), the disclosure of which is herein incorporated by reference.
  • reporter plasmid For the transactivation assay a reporter plasmid is introduced by transfection (a procedure to induce cells to take foreign genes) into the respective cells.
  • This reporter plasmid comprising the cDNA for a reporter protein, such as secreted alkaline phosphatase (SEAP), controlled by prostate specific antigen (PSA) upstream sequences containing androgen response elements (AREs).
  • SEAP secreted alkaline phosphatase
  • PSA prostate specific antigen
  • AREs upstream sequences containing androgen response elements
  • This reporter plasmid functions as a reporter for the transcription-modulating activity of the AR.
  • the reporter acts as a surrogate for the products (mRNA then protein) normally expressed by a gene under control of the AR and its native hormone.
  • the transactivation assay is carried out in the presence of constant concentration of the natural AR hormone (DHT) known to induce a defined reporter signal.
  • DHT natural AR hormone
  • Increasing concentrations of a suspected antagonist will decrease the reporter signal (e.g., SEAP production).
  • exposing the transfected cells to increasing concentrations of a suspected agonist will increase the production of the reporter signal.
  • LNCaP and MDA 453 cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in RPMI 1640 or DMEM medium supplemented with 10% fetal bovine serum (FBS; Gibco) respectively.
  • the respective cells were transiently transfected by electroporation according to the optimized procedure described by Heiser, 130 Methods Mol. Biol., 117 (2000), with the pSEAP2/PSA540/Enhancer reporter plasmid.
  • the reporter plasmid was constructed as follows: commercial human placental genomic DNA was used to generate by Polymerase Cycle Reaction (PCR) a fragment containing the BglII site (position 5284) and the Hind III site at position 5831 of the human prostate specific antigen promoter (Accession # U37672), Schuur, et al., J. Biol. Chem., 271 (12): 7043-51 (1996). This fragment was subcloned into the pSEAP2/basic (Clontech) previously digested with BglII and HindIII to generate the pSEAP2/PSA540 construct.
  • PCR Polymerase Cycle Reaction
  • Each cell suspension was distributed into two Gene Pulser Cuvetts (Bio-Rad) which then received 8 ⁇ g of the reporter construct, and electoporated using a Bio-Rad Gene Pulser at 210 volts and 960 ⁇ Faraday. Following the transfections the cells were washed and incubated with media containing charcoal stripped fetal bovine serum in the absence (blank) or presence (control) of 1 nM dihydrotestosterone (DHT; Sigma Chemical) and in the presence or absence of the standard anti-androgen bicalutamide or compounds of the present invention in concentrations ranging from 10 ⁇ 10 to 10 ⁇ 5 M (sample). Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory workstation.
  • a mix of a tetrazolium compound (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; MTS) and an electron coupling reagent (phenazine methosulfate; PMS) are added to the cells.
  • MTS Olet's reagent
  • PMS phenazine methosulfate
  • the reporter plasmid utilized was comprised of the cDNA for the reporter SEAP protein, as described for the AR specific transactivation assay. Expression of the reporter SEAP protein was controlled by the mouse mammary tumor virus long terminal repeat (MMTV LTR) sequences that contains three hormone response elements (HREs) that can be regulated by both GR and PR see, e.g. G. Chalepakis et al., Cell, 53(3), 371 (1988). This plasmid was transfected into A549 cells, which expresses endogenous GR, to obtain a GR specific transactivation assay.
  • MMTV LTR mouse mammary tumor virus long terminal repeat
  • HREs hormone response elements
  • A549 cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in RPMI 1640 supplemented with 10% fetal bovine serum (FBS; Gibco). Determination of the GR specific antagonist activity of the compounds of the present invention was identical to that described for the AR specific transactivation assay, except that the DHT was replaced with 5 nM dexamethasone (Sigma Chemicals), a specific agonist for GR. Determination of the GR specific agonist activity of the compounds of the present invention was performed as described for the AR transactivation assay, wherein one measures the activation of the GR specific reporter system by the addition of a test compound, in the absence of a known GR specific agonists ligand.
  • the reporter plasmid utilized was comprised of the cDNA for the reporter SEAP protein, as described for the AR specific transactivation assay. Expression of the reporter SEAP protein was controlled by the mouse mammary tumor virus long terminal repeat (MMTV LTR) sequences that contains three hormone response elements (HRE's) that can be regulated by both GR and PR. This plasmid was transfected into T47D, which expresses endogenous PR, to obtain a PR specific transactivation assay. T47D cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in DMEM medium supplemented with 10% fetal bovine serum (FBS; Gibco).
  • FBS fetal bovine serum
  • Determination of the PR specific antagonist activity of the compounds of the present invention was identical to that described for the AR specific transactivation assay, except that the DHT was replaced with 1 nM Promegastone (NEN), a specific agonist for PR. Determination of the PR specific agonist activity of the compounds of the present invention was performed as described for the AR transactivation assay, wherein one measures the activation of the PR specific reporter system by the addition of a test compound, in the absence of a known PR specific agonists ligand.
  • human LNCaP cells T877A mutant AR or MDA 453 (wild type AR) in 96-well microtiter plates containing RPMI 1640 or DMEM supplemented with 10% charcoal stripped CA-FBS (Cocaleco Biologicals) respectively, were incubated at 37° C. to remove any endogenous ligand that might be complexed with the receptor in the cells. After 48 h, either a saturation analysis to determine the K d for tritiated dihydrotestosterone, [ 3 H]-DHT, or a competitive binding assay to evaluate the ability of test compounds to compete with [ 3 H]-DHT were performed.
  • media RPMI 1640 or DMEM—0.2% CA-FBS
  • [ 3 H]-DHT in concentrations ranging from 0.1 nM to 16 nM
  • an aliquot of the total binding media at each concentration of [ 3 H]-DHT was removed to estimate the amount of free [ 3 H]-DHT.
  • the difference between the total binding and the non-specific binding was defined as specific binding.
  • the specific binding was evaluated by Scatchard analysis to determine the K d for [ 3 H]-DHT. See e.g. D.
  • test compounds media containing 1 nM [ 3 H]-DHT and compounds of the invention (“test compounds”) in concentrations ranging from 10 ⁇ 10 to 10 ⁇ 5 M were added to the cells. Two replicates were used for each sample. After 4 h at 37° C., cells were washed, harvested and counted as described above. The data was plotted as the amount of [ 3 H]-DHT (% of control in the absence of test compound) remaining over the range of the dose response curve for a given compound. The concentration of test compound that inhibited 50% of the amount of [ 3 H]-DHT bound in the absence of competing ligand was quantified (IC 50 ) after log-logit transformation.
  • K 1 IC 50 (1+( 3 H-DHT)/K d for 3 H-DHT) After correcting for non-specific binding, IC 50 values were determined.
  • the IC 50 is defined as the concentration of competing ligand needed to reduce specific binding by 50%.
  • the K d s for [ 3 H]-DHT for MDA 453 and LNCaP were 0.7 and 0.2 nM respectively.
  • the first assay uses a cell line, Stable 1 (clone #72), which expresses the full length rat androgen receptor but requires the transient transfection of an enhancer/reporter. This cell line was derived from C2C12 mouse moyoblast cells.
  • the second assay uses a cell line, Stable 2 (clone #133), derived from Stable 1 which expresses both rAR and the enhancer/luciferase reporter.
  • the enhancer/reporter construct used in this system is pGL3/2XDR-1/luciferase.
  • 2 ⁇ DR-1 was reported to be an AR specific response element in CV-1 cells, Brown et. al. The Journal of Biological Chemistry 272, 8227-8235, (1997). It was developed by random mutagenesis of an AR/GR consensus enhancer sequence.
  • Stable 1 cells are plated in 96 well format at 6,000 cells/well in high glucose DMEM without phenol red (Gibco BRL, Cat. No.: 21063-029) containing 10% charcoal and dextran treated FBS (HyClone Cat. No.: SH30068.02), 50 mM HEPES Buffer (Gibco BRL, Cat. No.: 15630-080), 1 ⁇ MEM Na Pyruvate (Gibco BRL, Cat. No.: 11360-070), 0.5 ⁇ Antibiotic-Antimycotic, and 800 ⁇ g/mL Geneticin (Gibco BRL, Cat. No.: 10131-035).
  • cells are transfected with pGL3/2XDR-1/luciferase using LipofectAMINE PlusTM Reagent (Gibco BRL, Cat. No.: 10964-013).
  • LipofectAMINE PlusTM Reagent Gibco BRL, Cat. No.: 10964-013
  • 5 ng/well pGL3/2XDR-1/luciferase DNA and 50 ng/well Salmon Sperm DNA (as carrier) are diluted with 5 ⁇ l/well Opti-MEMem media (Gibco BRL, Cat. No.: 31985-070).
  • Opti-MEMem media Gibco BRL, Cat. No.: 31985-070
  • 0.5 ⁇ l/well Plus reagent is added. This mixture is incubated for 15 min at rt.
  • LipofectAMINE reagent is diluted with 5 ⁇ l/well Opti-MEM.
  • the DNA mixture is then combined with the LipofectAMINE mixture and incubated for an additional 15 min at rt. During this time, the media from the cells is removed and replaced with 60 ⁇ l/well of Opti-MEM. To this is added 10 ⁇ l/well of the DNA/LipofectAMINE transfection mixture. The cells are incubated for 4 h.
  • the transfection mixture is removed from the cells and replaced with 90 ⁇ l of media as in #1 above.
  • the Steady-GloTM Luciferase Assay System is used to detect activity according to the manufacturer's instructions (Promega, Cat. No.: E2520).
  • Stable 2 cells are plated in 96 well format at 6,000 cells/well in high glucose DMEM without phenol red (Gibco BRL, Cat. No.: 21063-029) containing 10% charcoal and dextran treated FBS (HyClone Cat. No.: SH30068.02), 50 mM HEPES Buffer (Gibco BRL, Cat. No.: 15630-080), 1 ⁇ MEM Na Pyruvate (Gibco BRL, Cat. No.: 11360-070), 0.5 ⁇ Antibiotic-Antimycotic, 800 ⁇ g/mL Geneticin (Gibco BRL, Cat. No.: 10131-035) and 800 ⁇ g/mL Hygromycin ⁇ (Gibco BRL, Cat. No.: 10687-010).
  • the Steady-GloTM Luciferase Assay System is used to detect activity according to the manufacturer's instructions (Promega, Cat. No. E2520).
  • test compounds were tested (“test compounds”) on the proliferation of human prostate cancer cell lines.
  • MDA PCa2b cells a cell line derived from the metastasis of a patient that failed castration, Navone et al., Clin. Cancer Res., 3, 2493-500 (1997), were incubated with or without the test compounds for 72 h and the amount of [ 3 H]-thymidine incorporated into DNA was quantified as a way to assess number of cells and therefore proliferation.
  • the MDA PCa2b cell line was maintained in BRFF-HPC1 media (Biological Research Faculty & Facility Inc., MD) supplemented with 10% FBS.
  • cells were plated in Biocoated 96-well microplates and incubated at 37° C. in 10% FBS (charcoal-stripped)/BRFF-BMZERO (without androgens). After 24 h, the cells were treated in the absence (blank) or presence of 1 nM DHT (control) or with test compounds (sample) of the present invention in concentrations ranging from 10 ⁇ 10 to 10 ⁇ 5 M. Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory work station. Seventy-two h later 0.44 uCi.
  • % Inhibition 100 ⁇ (1 ⁇ [average control ⁇ average blank /average sample ⁇ average blank ]) Data was plotted and the concentration of compound that inhibited 50% of the [3H]_Thymidine incorporation was quantified (IC 50 ).
  • test compounds The ability of compounds of the present invention (“test compounds”) to modulate the function of the AR was determined by testing said compounds in a proliferation assay using the androgen responsive murine breast cell line derived from the Shionogi tumor, Hiraoka et al., Cancer Res., 47, 6560-6564 (1987).
  • Stable AR dependent clones of the parental Shionogi line were established by passing tumor fragments under the general procedures originally described in Tetuo, et. al., Cancer Research 25, 1168-1175 (1965). From the above procedure, one stable line, SC114, was isolated, characterized and utilized for the testing of example compounds.
  • SC114 cells were incubated with or without the test compounds for 72 h and the amount of [3H]-thymidine incorporated into DNA was quantified as a surrogate endpoint to assess the number of cells and therefore the proliferation rate as described in Suzuki et. al., J. Steroid Biochem. Mol. Biol. 37, 559-567 (1990).
  • the SC114 cell line was maintained in MEM containing 10 ⁇ 8 M testosterone and 2% DCC-treated FCS.
  • cells were plated in 96-well microplates in the maintenance media and incubated at 37° C.
  • the medium was changed to serum free medium [Ham's F-12:MEM (1; 1, v/v) containing 0.1% BSA] with (antagonist mode) or without (agonist mode) 10 ⁇ 8 M testosterone and the test compounds of the present invention in concentrations ranging from 10 ⁇ 10 to 10 ⁇ 5 M.
  • Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory work station. Seventy two h later 0.44uCi of [3H]-Thymidine (Amersham) was added per well and incubated for another 2 h followed by tripsinization, and harvesting of the cells onto GF/B filters. Micro-scint PS were added to the filters before counting them on a Beckman TopCount.
  • % Inhibition 100 ⁇ (1 ⁇ [average sample ⁇ average blank /average control ⁇ average blank ]) Data was plotted and the concentration of compound that inhibited 50% of the [ 3 H]-Thymidine incorporation was quantified (IC 50 ).
  • the AP-1 assay is a cell-based luciferase reporter assay.
  • A549 cells which contain endogenous glucocorticoid receptor, were stably transfected with an AP-1 DNA binding site attached to the luciferase gene. Cells are then grown in RPMI+10% fetal calf serum (charcoal-treated)+Penicillin/Streptomycin with 0.5 mg/mL geneticin. Cells are plated the day before the assay at approximately 40000 cells/well. On assay day, the media is removed by aspiration and 20 ⁇ L assay buffer (RPMI without phenol red+10% FCS (charcoal-treated)+Pen/Strep) is added to each well.
  • test compounds the compounds of the present invention
  • dexamethasome 100 nM in DMSO, positive control
  • the plates are then pre-incubated for 15 min at 37° C., followed by stimulation of the cells with 10 ng/mL PMA.
  • the plates are then incubated for 7 h at 37° C. after which 40 ⁇ L luciferase substrate reagent is added to each well.
  • Activity is measured by analysis in a luminometer as compared to control experiments treated with buffer or dexamethasome.
  • Activity is designated as % inhibition of the reporter system as compared to the buffer control with 10 ng/mL PMA alone.
  • the control, dexamethasone, at a concentration of ⁇ 10 ⁇ M typically suppresses activity by 65%.
  • Test compounds which demonstrate an inhibition of PMA induction of 50% or greater at a concentration of test compound of ⁇ 10 ⁇ M are deemed active.
  • Testosterone Propionate (TP) (3 mg/rat/day, subcutaneous)
  • TP plus Bicalutamide administered p.o. in PEGTW, QD
  • a recognized antiandrogen as a reference compound.
  • test compound a compound of the present invention was administered (p.o. in PEGTW, QD) with TP (s.c. as administered in group 2) in a range of doses.
  • test compound a compound of the present invention was administered alone (p.o. in PEGTW, QD) in a range of doses.
  • the animals were sacrificed by carbon dioxide, and the levator ani, seminal vesicle and ventral prostate weighed.
  • the levator ani muscle and sexual organ weights were first standardized as mg per 100 g of body weight, and the increase in organ weight induced by TP was considered as the maximum increase (100%).
  • Super-anova one factor was used for statistical analysis.
  • the gain and loss of sexual organ weight reflect the changes of the cell number (DNA content) and cell mass (protein content), depending upon the serum androgen concentration. See Y. Okuda et al., J. Urol., 145, 188-191 (1991), the disclosure of which is herein incorporated by reference. Therefore, measurement of organ wet weight is sufficient to indicate the bioactivity of androgens and androgen antagonist. In immature castrated rats, replacement of exogenous androgens increases levator ani, seminal vesicles (SV) and prostate in a dose dependent manner.
  • SV seminal vesicles
  • the maximum increase in organ weight was 4 to 5-fold when dosing 3 mg/rat/day of testosterone (T) or 1 mg/rat/day of testosterone propionate (TP) for 3 days.
  • the EC 50 of T and TP were about 1 mg and 0.03 mg, respectively.
  • the increase in the weight of the VP and SV also correlated with the increase in the serum T and DHT concentration.
  • administration of T showed 5-times higher serum concentrations of T and DHT at 2 h after subcutaneous injection than that of TP, thereafter, these high levels declined very rapidly.
  • the serum concentrations of T and DHT in TP-treated animals were fairly consistent during the 24 h, and therefore, TP showed about 10-30-fold higher potency than free T.
  • the title compound was prepared from 4-cyanonaphthylamine in a similar fashion to that described in Step 5 of Example 1 for the preparation of an aryl isocyanate from an aryl amine.
  • Step 1 compound (0.51 g, 1.96 mmol) in CH 2 Cl 2 (2 mL) cooled to 0° C. was added i-Pr 2 EtN (0.37 mL, 2.16 mmol). After stirring at 0° C. for 20 min, cis-3-hydroxyproline methyl ester (0.34 g, 1.75 mmol) in CH 2 Cl 2 (1 mL) solution was added, along with 4 ⁇ molecular sieves (0.5 g) and the resulting mixture stirred at rt until urea formation was complete ( ⁇ 1 h).
  • Step 1 compound (6.20 g, 26.7 mmol) in MeOH (100 mL) was treated with saturated aqueous LiOH (15 mL), and the reaction mixture was stirred at rt overnight ( ⁇ 16 h). The resulting mixture was concentrated in vacuo to dryness and partitioned between a mixed solvent (50% Et 2 O:50% EtOAc) and H 2 O. The H 2 O layer was acidified to pH 2-3 and extracted with EtOAc (3 ⁇ 20 mL). The EtOAc was dried over Na 2 SO 4 and concentrated in vacuo to give a yellow solid (2.1 g) in 38% yield.
  • Step 2 compound (1.37 g, 5.3 mmol) in DMF (30 mL) was added HOBT (1.94 g, 14.4 mmol), EDAC (1.85 g, 9.6 mmol), followed by TEA (2 mL, 14.4 mmol), and the reaction was stirred at rt overnight ( ⁇ 16 h). The resulting mixture was partitioned between EtOAc (100 mL) and H 2 O (4 ⁇ 50 mL). The EtOAc layer was dried over Na 2 SO 4 and was evaporated in vacuo. The crude product was cleaned up by silica gel chromatography using 10% MeOH in CH 2 Cl 2 to afford 1.01 g of the title compound as an off-white solid.

Abstract

There are provided nuclear hormone receptor modulating compounds of formula I
Figure US20080108691A1-20080508-C00001

wherein R1, R2, R3, X, Y, n and G are as described herein. Further provided are methods of using such compounds for the treatment of nuclear hormone receptor-associated conditions, such as age related diseases, for example sarcopenia, and also provided are pharmaceutical compositions containing such compounds. Other embodiments are also disclosed.

Description

    RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 10/712,456, filed Nov. 13, 2003, and thus claims priority benefit under 35 U.S.C. § 119(e) of U.S. provisional Application No. 60/426,694, filed Nov. 15, 2002, the contents of which are herein incorporated by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to open chain prolyl urea and thiourea related compounds, methods of using such compounds in the treatment of androgen receptor-associated conditions, such as age-related diseases, for example sarcopenia, and to pharmaceutical compositions containing such compounds.
  • BACKGROUND OF THE INVENTION
  • Nuclear hormone receptors (NHR's) constitute a large super-family of structurally-related and sequence-specific gene regulators scientists have named “ligand-dependent transcription factors.” R. M. Evans, Science, 240:889 (1988). The steroid binding NHR's (SB-NHR's) form a recognized subset of the NHR's, including the progesterone receptor (PR), androgen receptor (AR), estrogen receptor (ER), glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). The conventional nuclear hormone receptors are generally transactivators in the presence of ligand, which selectively bind to the NHR in a way that effects gene transcription. In the absence of a corresponding ligand, some of the orphan receptors behave as if they are transcriptionally inert. Others, however, behave as either constitutive activators or repressors. These orphan nuclear hormone receptors are either under the control of ubiquitous ligands that have not been identified, or do not need to bind ligand to exert these activities.
  • The AR is a ligand-activated transcriptional regulatory protein that mediates induction of male sexual development and function through its activity with endogenous androgens. In addition, androgens are associated with male and female maintenance of muscle mass and strength, bone mass and erythropoiesis. Androgens, such as testosterone, also play an important role in many physiological processes, such as differentiation of male internal and external genitalia, development and maintenance of male secondary sexual characteristics (e.g., the development of prostate, seminal vesicles, penis, scrotum, skeletal muscle, redistribution of body fat, stimulation of long bone growth, closure of epiphyses, development of male hair growth pattern and enlargement of larynx), the maintenance of sexual behavior and function (e.g., libido and potency) and spermatogenesis (in man).
  • As one ages, the serum androgen concentration in the body declines. The age dependent decline in androgens is associated with changes in body composition for men and women, such as a lower percentage of muscle mass and an increase in body fat, e.g., sarcopenia. In this regard, modulation of the AR gene can have an impact on the physiological effects associated with androgen production. However, the effectiveness of known modulators of steroid receptors is often tempered by their undesired side-effect profile, particularly during long-term administration. For example, the administration of synthetic androgens has been associated with liver damage, prostate cancer, adverse effects on male sexual function and adverse effects associated with cardiovascular and erythropoietic function.
  • Numerous synthetically-derived steroidal and non-steroidal agonists and antagonists have been described for the members of the SB-NHR family. Many of these agonist and antagonist ligands are used clinically in man to treat a variety of medical conditions. RU486 (mifepristone) is an example of a synthetic antagonist of the PR, which is utilized as a birth control agent (Vegeto et al., Cell 69: 703-713 (1992)). Flutamide is an example of an antagonist of the AR, which is utilized for the treatment of prostate cancer (Neri et al, Endo. 91, 427-437 (1972)). Tamoxifen is an example of a tissue-selective modulator of the ER function, that is used in the treatment of breast cancer (Smigel J. Natl. Cancer Inst. 90, 647-648 (1998)). Tamoxifen can function as an antagonist of the ER in breast tissue while acting as an agonist of the ER in bone (Grese et al., Proc. Natl. Acad. Sci. USA 94, 14105-14110 (1997)). Because of the tissue-selective effects seen for Tamoxifen, this agent, and agents like it, are referred to as tissue-selective estrogen receptor modulators. In addition to synthetically-derived non-endogenous ligands, non-endogenous ligands for NHR's can be obtained from food sources (Regal et al., Proc. Soc. Exp. Biol. Med. 223, 372-378 (2000) and Hempstock et al., J. Med. Food 2, 267-269 (1999)). The flavanoid phytoestrogens are an example of an unnatural ligand for SB-NHR's that are readily obtained from a food source such as soy (Quella et al., J. Clin. Oncol. 18, 1068-1074 (2000) and Banz et al., J. Med. Food 2, 271-273 (1999)). The ability to modulate the transcriptional activity of an individual NHR by the addition of a small molecule ligand, makes these receptors ideal targets for the development of pharmaceutical agents for a variety of disease states.
  • As mentioned above, non-natural ligands can be synthetically engineered to serve as modulators of the function of NHR's. In the case of SB-NHR's, engineering of an unnatural ligand can include the identification of a core structure which mimics the natural steroid core system. This can be achieved by random screening against several SB-NHR's, or through directed approaches using the available crystal structures of a variety of NHR ligand binding domains (Bourguet et al., Nature 375, 377-382 (1995), Brzozowski, et al., Nature 389, 753-758 (1997), Shiau et al., Cell 95, 927-937 (1998) and Tanenbaum et al., Proc. Natl. Acad. Sci. USA 95, 5998-6003 (1998)). Differential substitution about such a steroid mimic core can provide agents with selectivity for one receptor versus another. In addition, such modifications can be employed to obtain agents with agonist or antagonist activity for a particular SB-NHR. Differential substitution about the steroid mimic core can result in the formation of a series of high affinity agonists and antagonists with specificity for, for example, ER versus PR versus AR versus GR versus MR. Such an approach of differential substitution has been reported, for example, for quinoline based modulators of steroid NHR in J. Med. Chem., 41, 623 (1998); WO 9749709; U.S. Pat. No. 5,696,133; U.S. Pat. No. 5,696,130; U.S. Pat. No. 5,696,127; U.S. Pat. No. 5,693,647; U.S. Pat. No. 5,693,646; U.S. Pat. No. 5,688,810; U.S. Pat. No. 5,688,808 and WO 9619458, all incorporated herein by reference.
  • Accordingly, identification of compounds which have good specificity for one or more steroid receptors, but which have reduced or no cross-reactivity for other steroid or intracellular receptors, would be of significant value in the treatment of male and female hormone-responsive diseases. There is, therefore, a need in the art for the identification of selective modulators of the steroid binding nuclear hormone receptors, particularly non-steroidal, non-toxic tissue selective androgen receptor modulators, which activate the androgen receptor in skeletal muscle while demonstrating limited or neutral effect on other androgen responsive (e.g., prostate) tissues.
  • SUMMARY OF THE INVENTION
  • In accordance with illustrative embodiments and demonstrating features of the present invention, compounds are provided which are capable of modulating the function of a nuclear hormone receptor. Preferably the compounds are selective androgen receptor modulators, and have the general formula I
    Figure US20080108691A1-20080508-C00002

    wherein
  • R1 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl and CH2OR4;
  • R2 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo, heteroaryl or substituted heteroaryl and CH2OR4;
  • R3 is selected from the group consisting of hydrogen (H), alkyl or substituted alkyl, CH2OR4, OR2, SR2, halo, NHR2, NHCOR4, NHCO2R4, NHCONR4R4′ and NHSO2R4;
  • R4 and R4′ for each occurrence are each independently selected from the group consisting of hydrogen(H), alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo and heteroaryl or substituted heteroaryl;
  • G is a mono- or polycyclic ring system selected from the group consisting of aryl, heterocyclo and heteroaryl, wherein said ring system may optionally substituted with one or more substituents selected from the group consisting of hydrogen, halo, CN, CF3, OR4, CO2R4, NR4R4′, CONR4R4′, CH2OR4, SR4, SOR4, SO2R4, NO2, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
  • X is a linking group selected from the group consisting of NR4 and CHR4;
  • Y is selected from the group consisting of oxygen (O), NR4, NOR4 and sulfur (S);
  • Z is oxygen (—O—) or NR4; and
  • n is an integer of 1 or 2.
  • The definition of formula I above includes of all prodrug esters, stereoisomers and pharmaceutically acceptable salts of formula I.
  • The compounds of formula I modulate the function of the nuclear hormone receptors, particularly the androgen receptor, and include compounds which are, for example, selective agonists, partial agonists, antagonists or partial antagonists of the androgen receptor. Preferably the compounds of formula I possess activity as agonists of the androgen receptor and may be used in the treatment of diseases or disorders associated with androgen receptor activity, such as maintenance of muscle strength and function (e.g., in the elderly); reversal or prevention of frailty or age-related functional decline (“ARFD”) in the elderly (e.g., sarcopenia); prevention of catabolic side effects of glucocorticoids; prevention and treatment of reduced bone density or growth (e.g., osteoporosis and osteopenia); treatment of chronic fatigue syndrome (CFS); chronic myalgia; treatment of acute fatigue syndrome and muscle loss following elective surgery (e.g., post-surgical rehabilitation); acceleration of wound healing; accelerating bone fracture repair (such as accelerating the recovery of hip fracture patients); treatment of wasting secondary to fractures and wasting in connection with chronic obstructive pulmonary disease (COPD), chronic liver disease, AIDS, weightlessness, cancer cachexia, burn and trauma recovery, chronic catabolic state (e.g., coma), eating disorders (e.g., anorexia) and chemotherapy.
  • The present invention provides for compounds of formula I, pharmaceutical compositions employing such compounds and for methods of using such compounds. In particular, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I, alone or in combination with a pharmaceutically acceptable carrier.
  • Further, in accordance with the present invention, a method is provided for preventing, inhibiting or treating the progression or onset of diseases or disorders associated with nuclear hormone receptors, particularly, the androgen receptor, such as the diseases or disorders defined above and hereinafter, wherein a therapeutically effective amount of a compound of formula I is administered to a mammalian, i.e., human, patient in need of treatment.
  • The compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more other agent(s) active in the therapeutic areas described herein.
  • In addition, a method is provided for preventing, inhibiting or treating the diseases as defined above and hereinafter, wherein a therapeutically effective amount of a combination of a compound of formula I and another type of therapeutic agent, is administered to a human patient in need of treatment.
  • Preferred are compounds of formula I wherein
  • R1 is hydrogen (H) or alkyl;
  • R2 is hydrogen (H) or alkyl;
  • R3 is hydroxyl (OH);
  • X is NR4;
  • Y is O;
  • Z is O; and
  • n is 1
  • Further preferred embodiments include compounds of formula I where G is selected from:
    Figure US20080108691A1-20080508-C00003

    wherein
  • R8, R9, R10 and R11 in each functional group are each independently selected from the group consisting of hydrogen (H), NO2, CN, CF3, OR4, CO2R4, NR4′, CONR4R4′, CH2OR4, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
  • A to F is each independently selected from N or CR1;
  • J, K, L, P and Q are each independently selected from NR12, O, S, SO, SO2 or CR12R12′;
  • R12 and R12′ in each functional group are each independently selected from a bond or R1; and
  • m is an integer of 0 or 1.
  • Preferred are compounds of formula I where RS is CN.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following abbreviations are employed herein:
  • Chiralpak®=Trademark of Chiral Technologies, Inc. Eaton, Pa.
  • DBU=1,8-diazabicyclo[5.4.0]undec-7-ene
  • AcOH=acetic acid
  • DMPU=1,3-dimethyl-3,4,5,6-tetrahydro-2(1H)-pyrimidinone
  • EtOAc=ethyl acetate
  • HPLC=high performance liquid chromatography
  • MeOH=methanol
  • MS or Mass Spec=mass spectrometry
  • YMC®=trademark of YMC Co, Ltd., Kyoto, Japan
  • CuBr=copper(I) bromide
  • CuCN=copper(I) cyanide
  • CsF=cesium fluoride
  • Et3N=triethylamine
  • DCC=1,3-dicyclohexylcarbodiimide
  • DEAD=diethyl azodicarboxylate
  • LDA=lithium diisopropylamide
  • NMP=1-methyl-2-pyrrolidinone
  • KOH=potassium hydroxide
  • Pd/C=palladium on activated charcoal
  • TFA=trifluoroacetic acid
  • THF=tetrahydrofuran
  • mp.=melting point
  • min=minute(s)
  • h=hour(s)
  • L=liter
  • mL=milliliter
  • t=microliter
  • g=gram(s)
  • mg=milligram(s)
  • mol=moles
  • mmol=millimole(s)
  • nM=nanomolar
  • rt=room temperature
  • The following definitions apply to the terms as used throughout this specification, unless otherwise limited in specific instances.
  • As used herein, the term “alkyl” denotes branched or unbranched hydrocarbon chains, preferably having about 1 to about 8 carbons, such as, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, 2-methylpentyl pentyl, hexyl, isohexyl, heptyl, 4,4-dimethyl pentyl, octyl, 2,2,4-trimethylpentyl and the like. “Substituted alkyl” includes an alkyl group optionally substituted with one or more functional groups which are commonly attached to such chains, such as, hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, amido, and the like to form alkyl groups such as trifluoro methyl, 3-hydroxyhexyl, 2-carboxypropyl, 2-fluoroethyl, carboxymethyl, cyanobutyl and the like.
  • Unless otherwise indicated, the term “cycloalkyl” as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl,
    Figure US20080108691A1-20080508-C00004
  • “Substituted cycloalkyl” includes a cycloalkyl group optionally substituted with 1 or more substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol and/or alkylthio and/or any of the substituents included in the definition of “substituted alkyl.”
  • Unless otherwise indicated, the term “alkenyl” as used herein by itself or as part of another group refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbons in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12-tetradecatrienyl, and the like. “Substituted alkenyl” includes an alkenyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • Unless otherwise indicated, the term “alkynyl” as used herein by itself or as part of another group refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons in the normal chain, which include one or more triple bonds in the normal chain, such as 2-propynyl, 3butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl,3-undecynyl, 4-dodecynyl and the like. “Substituted alkynyl” includes an alkynyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • The terms “arylalkyl”, “arylalkenyl” and “arylalkynyl” as used alone or as part of another group refer to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent. Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like. “Substituted arylalkyl” includes arylalkyl groups wherein the aryl portion is optionally substituted with one or more substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.”
  • The term “halogen” or “halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
  • Unless otherwise indicated, the term “aryl” or “Ar” as employed herein alone or as part of another group refers to monocyclic and polycyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2-naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings), for example
    Figure US20080108691A1-20080508-C00005
  • “Substituted aryl” includes an aryl group optionally substituted with one or more functional groups, such as halo, haloalkyl, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl-alkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino includes 1 or 2 substituents (which are alkyl, aryl or any of the other aryl compounds mentioned in the definitions), thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl, alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino or arylsulfonaminocarbonyl and/or any of the alkyl substituents set out herein.
  • Unless otherwise indicated, the term “heteroaryl” as used herein alone or as part of another group refers to a 5- or 7-membered aromatic ring which includes 1, 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocycloalkyl ring (e.g. benzothiophenyl, indolyl), and includes possible N-oxides. “Substituted heteroaryl” includes a heteroaryl group optionally substituted with 1 to 4 substituents, such as the substituents included above in the definition of “substituted alkyl” and “substituted cycloalkyl.” Examples of heteroaryl groups include the following:
    Figure US20080108691A1-20080508-C00006

    and the like.
  • The term “heterocyclo”, heterocycle or heterocyclic ring, as used herein, represents an unsubstituted or substituted stable 5- to 7-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, and oxadiazolyl.
  • The compounds of formula I can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of formula I have, for example, at least one basic center, they can form acid addition salts. These are formed, for example, with strong inorganic acids, such as mineral acids, for example sulfuric acid, phosphoric acid or a hydrohalic acid, with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted, for example, by halogen, for example acetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (C1-C4) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen, for example methyl- or p-toluene-sulfonic acid. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds of formula I having at least one acid group (for example COOH) can also form salts with bases. Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as morpholine, thiomorpholine, piperidine, pyrrolidine, a mono, di or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine. Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula I or their pharmaceutically acceptable salts, are also included.
  • Preferred salts of the compounds of formula I which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate or nitrate.
  • Preferred salts of the compounds of formula I which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines.
  • The term “modulator” refers to a chemical compound with capacity to either enhance (e.g., “agonist” activity) or inhibit (e.g., “antagonist” activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • The term “prodrug esters” as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of formula I with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like.
  • Any compound that can be converted in vivo to provide the bioactive agent (i.e., the compound of formula I) is a prodrug within the scope and spirit of the invention.
  • Various forms of prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in:
  • a) The Practice of Medicinal Chemistry, Camille G. Wermuth et al., Ch. 31, (Academic Press, 1996),
  • b) Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985);
  • c) A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds. Ch 5, pgs 113-191 (Harwood Academic Publishers, 1991).
  • Said references are incorporated herein by reference.
  • An administration of a therapeutic agent of the invention includes administration of a therapeutically effective amount of the agent of the invention. The term “therapeutically effective amount” as used herein refers to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance.
  • All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. The compounds of the present invention can have asymmetric centers at any of the carbon atoms including any one of the R substituents. Consequently, compounds of formula I can exist in enantiomeric or diastereomeric forms or in mixtures thereof. The processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When diastereomeric or enantiomeric products are prepared, they can be separated by conventional methods for example, chromatographic, chiral HPLC or fractional crystallization.
  • The compounds of formula I of the invention can be prepared as shown in the following reaction schemes and description thereof, as well as relevant published literature procedures that may be used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working Examples.
    Figure US20080108691A1-20080508-C00007
  • As illustrated in Scheme I, selected compounds of formula I can be prepared from suitably protected intermediates of formula III. Intermediates of formula II an be obtained commercially, can be prepared by methods known in the literature or can be readily prepared by one skilled in the art. Intermediates of formula III can be obtained commercially, can be prepared by methods known in the literature or can be readily prepared by one skilled in the art. Treatment of an intermediate of formula II with suitable coupling reagents, such as EDAC and HOBT together with a suitable coupling partner such as a compound of the formula HZR1 yields an intermediate of the formula III. Treatment of III with an intermediate of formula G-NCY yields compounds of the formula I. The intermediates of formula G-NCY can be obtained, for example, from commercially available isocyanates, thioisocyanates and carbodiimides or can be readily prepared by one skilled in the art. A compound of formula Ia can subsequently be obtained by any one of various methods for amide alkylation, such as for example, treatment of a compound of formula I with a base such as NaH, followed by treatment with an electrophile such as R4—Br to give the alkylated product Ia.
    Figure US20080108691A1-20080508-C00008
  • Scheme II describes a method for preparing compounds of formula I wherein a suitably protected intermediate of formula III is treated with a phosgene like reagent of formula C1-E-Cl in the presence of a base, such as NaHCO3 or triethylamine, to yield an intermediate of formula IV. The phosgene like intermediates of formula C1-E-Cl can be obtained from commercially available sources, can be prepared by methods known in the literature or can be readily be prepared by one skilled in the art.
  • Phosgene equivalents such as carbonyldiimidazoles, may alternatively be substituted for Cl-E-Cl. The intermediate of formula IV can be reacted with an amine of formula H2N-G in the presence of a base, such as triethylamine, to give a compound of formula I. The amine intermediates H2N-G can be obtained from commercially available sources, can be prepared by methods known in the literature or can be readily be prepared by one skilled in the art.
    Figure US20080108691A1-20080508-C00009
  • Scheme III describes a method to prepare isocyanates and isothiocyanates of general formula Y═C═N-G, wherein intermediates H2N-G are treated with phosgene, thiophosgene, or a (thio)phosgene-like reagent in the presence of an inorganic base such as sodium bicarbonate, or a organic base such as diisopropylethylamine in a solvent such as dichloromethane to afford an isocyanate or isothiocyanate of formula Y═C═N-G.
    Figure US20080108691A1-20080508-C00010
  • Scheme IV describes a method of preparing certain intermediate compounds of the general formula IX (G) for incorporation into compounds of the general formula I by the methods described in the preceeding schemes. Alkyl or other functionality can be introduced into the 2-position of substituted anilines such as 4-chloro-3-trifluoromethylaniline (intermediate V, where R11═CF3, Hal=Cl) for example by first protecting the amino functionality with a suitable protecting group, such as tert-butyloxycarbonyl (BOC) using tert-butyloxycarbonyl (BOC) chloride or tert-butyloxycarbonyl anhydride and a suitable base such as triethylamine to give an intermediate of the formula VI. Optional functionality, such as an alkyl group (R10=alkyl) can then be installed at the 2-position of the aryl ring by first metallation of a compound of the formula VI with a suitable base such as n-butyllithium in an appropriate solvent such as THF, followed by quenching with a suitable electrophile such as an alkyl halide to give an intermediate of the formula VII. A halogen group at the 4-position of the aryl ring of a compound of the formula VII can optionally be converted into, for example, a nitrile group to give a compound of the formula VIII by treatment with a suitable reagent, such as CuCN. Removal of the protective group can be accomplished, for example in the case of a tert-butyloxycarbonyl (BOC) group, treatment of a compound of the formula VIII under suitable cleaving conditions such as HCl in EtOH or TFA in CH2Cl2 to give a compound of the formula IX (G).
    Figure US20080108691A1-20080508-C00011
  • Scheme V describes a method of preparing certain compounds of formula I. Beginning with a 4-cyano 1-alkylaromatic compound of formula X, such as 4-cyano-1-methylnaphthalene (compound of formula X, where R4═H and R10 and R11 are taken together to form the remainder of a naphthyl ring), treatment with a strong base such as lithium diisopropyl amide, followed by quenching with a suitable electrophile, such as methyl chloroformate, affords a compound of the formula XI. A compound of the formula XI can be hydrolysed to the free acid by standard methods known to those skilled in the art, such as by treatment with LiOH in MeOH to provide a compound of the formula XII. A compound of the formula XII can then be used as an intermediate to prepare certain compounds of the formula I (where X═CH2) by the employment of, for example, standard peptide coupling conditions, such as EDAC and HOBT with triethylamine in a reaction with a suitable pyrrolidine or piperidinyl based coupling partner such as a compound of the formula III.
  • Use and Utility A. Utilities
  • The compounds of the present invention modulate the function of the nuclear hormone receptors, particularly the androgen receptor, and include compounds which are, for example, selective agonists, partial agonists, antagonists or partial antagonists of the androgen receptor (AR). Thus, the present compounds are useful in the treatment of AR-associated conditions An “AR-associated condition,” as used herein, denotes a condition or disorder which can be treated by modulating the function or activity of an AR in a subject, wherein treatment comprises prevention, partial alleviation or cure of the condition or disorder. Modulation may occur locally, for example, within certain tissues of the subject, or more extensively throughout a subject being treated for such a condition or disorder.
  • The compounds of the present invention can be administered to animals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to maintenance of muscle strength and function (e.g., in the elderly); reversal or prevention of frailty or age-related functional decline (“ARFD”) in the elderly (e.g., sarcopenia); treatment of catabolic side effects of glucocorticoids; prevention and/or treatment of reduced bone mass, density or growth (e.g., osteoporosis and osteopenia); treatment of chronic fatigue syndrome (CFS); chronic myalgia; treatment of acute fatigue syndrome and muscle loss following elective surgery (e.g., post-surgical rehabilitation); accelerating of wound healing; accelerating bone fracture repair (such as accelerating the recovery of hip fracture patients); accelerating healing of complicated fractures, e.g. distraction osteogenesis; in joint replacement; prevention of post-surgical adhesion formation; acceleration of tooth repair or growth; maintenance of sensory function (e.g., hearing, sight, olefaction and taste); treatment of periodontal disease; treatment of wasting secondary to fractures and wasting in connection with chronic obstructive pulmonary disease (COPD), chronic liver disease, AIDS, weightlessness, cancer cachexia, burn and trauma recovery, chronic catabolic state (e.g., coma), eating disorders (e.g., anorexia) and chemotherapy; treatment of cardiomyopathy; treatment of thrombocytopenia; treatment of growth retardation in connection with Crohn's disease; treatment of short bowel syndrome; treatment of irritable bowel syndrome; treatment of inflammatory bowel disease; treatment of Crohn's disease and ulcerative colits; treatment of complications associated with transplantation; treatment of physiological short stature including growth hormone deficient children and short stature associated with chronic illness; treatment of obesity and growth retardation associated with obesity; treatment of anorexia (e.g., associated with cachexia or aging); treatment of hypercortisolism and Cushing's syndrome; Paget's disease; treatment of osteoarthritis; induction of pulsatile growth hormone release; treatment of osteochondrodysplasias; treatment of depression, nervousness, irritability and stress; treatment of reduced mental energy and low self-esteem (e.g., motivation/assertiveness); improvement of cognitive function (e.g., the treatment of dementia, including Alzheimer's disease and short term memory loss); treatment of catabolism in connection with pulmonary dysfunction and ventilator dependency; treatment of cardiac dysfunction (e.g., associated with valvular disease, myocardial infarction, cardiac hypertrophy or congestive heart failure); lowering blood pressure; protection against ventricular dysfunction or prevention of reperfusion events; treatment of adults in chronic dialysis; reversal or slowing of the catabolic state of aging; attenuation or reversal of protein catabolic responses following trauma (e.g., reversal of the catabolic state associated with surgery, congestive heart failure, cardiac myopathy, burns, cancer, COPD etc.); reducing cachexia and protein loss due to chronic illness such as cancer or AIDS; treatment of hyperinsulinemia including nesidioblastosis; treatment of immunosuppressed patients; treatment of wasting in connection with multiple sclerosis or other neurodegenerative disorders; promotion of myelin repair; maintenance of skin thickness; treatment of metabolic homeostasis and renal homeostasis (e.g., in the frail elderly); stimulation of osteoblasts, bone remodeling and cartilage growth; regulation of food intake; treatment of insulin resistance, including NIDDM, in mammals (e.g., humans); treatment of insulin resistance in the heart; improvement of sleep quality and correction of the relative hyposomatotropism of senescence due to high increase in REM sleep and a decrease in REM latency; treatment of hypothermia; treatment of congestive heart failure; treatment of lipodystrophy (e.g., in patients taking HIV or AIDS therapies such as protease inhibitors); treatment of muscular atrophy (e.g., due to physical inactivity, bed rest or reduced weight-bearing conditions); treatment of musculoskeletal impairment (e.g., in the elderly); improvement of the overall pulmonary function; treatment of sleep disorders; and the treatment of the catabolic state of prolonged critical illness; treatment of hirsutism, acne, seborrhea, androgenic alopecia, anemia, hyperpilosity, benign prostate hypertrophy, adenomas and neoplasies of the prostate (e.g., advanced metastatic prostate cancer) and malignant tumor cells containing the androgen receptor, such as is the case for breast, brain, skin, ovarian, bladder, lymphatic, liver and kidney cancers; cancers of the skin, pancreas, endometrium, lung and colon; osteosarcoma; hypercalcemia of malignancy; metastatic bone disease; treatment of spermatogenesis, endometriosis and polycystic ovary syndrome; conteracting preeclampsia, eclampsia of pregnancy and preterm labor; treatment of premenstural syndrome; treatment of vaginal dryness; age related decreased testosterone levels in men, male menopause, hypogonadism, male hormone replacement, male and female sexual dysfunction (e.g., erectile dysfunction, decreased sex drive, sexual well-being, decreased libido), urinary incontinence, male and female contraception, hair loss, Reaven's Syndrome and the enhancement of bone and muscle performance/strength. The term treatment is also intended to include prophylactic treatment.
  • In addition, the conditions, diseases, and maladies collectively referenced to as “Syndrome X” or Metabolic Syndrome as detailed in Johannsson J. Clin. Endocrinol. Metab., 82, 727-34 (1997), may be treated employing the compounds of the invention.
  • B. Combinations
  • The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of formula I, alone or in combination with a pharmaceutical carrier or diluent. Optionally, compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antibiotic or other pharmaceutically active material.
  • The compounds of the present invention may be combined with growth promoting agents, such as, but not limited to, TRH, diethylstilbesterol, theophylline, enkephalins, E series prostaglandins, compounds disclosed in U.S. Pat. No. 3,239,345, e.g., zeranol, and compounds disclosed in U.S. Pat. No. 4,036,979, e.g., sulbenox or peptides disclosed in U.S. Pat. No. 4,411,890.
  • The compounds of the invention may also be used in combination with growth hormone secretagogues such as GHRP-6, GHRP-1 (as described in U.S. Pat. No. 4,411,890 and publications WO 89/07110 and WO 89/07111), GHRP-2 (as described in WO 93/04081), NN703 (Novo Nordisk), LY444711 (Lilly), MK-677 (Merck), CP424391 (Pfizer) and B-HT920, or with growth hormone releasing factor and its analogs or growth hormone and its analogs or somatomedins including IGF-1 and IGF-2, or with alpha-adrenergic agonists, such as clonidine or serotinin 5-HTD agonists, such as sumatriptan, or agents which inhibit somatostatin or its release, such as physostigmine and pyridostigmine. A still further use of the disclosed compounds of the invention is in combination with parathyroid hormone, PTH(1-34) or bisphosphonates, such as MK-217 (alendronate).
  • A still further use of the compounds of the invention is in combination with estrogen, testosterone, a selective estrogen receptor modulator, such as tamoxifen or raloxifene, or other androgen receptor modulators, such as those disclosed in Edwards, J. P. et. al., Bio. Med. Chem. Let., 9, 1003-1008 (1999) and Hamann, L. G. et. al., J. Med. Chem., 42, 210-212 (1999).
  • A further use of the compounds of this invention is in combination with progesterone receptor agonists (“PRA”), such as levonorgestrel, medroxyprogesterone acetate (MPA).
  • The compounds of the present invention may be employed alone or in combination with each other and/or other modulators of nuclear hormone receptors or other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-diabetic agents; anti-osteoporosis agents; anti-obesity agents; anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; anti-platelet agents; anti-thrombotic and thrombolytic agents; cardiac glycosides; cholesterol/lipid lowering agents; mineralocorticoid receptor antagonists; phosphodiesterase inhibitors; protein tyrosine kinase inhibitors; thyroid mimetics (including thyroid receptor agonists); anabolic agents; HIV or AIDS therapies; therapies useful in the treatment of Alzheimer's disease and other cognitive disorders; therapies useful in the treatment of sleeping disorders; anti-proliferative agents; and anti-tumor agents.
  • Examples of suitable anti-diabetic agents for use in combination with the compounds of the present invention include biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins (including insulin secretagogues or insulin sensitizers), meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide and glipizide), biguanide/glyburide combinations (e.g., Glucovance®), thiazolidinediones (e.g., troglitazone, rosiglitazone and pioglitazone), PPAR-alpha agonists, PPAR-gamma agonists, PPAR alpha/gamma dual agonists, SGLT2 inhibitors, glycogen phosphorylase inhibitors, inhibitors of fatty acid binding protein (aP2) such as those disclosed in U.S. Ser. No. 09/519,079 filed Mar. 6, 2000, glucagon-like peptide-1 (GLP-1), and dipeptidyl peptidase IV (DPP4) inhibitors such as those disclosed in WO 0168603.
  • Examples of suitable anti-osteoporosis agents for use in combination with the compounds of the present invention include alendronate, risedronate, PTH, PTH fragment, raloxifene, calcitonins, steroidal or non-steroidal progesterone receptor agonists, RANK ligand antagonists, calcium sensing receptor antagonists, TRAP inhibitors, selective estrogen receptor modulators (SERM's), estrogen and AP-1 inhibitors.
  • Examples of suitable anti-obesity agents for use in combination with the compounds of the present invention include aP2 inhibitors, such as those disclosed in U.S. Ser. No. 09/519,079 filed Mar. 6, 2000, PPAR gamma antagonists, PPAR delta agonists, beta 3 adrenergic agonists, such as AJ9677 (Takeda/Dainippon), L750355 (Merck), or CP331648 (Pfizer) or other known beta δ agonists as disclosed in U.S. Pat. Nos. 5,541,204, 5,770,615, 5,491,134, 5,776,983 and 5,488,064, a lipase inhibitor, such as orlistat or ATL-962 (Alizyme), a serotonin (and dopamine) reuptake inhibitor, such as sibutramine, topiramate (Johnson & Johnson) or axokine (Regeneron), a thyroid receptor beta drug, such as a thyroid receptor ligand as disclosed in WO 97/21993 (U. Cal SF), WO 99/00353 (KaroBio) and GB98/284425 (KaroBio), and/or an anorectic agent, such as dexamphetamine, phentermine, phenylpropanolamine or mazindol.
  • Examples of suitable anti-inflammatory agents for use in combination with the compounds of the present invention include prednisone, dexamethasone, Enbrel®, cyclooxygenase inhibitors (i.e., COX-1 and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, Naproxen®, Celebrex®, Vioxx®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CellCept®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-1, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR1384), prostaglandin synthesis inhibitors, budesonide, clofazimine, CNI-1493, CD4 antagonists (e.g., priliximab), p38 mitogen-activated protein kinase inhibitors, protein tyrosine kinase (PTK) inhibitors, IKK inhibitors, and therapies for the treatment of irritable bowel syndrome (e.g., Zelmac® and Maxi-K® openers such as those disclosed in U.S. Pat. No. 6,184,231 B1).
  • Examples of suitable anti-anxiety agents for use in combination with the compounds of the present invention include diazepam, lorazepam, buspirone, oxazepam, and hydroxyzine pamoate.
  • Examples of suitable anti-depressants for use in combination with the compounds of the present invention include citalopram, fluoxetine, nefazodone, sertraline, and paroxetine.
  • Examples of suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and T-type; e.g. diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril), AT-1 receptor antagonists (e.g., losartan, irbesartan, valsartan), ET receptor antagonists (e.g., sitaxsentan, atrsentan and compounds disclosed in U.S. Pat. Nos. 5,612,359 and 6,043,265), Dual ET/AII antagonist (e.g., compounds disclosed in WO 00/01389), neutral endopeptidase (NEP) inhibitors, vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., omapatrilat and gemopatrilat), and nitrates.
  • Examples of suitable anti-platelet agents for use in combination with the compounds of the present invention include GPIIb/IIIa blockers (e.g., abciximab, eptifibatide, tirofiban), P2Y12 antagonists (e.g., clopidogrel, ticlopidine, CS-747), thromboxane receptor antagonists (e.g., ifetroban), aspirin, and PDE-III inhibitors (e.g., dipyridamole) with or without aspirin.
  • Examples of suitable cardiac glycosides for use in combination with the compounds of the present invention include digitalis and ouabain.
  • Examples of suitable cholesterol/lipid lowering agents for use in combination with the compounds of the present invention include HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin) and ZD-4522 (a.k.a. rosuvastatin, or atavastatin or visastatin)), squalene synthetase inhibitors, fibrates, bile acid sequestrants, ACAT inhibitors, MTP inhibitors, lipooxygenase inhibitors, cholesterol absorption inhibitors, and cholesterol ester transfer protein inhibitors (e.g., CP-529414).
  • Examples of suitable mineralocorticoid receptor antagonists for use in combination with the compounds of the present invention include spironolactone and eplerinone.
  • Examples of suitable phosphodiesterase (PDE) inhibitors for use in combination with the compounds of the present invention include PDE-3 inhibitors such as cilostazol, and phosphodiesterase-5 inhibitors (PDE-5 inhibitors) such as sildenafil.
  • Examples of suitable thyroid mimetics for use in combination with the compounds of the present invention include thyrotropin, polythyroid, KB-130015, and dronedarone.
  • Examples of suitable anabolic agents for use in combination with the compounds of the present invention include testosterone, TRH diethylstilbesterol, estrogens, β-agonists, theophylline, anabolic steroids, dehydroepiandrosterone, enkephalins, E-series prostagladins, retinoic acid and compounds as disclosed in U.S. Pat. No. 3,239,345, e.g., Zeranol®; U.S. Pat. No. 4,036,979, e.g., Sulbenox® or peptides as disclosed in U.S. Pat. No. 4,411,890.
  • Examples of suitable HIV or AIDS therapies for use in combination with the compounds of the present invention include indinavir sulfate, saquinavir, saquinavir mesylate, ritonavir, lamivudine, zidovudine, lamivudine/zidovudine combinations, zalcitabine, didanosine, stavudine, and megestrol acetate.
  • Examples of suitable therapies for treatment of Alzheimer's disease and cognitive disorders for use in combination with the compounds of the present invention include donepezil, tacrine, revastigmine, 5HT6, gamma secretase inhibitors, beta secretase inhibitors, SK channel blockers, Maxi-K blockers, and KCNQs blockers.
  • Examples of suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, MLIB agonists, and GABA/NMDA receptor antagonists.
  • Examples of suitable anti-proliferative agents for use in combination with the compounds of the present invention include cyclosporin A, paclitaxel, FK-506, and adriamycin.
  • Examples of suitable anti-tumor agents for use in combination with the compounds of the present invention include paclitaxel, adriamycin, epothilones, cisplatin and carboplatin.
  • Compounds of the present invention may further be used in combination with nutritional supplements such as those described in U.S. Pat. No. 5,179,080, especially in combination with whey protein or casein, amino acids (such as leucine, branched amino acids and hydroxymethylbutyrate), triglycerides, vitamins (e.g., A, B6, B12, folate, C, D and E), minerals (e.g., selenium, magnesium, zinc, chromium, calcium and potassium), carnitine, lipoic acid, creatinine, B-hyroxy-B-methylbutyriate (Juven) and coenzyme Q-10.
  • In addition, compounds of the present invention may be used in combination with therapeutic agents used in the treatment of sexual dysfunction, including but not limited to PDE-5 inhibitors, such as sildenafil or IC-351.
  • Compounds of the present invention may further be used in combination with antiresorptive agents, hormone replacement therapies, vitamin D analogues, elemental calcium and calcium supplements, cathepsin K inhibitors, MMP inhibitors, vitronectin receptor antagonists, Src SH2 antagonists, vacular—H+-ATPase inhibitors, ipriflavone, fluoride, Tibolone, prostanoids, 17-beta hydroxysteroid dehydrogenase inhibitors and Src kinase inhibitors.
  • Compounds of the present invention may be used in combination with male contraceptives, such as nonoxynol 9 or therapeutic agents for the treatment of hair loss, such as minoxidil and finasteride or chemotherapeutic agents, such as with LHRH agonists.
  • Further, the compounds of the present invention may be used in combination with anti-cancer and cytotoxic agents, including but not limited to alkylating agents such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes such as L-asparaginase; farnesyl-protein transferase inhibitors; 5α-reductase inhibitors; inhibitors of 17β-hydroxysteroid dehydrogenase type 3; hormonal agents such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-disruptor agents, such as ecteinascidins or their analogs and derivatives; microtubule-stabilizing agents such as taxanes, for example, paclitaxel (Taxol®), docetaxel (Taxoter®), and their analogs, and epothilones, such as epothilones A-F and their analogs; plant-derived products, such as vinca alkaloids, epipodophyllotoxins, taxanes; and topiosomerase inhibitors; prenyl-protein transferase inhibitors; and miscellaneous agents such as hydroxyurea, procarbazine, mitotane, hexamethylmelamine, platinum coordination complexes such as cisplatin and carboplatin; and other agents used as anti-cancer and cytotoxic agents such as biological response modifiers, growth factors; immune modulators and monoclonal antibodies. The compounds of the invention may also be used in conjunction with radiation therapy.
  • Representative examples of these classes of anti-cancer and cytotoxic agents include but are not limited to mechlorethamine hydrochloride, cyclophosphamide, chlorambucil, melphalan, ifosfamide, busulfan, carmustin, lomustine, semustine, streptozocin, thiotepa, dacarbazine, methotrexate, thioguanine, mercaptopurine, fludarabine, pentastatin, cladribin, cytarabine, fluorouracil, doxorubicin hydrochloride, daunorubicin, idarubicin, bleomycin sulfate, mitomycin C, actinomycin D, safracins, saframycins, quinocarcins, discodermolides, vincristine, vinblastine, vinorelbine tartrate, etoposide, etoposide phosphate, teniposide, paclitaxel, tamoxifen, estramustine, estramustine phosphate sodium, flutamide, buserelin, leuprolide, pteridines, diyneses, levamisole, aflacon, interferon, interleukins, aldesleukin, filgrastim, sargramostim, rituximab, BCG, tretinoin, irinotecan hydrochloride, betamethosone, gemcitabine hydrochloride, altretamine, and topoteca and any analogs or derivatives thereof.
  • Preferred member of these classes include, but are not limited to, paclitaxel, cisplatin, carboplatin, doxorubicin, caminomycin, daunorubicin, aminopterin, methotrexate, methopterin, mitomycin C, ecteinascidin 743, or porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podophyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine and leurosine.
  • Examples of anticancer and other cytotoxic agents include the following: epothilone derivatives as found in German Patent No. 4138042.8; WO 97/19086, WO 98/22461, WO 98/25929, WO 98/38192, WO 99/01124, WO 99/02224, WO 99/02514, WO 99/03848, WO 99/07692, WO 99/27890, WO 99/28324, WO 99/43653, WO 99/54330, WO 99/54318, WO 99/54319, WO 99/65913, WO 99/67252, WO 99/67253 and WO 00/00485; cyclin dependent kinase inhibitors as found in WO 99/24416 (see also U.S. Pat. No. 6,040,321); and prenyl-protein transferase inhibitors as found in WO 97/30992 and WO 98/54966; and agents such as those described generically and specifically in U.S. Pat. No. 6,011,029 (the compounds of which U.S. patent can be employed together with any NHR modulators (including, but not limited to, those of present invention) such as AR modulators, ER modulators, with LHRH modulators, or with surgical castration, especially in the treatment of cancer).
  • The above other therapeutic agents, when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • The compounds of the formula I can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds can also be administered liposomally.
  • Exemplary compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The compounds of formula I can also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • Exemplary compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • Exemplary compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug.
  • Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • The effective amount of a compound of the present invention can be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for an adult human of from about 0.01 to 2000 mg of active compound per day, which can be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats and the like, subject to NHR-associated conditions.
  • Transactivation Assays A. AR Specific Assay
  • Compounds of the present invention were tested in transactivation assays of a transfected reporter construct and using the endogenous androgen receptor of the host cells. The transactivation assay provides a method for identifying functional agonists and partial agonists that mimic, or antagonists that inhibit, the effect of native hormones, in this case, dihydrotestosterone (DHT). This assay can be used to predict in vivo activity as there is a good correlation in both series of data. See, e.g. T. Berger et al., J. Steroid Biochem. Molec. Biol. 773 (1992), the disclosure of which is herein incorporated by reference.
  • For the transactivation assay a reporter plasmid is introduced by transfection (a procedure to induce cells to take foreign genes) into the respective cells. This reporter plasmid, comprising the cDNA for a reporter protein, such as secreted alkaline phosphatase (SEAP), controlled by prostate specific antigen (PSA) upstream sequences containing androgen response elements (AREs). This reporter plasmid functions as a reporter for the transcription-modulating activity of the AR. Thus, the reporter acts as a surrogate for the products (mRNA then protein) normally expressed by a gene under control of the AR and its native hormone. In order to detect antagonists, the transactivation assay is carried out in the presence of constant concentration of the natural AR hormone (DHT) known to induce a defined reporter signal. Increasing concentrations of a suspected antagonist will decrease the reporter signal (e.g., SEAP production). On the other hand, exposing the transfected cells to increasing concentrations of a suspected agonist will increase the production of the reporter signal.
  • For this assay, LNCaP and MDA 453 cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in RPMI 1640 or DMEM medium supplemented with 10% fetal bovine serum (FBS; Gibco) respectively. The respective cells were transiently transfected by electroporation according to the optimized procedure described by Heiser, 130 Methods Mol. Biol., 117 (2000), with the pSEAP2/PSA540/Enhancer reporter plasmid. The reporter plasmid, was constructed as follows: commercial human placental genomic DNA was used to generate by Polymerase Cycle Reaction (PCR) a fragment containing the BglII site (position 5284) and the Hind III site at position 5831 of the human prostate specific antigen promoter (Accession # U37672), Schuur, et al., J. Biol. Chem., 271 (12): 7043-51 (1996). This fragment was subcloned into the pSEAP2/basic (Clontech) previously digested with BglII and HindIII to generate the pSEAP2/PSA540 construct. Then a fragment bearing the fragment of human PSA upstream sequence between positions-5322 and -3873 was amplified by PCR from human placental genomic DNA. A XhoI and a BglII sites were introduced with the primers. The resulting fragment was subcloned into pSEAP2/PSA540 digested with XhoI and BglII respectively, to generate the pSEAP2/PSA540/Enhancer construct. LNCaP and MDA MB-453 cells were collected in media containing 10% charcoal stripped FBS. Each cell suspension was distributed into two Gene Pulser Cuvetts (Bio-Rad) which then received 8 μg of the reporter construct, and electoporated using a Bio-Rad Gene Pulser at 210 volts and 960 μFaraday. Following the transfections the cells were washed and incubated with media containing charcoal stripped fetal bovine serum in the absence (blank) or presence (control) of 1 nM dihydrotestosterone (DHT; Sigma Chemical) and in the presence or absence of the standard anti-androgen bicalutamide or compounds of the present invention in concentrations ranging from 10−10 to 10−5 M (sample). Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory workstation.
  • After 48 h, a fraction of the supernatant was assayed for SEAP activity using the Phospha-Light Chemiluminescent Reporter Gene Assay System (Tropix, Inc). Viability of the remaining cells was determined using the CellTiter 96 Aqueous Non-Radioactive Cell Proliferation Assay (MTS Assay, Promega). Briefly, a mix of a tetrazolium compound (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; MTS) and an electron coupling reagent (phenazine methosulfate; PMS) are added to the cells. MTS (Owen's reagent) is bioreduced by cells into a formazan that is soluble in tissue culture medium, and therefore its absorbance at 490 nm can be measured directly from 96 well assay plates without additional processing. The quantity of formazan product as measured by the amount of 490 μm absorbance is directly proportional to the number of living cells in culture. For each replicate the SEAP reading was normalized by the Abs490 value derived from the MTS assay. For the antagonist mode, the % Inhibition was calculated as:
    % Inhibition=100×(1−[average control−average blank/average sample−average blank])
    Data was plotted and the concentration of compound that inhibited 50% of the normalized SEAP was quantified (IC50).
  • For the agonist mode % Control was referred as the effect of the tested compound compared to the maximal effect observed with the natural hormone, in this case DHT, and was calculated as:
    % Control=100×average sample−average blank/average control−average blank
    Data was plotted and the concentration of compound that activates to levels 50% of the normalized SEAP for the control was quantified (EC50).
  • B. GR Specificity Assay
  • The reporter plasmid utilized was comprised of the cDNA for the reporter SEAP protein, as described for the AR specific transactivation assay. Expression of the reporter SEAP protein was controlled by the mouse mammary tumor virus long terminal repeat (MMTV LTR) sequences that contains three hormone response elements (HREs) that can be regulated by both GR and PR see, e.g. G. Chalepakis et al., Cell, 53(3), 371 (1988). This plasmid was transfected into A549 cells, which expresses endogenous GR, to obtain a GR specific transactivation assay. A549 cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in RPMI 1640 supplemented with 10% fetal bovine serum (FBS; Gibco). Determination of the GR specific antagonist activity of the compounds of the present invention was identical to that described for the AR specific transactivation assay, except that the DHT was replaced with 5 nM dexamethasone (Sigma Chemicals), a specific agonist for GR. Determination of the GR specific agonist activity of the compounds of the present invention was performed as described for the AR transactivation assay, wherein one measures the activation of the GR specific reporter system by the addition of a test compound, in the absence of a known GR specific agonists ligand.
  • C. PR Specific Assay
  • The reporter plasmid utilized was comprised of the cDNA for the reporter SEAP protein, as described for the AR specific transactivation assay. Expression of the reporter SEAP protein was controlled by the mouse mammary tumor virus long terminal repeat (MMTV LTR) sequences that contains three hormone response elements (HRE's) that can be regulated by both GR and PR. This plasmid was transfected into T47D, which expresses endogenous PR, to obtain a PR specific transactivation assay. T47D cells were obtained from the American Type Culture Collection (Rockville, Md.), and maintained in DMEM medium supplemented with 10% fetal bovine serum (FBS; Gibco). Determination of the PR specific antagonist activity of the compounds of the present invention was identical to that described for the AR specific transactivation assay, except that the DHT was replaced with 1 nM Promegastone (NEN), a specific agonist for PR. Determination of the PR specific agonist activity of the compounds of the present invention was performed as described for the AR transactivation assay, wherein one measures the activation of the PR specific reporter system by the addition of a test compound, in the absence of a known PR specific agonists ligand.
  • D. AR Binding Assay
  • For the whole-cell binding assay, human LNCaP cells (T877A mutant AR) or MDA 453 (wild type AR) in 96-well microtiter plates containing RPMI 1640 or DMEM supplemented with 10% charcoal stripped CA-FBS (Cocaleco Biologicals) respectively, were incubated at 37° C. to remove any endogenous ligand that might be complexed with the receptor in the cells. After 48 h, either a saturation analysis to determine the Kd for tritiated dihydrotestosterone, [3H]-DHT, or a competitive binding assay to evaluate the ability of test compounds to compete with [3H]-DHT were performed. For the saturation analysis, media (RPMI 1640 or DMEM—0.2% CA-FBS) containing [3H]-DHT (in concentrations ranging from 0.1 nM to 16 nM) in the absence (total binding) or presence (non-specific binding) of a 500-fold molar excess of unlabeled DHT were added to the cells. After 4 h at 37° C., an aliquot of the total binding media at each concentration of [3H]-DHT was removed to estimate the amount of free [3H]-DHT. The remaining media was removed, cells were washed three times with PBS and harvested onto UniFilter GF/B plates (Packard), Microscint (Packard) was added and plates counted in a Top-Counter (Packard) to evaluate the amount of bound [3H]-DHT.
  • For the saturation analysis, the difference between the total binding and the non-specific binding, was defined as specific binding. The specific binding was evaluated by Scatchard analysis to determine the Kd for [3H]-DHT. See e.g. D.
  • Rodbard, Mathematics and statistics of ligand assays: an illustrated guide: In: J. Langon and J. J. Clapp, eds., Ligand Assay, Masson Publishing U.S.A., Inc., New York, pp. 45-99, (1981), the disclosure of which is herein incorporated by reference.
  • For the competition studies, media containing 1 nM [3H]-DHT and compounds of the invention (“test compounds”) in concentrations ranging from 10−10 to 10−5 M were added to the cells. Two replicates were used for each sample. After 4 h at 37° C., cells were washed, harvested and counted as described above. The data was plotted as the amount of [3H]-DHT (% of control in the absence of test compound) remaining over the range of the dose response curve for a given compound. The concentration of test compound that inhibited 50% of the amount of [3H]-DHT bound in the absence of competing ligand was quantified (IC50) after log-logit transformation. The K1 values were determined by application of the Cheng-Prusoff equation to the IC50 values, where:
    K1=IC50
    (1+(3H-DHT)/Kd for 3H-DHT)
    After correcting for non-specific binding, IC50 values were determined. The IC50 is defined as the concentration of competing ligand needed to reduce specific binding by 50%. The Kds for [3H]-DHT for MDA 453 and LNCaP were 0.7 and 0.2 nM respectively.
  • E. C2C12 Mouse Myoblast Transactivation Assay
  • Two functional transactivation assays were developed to assess the efficacy of androgen agonists in a muscle cell background using a luciferase reporter. The first assay (ARTA Stable 1) uses a cell line, Stable 1 (clone #72), which expresses the full length rat androgen receptor but requires the transient transfection of an enhancer/reporter. This cell line was derived from C2C12 mouse moyoblast cells. The second assay (ARTA Stable 2) uses a cell line, Stable 2 (clone #133), derived from Stable 1 which expresses both rAR and the enhancer/luciferase reporter.
  • The enhancer/reporter construct used in this system is pGL3/2XDR-1/luciferase. 2×DR-1 was reported to be an AR specific response element in CV-1 cells, Brown et. al. The Journal of Biological Chemistry 272, 8227-8235, (1997). It was developed by random mutagenesis of an AR/GR consensus enhancer sequence.
  • F. ARTA Stable 1
  • 1. Stable 1 cells are plated in 96 well format at 6,000 cells/well in high glucose DMEM without phenol red (Gibco BRL, Cat. No.: 21063-029) containing 10% charcoal and dextran treated FBS (HyClone Cat. No.: SH30068.02), 50 mM HEPES Buffer (Gibco BRL, Cat. No.: 15630-080), 1×MEM Na Pyruvate (Gibco BRL, Cat. No.: 11360-070), 0.5×Antibiotic-Antimycotic, and 800 μg/mL Geneticin (Gibco BRL, Cat. No.: 10131-035).
  • 2. 48 h later, cells are transfected with pGL3/2XDR-1/luciferase using LipofectAMINE Plus™ Reagent (Gibco BRL, Cat. No.: 10964-013). Specifically, 5 ng/well pGL3/2XDR-1/luciferase DNA and 50 ng/well Salmon Sperm DNA (as carrier) are diluted with 5 μl/well Opti-MEMem media (Gibco BRL, Cat. No.: 31985-070). To this, 0.5 μl/well Plus reagent is added. This mixture is incubated for 15 min at rt. In a separate vessel, 0.385 μl/well LipofectAMINE reagent is diluted with 5 μl/well Opti-MEM. The DNA mixture is then combined with the LipofectAMINE mixture and incubated for an additional 15 min at rt. During this time, the media from the cells is removed and replaced with 60 μl/well of Opti-MEM. To this is added 10 μl/well of the DNA/LipofectAMINE transfection mixture. The cells are incubated for 4 h.
  • 3. The transfection mixture is removed from the cells and replaced with 90 μl of media as in #1 above.
  • 4. 10 μl/well of appropriate drug dilution is placed in each well.
  • 5. 24 h later, the Steady-Glo™ Luciferase Assay System is used to detect activity according to the manufacturer's instructions (Promega, Cat. No.: E2520).
  • G. ARTA Stable 2
  • 1. Stable 2 cells are plated in 96 well format at 6,000 cells/well in high glucose DMEM without phenol red (Gibco BRL, Cat. No.: 21063-029) containing 10% charcoal and dextran treated FBS (HyClone Cat. No.: SH30068.02), 50 mM HEPES Buffer (Gibco BRL, Cat. No.: 15630-080), 1×MEM Na Pyruvate (Gibco BRL, Cat. No.: 11360-070), 0.5×Antibiotic-Antimycotic, 800 μg/mL Geneticin (Gibco BRL, Cat. No.: 10131-035) and 800 μg/mL Hygromycin β (Gibco BRL, Cat. No.: 10687-010).
  • 2. 48 h later, the media on the cells is removed and replaced with 90 μl fresh. 10 μl/well of appropriate drug dilution is placed in each well.
  • 3. 24 h later, the Steady-Glo™ Luciferase Assay System is used to detect activity according to the manufacturer's instructions (Promega, Cat. No. E2520).
  • Proliferation Assays A. Human Prostate Cell Proliferation Assay
  • Compounds of the present invention were tested (“test compounds”) on the proliferation of human prostate cancer cell lines. For that, MDA PCa2b cells, a cell line derived from the metastasis of a patient that failed castration, Navone et al., Clin. Cancer Res., 3, 2493-500 (1997), were incubated with or without the test compounds for 72 h and the amount of [3H]-thymidine incorporated into DNA was quantified as a way to assess number of cells and therefore proliferation. The MDA PCa2b cell line was maintained in BRFF-HPC1 media (Biological Research Faculty & Facility Inc., MD) supplemented with 10% FBS. For the assay, cells were plated in Biocoated 96-well microplates and incubated at 37° C. in 10% FBS (charcoal-stripped)/BRFF-BMZERO (without androgens). After 24 h, the cells were treated in the absence (blank) or presence of 1 nM DHT (control) or with test compounds (sample) of the present invention in concentrations ranging from 10−10 to 10−5 M. Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory work station. Seventy-two h later 0.44 uCi. of [3H]-Thymidine (Amersham) was added per well and incubated for another 24 h followed by tripsinization, harvesting of the cells onto GF/B filters. Micro-scint PS were added to the filters before counting them on a Beckman TopCount.
  • The % Inhibition was calculated as:
    % Inhibition=100×(1−[averagecontrol−averageblank/averagesample−averageblank])
    Data was plotted and the concentration of compound that inhibited 50% of the [3H]_Thymidine incorporation was quantified (IC50).
  • B. Murine Breast Cell Proliferation Assay
  • The ability of compounds of the present invention (“test compounds”) to modulate the function of the AR was determined by testing said compounds in a proliferation assay using the androgen responsive murine breast cell line derived from the Shionogi tumor, Hiraoka et al., Cancer Res., 47, 6560-6564 (1987). Stable AR dependent clones of the parental Shionogi line were established by passing tumor fragments under the general procedures originally described in Tetuo, et. al., Cancer Research 25, 1168-1175 (1965). From the above procedure, one stable line, SC114, was isolated, characterized and utilized for the testing of example compounds. SC114 cells were incubated with or without the test compounds for 72 h and the amount of [3H]-thymidine incorporated into DNA was quantified as a surrogate endpoint to assess the number of cells and therefore the proliferation rate as described in Suzuki et. al., J. Steroid Biochem. Mol. Biol. 37, 559-567 (1990). The SC114 cell line was maintained in MEM containing 10−8 M testosterone and 2% DCC-treated FCS. For the assay, cells were plated in 96-well microplates in the maintenance media and incubated at 37° C. On the following day, the medium was changed to serum free medium [Ham's F-12:MEM (1; 1, v/v) containing 0.1% BSA] with (antagonist mode) or without (agonist mode) 10−8 M testosterone and the test compounds of the present invention in concentrations ranging from 10−10 to 10−5 M. Duplicates were used for each sample. The compound dilutions were performed on a Biomek 2000 laboratory work station. Seventy two h later 0.44uCi of [3H]-Thymidine (Amersham) was added per well and incubated for another 2 h followed by tripsinization, and harvesting of the cells onto GF/B filters. Micro-scint PS were added to the filters before counting them on a Beckman TopCount.
  • For the antagonist mode, the % Inhibition was calculated as:
    % Inhibition=100×(1−[averagesample−averageblank/averagecontrol−averageblank])
    Data was plotted and the concentration of compound that inhibited 50% of the [3H]-Thymidine incorporation was quantified (IC50).
  • For the agonist mode % Control was referred as the effect of the tested compound compared to the maximal effect observed with the natural hormone, in this case DHT, and was calculated as:
    % Control=100×(averagesample−averageblank)/(averagecontrol−average blank)
    Data was plotted and the concentration of compound that inhibited 50% of the [3H]-Thymidine incorporation was quantified (EC50).
  • C. In Vitro Assay to Measure GR-Induced AP-1 Transrepression
  • The AP-1 assay is a cell-based luciferase reporter assay. A549 cells, which contain endogenous glucocorticoid receptor, were stably transfected with an AP-1 DNA binding site attached to the luciferase gene. Cells are then grown in RPMI+10% fetal calf serum (charcoal-treated)+Penicillin/Streptomycin with 0.5 mg/mL geneticin. Cells are plated the day before the assay at approximately 40000 cells/well. On assay day, the media is removed by aspiration and 20 μL assay buffer (RPMI without phenol red+10% FCS (charcoal-treated)+Pen/Strep) is added to each well. At this point either 20 μL assay buffer (control experiments), the compounds of the present invention (“test compounds”) (dissolved in DMSO and added at varying concentrations) or dexamethasome (100 nM in DMSO, positive control) are added to each well. The plates are then pre-incubated for 15 min at 37° C., followed by stimulation of the cells with 10 ng/mL PMA. The plates are then incubated for 7 h at 37° C. after which 40 μL luciferase substrate reagent is added to each well. Activity is measured by analysis in a luminometer as compared to control experiments treated with buffer or dexamethasome. Activity is designated as % inhibition of the reporter system as compared to the buffer control with 10 ng/mL PMA alone. The control, dexamethasone, at a concentration of ≧10 μM typically suppresses activity by 65%. Test compounds which demonstrate an inhibition of PMA induction of 50% or greater at a concentration of test compound of ≧10 μM are deemed active.
  • In Vivo Assays Levator Ani & Wet Prostate Weight Assay AR Agonist Assay
  • The activity of compounds of the present invention as AR agonists was investigated in an immature male rat model, a recognized test of anabolic effects in muscle and sustaining effects in sex organs for a given compound, as described in L. G. Hershberger et al., Proc. Soc. Expt. Biol. Med., 83, 175 (1953); B. L. Beyler et al, “Methods for evaluating anabolic and catabolic agents in laboratory animals”, J. Amer. Med. Women's Ass., 23, 708 (1968); H. Fukuda et al., “Investigations of the levator ani muscle as an anabolic steroid assay”, Nago Dqi. Yak. Ken. Nem. 14, 84 (1966) the disclosures of which are herein incorporated by reference.
  • The basis of this assay lies in the well-defined action of androgenic agents on the maintenance and growth of muscle tissues and sexual accessory organs in animals and man. Androgenic steroids, such as testosterone (T), have been well characterized for their ability to maintain muscle mass. Treatment of animals or humans after castrations with an exogenous source of T results in a reversal of muscular atrophy. The effects of T on muscular atrophy in the rat levator ani muscle have been well characterized. M. Masuoka et al., “Constant cell population in normal, testosterone deprived and testosterone stimulated levator ani muscles” Am. J. Anat. 119, 263 (1966); Z. Gori et al., “Testosterone hypertrophy of levator ani muscle of castrated rats. I. Quantitative data” Boll.—Soc. Ital. Biol. Sper. 42, 1596 (1966); Z. Gori et al., “Testosterone hypertrophy of levator ani muscle of castrated rats. II. Electron-microscopic observations” Boll.—Soc. Ital. Biol. Sper. 42, 1600 (1966); A. Boris et al., Steroids 15, 61 (1970). As described above, the effects of androgens on maintenance of male sexual accessory organs, such as the prostate and seminal vesicles, is well described. Castration results in rapid involution and atrophy of the prostate and seminal vesicles. This effect can be reversed by exogenous addition of androgens. Since both the levator ani muscle and the male sex organs are the tissues most responsive to the effects of androgenic agents, this model is used to determine the androgen dependent reversal of atrophy in the levator ani muscle and the sex accessory organs in immature castrated rats. Sexually mature rats (200-250 g, 6-8 weeks-old, Sprague-Dawley, Harlan) were acquired castrated from the vendor (Taconic). The rats were divided into groups and treated daily for 7 to 14 days with one of the following:
  • 1. Control vehicle
  • 2. Testosterone Propionate (TP) (3 mg/rat/day, subcutaneous)
  • 3. TP plus Bicalutamide (administered p.o. in PEGTW, QD), a recognized antiandrogen, as a reference compound.
  • 4. To demonstrate antagonist activity, a compound of the present invention (“test compound”) was administered (p.o. in PEGTW, QD) with TP (s.c. as administered in group 2) in a range of doses.
  • 5. To demonstrate agonist activity a compound of the present invention (“test compound”) was administered alone (p.o. in PEGTW, QD) in a range of doses.
  • At the end of the 7-14-day treatment, the animals were sacrificed by carbon dioxide, and the levator ani, seminal vesicle and ventral prostate weighed. To compare data from different experiments, the levator ani muscle and sexual organ weights were first standardized as mg per 100 g of body weight, and the increase in organ weight induced by TP was considered as the maximum increase (100%). Super-anova (one factor) was used for statistical analysis.
  • The gain and loss of sexual organ weight reflect the changes of the cell number (DNA content) and cell mass (protein content), depending upon the serum androgen concentration. See Y. Okuda et al., J. Urol., 145, 188-191 (1991), the disclosure of which is herein incorporated by reference. Therefore, measurement of organ wet weight is sufficient to indicate the bioactivity of androgens and androgen antagonist. In immature castrated rats, replacement of exogenous androgens increases levator ani, seminal vesicles (SV) and prostate in a dose dependent manner.
  • The maximum increase in organ weight was 4 to 5-fold when dosing 3 mg/rat/day of testosterone (T) or 1 mg/rat/day of testosterone propionate (TP) for 3 days. The EC50 of T and TP were about 1 mg and 0.03 mg, respectively. The increase in the weight of the VP and SV also correlated with the increase in the serum T and DHT concentration. Although administration of T showed 5-times higher serum concentrations of T and DHT at 2 h after subcutaneous injection than that of TP, thereafter, these high levels declined very rapidly. In contrast, the serum concentrations of T and DHT in TP-treated animals were fairly consistent during the 24 h, and therefore, TP showed about 10-30-fold higher potency than free T.
  • EXAMPLES
  • The following examples serve to better illustrate, but not limit, some of the preferred embodiments of the invention.
  • Example 1 1-(4-Cyano-2-ethyl-3-(trifluoromethyl)phenyl-1-carbamoyl)-3-hydroxy-pyrrolidine-2-carboxylic acid methyl ester
  • Figure US20080108691A1-20080508-C00012
  • Step 1. N-(4-Chloro-3-trifluoromethylphenyl)-2,2-dimethylpropionamide
  • Figure US20080108691A1-20080508-C00013
  • To a solution of 15.0 g (76.7 mmol) of 4-chloro-3-(trifluoromethyl)aniline in 200 mL of anhydrous THF cooled to 0-5° C. was added 11.7 mL (84.4 mmol) of NEt3 followed by 10.4 mL (84.4 mmol) of pivaloyl chloride over 30 min. The ice bath was removed, and the mixture was stirred for 1 h, then diluted with ether and filtered. The filtrate was washed with water (2×) and brine, dried (MgSO4), filtered and concentrated. The residue was triturated with hexanes and the solid filtered and dried to give 20.4 g (95%) of the Step 1 compound. 1H NMR (DMSO-d6) δ 1.22 (s, 9H), 7.63 (d, J=8.8, 1H), 8.00 (dd, J=2.8, 8.8, 1H), 8.24 (d, J=2.2, 1H), 9.62 (s, 1H); 13C NMR (DMSO-d6)δ26.94, 118.6, 121.7, 123.6, 124.6, 126.4, 126.6, 131.7, 138.9, 177.1; HPLC-1: 3.62 min retention time, column: Phenominex ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; HPLC-2: 3.53 min retention time (99%), column: Shimadzu Shim-Pack VP-ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UVdetection at 220 nm; MS: 280 [M+H]+.
  • Step 2. N-(4-Chloro-2-ethyl-3-trifluoromethylphenyl)-2,2-dimethylpropionamide
  • Figure US20080108691A1-20080508-C00014
  • To a solution of 3.00 g (10.7 mmol) of the Step 1 compound in 30 mL of anhydrous THF cooled to 0-5° C. was added 16.1 mL (25.7 mmol) of 1.6 M n-BuLi in hexanes over 30 min, keeping the temperature below 5° C. The solution was stirred at 0-5° C. for 1.5 h. A solution of 0.94 mL (11.8 mmol) of iodoethane in 2.5 mL of petroleum ether was added over 20 min, keeping the temperature below 5° C. The suspension was then stirred at 0-5° C. for 1 h and diluted with water and ether. The aqueous layer was extracted with ether and the combined organic layers were washed with brine, dried (MgSO4), filtered and concentrated. The residue was purified on silica gel eluted using CH2Cl2 to give 2.07 g (63%) of the Step 2 compound. 1H NMR (CDCl3) δ 1.22 (t, 3H), 1.35 (s, 9H), 2.79 (m, 2H), 7.37 (br d, J=8.8, 2H), 7.93 (d, J=8.8, 1H); 13C NMR (CDCl3) δ 14.2, 22.2, 27.5, 39.8, 125.0, 127.0, 128.3, 129.0, 130.3, 135.4, 136.8, 176.8; HPLC-1: 3.54 min retention time, column: Phenominex ODS C18 4.6×50 mm, 4 min gradient 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm, HPLC-2: 3.47 min retention time (96%), column: Shimadzu Shim-Pack VP-ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; MS 308 [M+H]+.
  • Step 3. N-(4-Cyano-2-ethyl-3-trifluoromethylphenyl)-2,2-dimethylpropionamide
  • Figure US20080108691A1-20080508-C00015
  • A suspension of 2.00 g (6.50 mmol) of the Step 2 compound and 1.46 g (16.2 mmol) of CuCN in 20 mL of anhydrous N-methylpyrrolidinone was refluxed for 18 h. The suspension was cooled to rt and poured into ice water with stirring. The solid was filtered, washed with water, dried, triturated with MeOH and filtered. The filtrate was concentrated to give 2.00 g of the Step 3 compound. 1H NMR (DMSO-d6) δ 1.01 (t, 3H), 1.24 (s, 9H), 2.79 (m, 2H), 7.71 (d, J=8.7, 2H), 7.95 (d, J=8.3, 1H), 9.30 (s, 1H); 13C NMR (DMSO-d6) δ 13.8, 21.0, 26.9, 107.0, 116.0, 132.6, 133.7, 141.0, 143.0, 177.0; HPLC-1: 3.25 min retention time, column: Phenominex ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; HPLC-2: 3.20 min retention time (85%), column: Shimadzu Shim-Pack VP-ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; MS: 299 [M+H]+.
  • Step 4. 4-Amino-3-ethyl-2-trifluoromethylbenzonitrile
  • Figure US20080108691A1-20080508-C00016
  • A solution of 1.94 g (6.50 mmol) of the Step 3 compound in 30 mL of concentrated HCl/EtOH (1:1) was refluxed for 17 h. The solution was concentrated and the residue suspended in EtOAc, washed with saturated NaHCO3 solution (2×) and brine, dried over MgSO4, filtered and concentrated. The residue was purified on silica gel eluting using CHCl3/MeOH (98:2) to give 1.07 g (77%) of the Step 4 compound. 1H NMR (CDCl3) δ 1.20 (t, 3H), 2.70 (m, 2H), 4.44 (br s, 2H), 6.83 (d, J=8.8, 1H), 7.44 (d, J=8.8, 1H); 13C NMR (CDCl3) δ 12.15, 21.02, 98.22, 117.30, 118.07, 122.3, 124.5, 127.43, 131.5, 134.1, 149.4; HPLC-1: 2.91 min retention time, column: Phenominex ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; HPLC-2: 2.81 min retention time (99%), column: Shimadzu Shim-Pack VP-ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; MS: 215 [M+H]+.
  • Step 5. 1-(4-Cyano-2-ethyl-3-trifluoromethylphenylcarbamoyl)-3-hydroxypyrrolidine-2-carboxylic acid
  • Figure US20080108691A1-20080508-C00017
  • To a suspension of 0.495 g (2.32 mmol) of the Step 4 compound, or an equivalent molar amount of the hydrochloride salt thereof, in 20 mL of CH2Cl2 was added 1.94 g (23.2 mmol) of NaHCO3 followed by 9.80 mL (18.5 mmol) of phosgene (20% solution in toluene), and the suspension was stirred at rt for 1 h. Another 2.5 mL (4.72 mmol) of 20% phosgene in toluene solution was added, and the suspension stirred for 1.5 h. The suspension was then filtered and the filtrate concentrated. The residue was azeotroped with toluene three times and concentrated to dryness. The resulting solid was then added to a suspension of 0.48 mL (2.78 mmol) of diisopropylethylamine, 0.60 g (2.32 mmol) of cis-3-hydroxyproline and 0.30 g of 4 Å molecular sieves in 25 mL of CH2Cl2, and the suspension was stirred at rt for 13.5 h. To the suspension was added 0.42 mL (2.78 mmol) of DBU and the suspension stirred at rt for 3 days. The suspension was filtered and the filtrate concentrated. The residue was purified on a reverse phase prep C18 column eluted using water/methanol (0-100% gradient) to give 83 mg (10%) of the compound of Example 1. 1H NMR (CD3OD) δ 1.17 (t, 3H), 2.05 (m, 1H), 2.18 (m, 1H), 2.92 (m, 2H), 3.75 (m, 2H), 4.42 (s, 1H), 2.52 (br s, 1H), 7.76 (d, J=8.4, 1H), 7.86 (d, J=8.4, 1H); 13C NMR (CD3OD) δ 14.3, 22.6, 33.6, 45.7, 69.6, 74.9, 107.5, 117.8, 123.45, 126.2, 131.0, 131.3, 131.6, 131.9, 132.2, 140.9, 144.5, 156.8, 173.9; HPLC-1: 2.50 min retention time, column: Phenominex ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; HPLC-2: 2.38 min retention time (96%), column: Shimadzu Shim-Pack VP-ODS C18 4.6×50 mm, 4 min gradient, 10% MeOH/90% H2O/0.1% TFA to 90% MeOH/10% H2O/0.1% TFA; 1 min hold; 4 mL/min, UV detection at 220 nm; MS: 372 [M+H]+.
  • Example 2 1-(4-Cyanonaphthalen-1-ylcarbamoyl)-3-hydroxy-pyrrolidine-2-carboxylic acid methyl ester
  • Figure US20080108691A1-20080508-C00018
  • Step 1. 4-Isocyanato-naphthalene-1-carbonitrile
  • Figure US20080108691A1-20080508-C00019
  • The title compound was prepared from 4-cyanonaphthylamine in a similar fashion to that described in Step 5 of Example 1 for the preparation of an aryl isocyanate from an aryl amine.
  • Step 2. 1-(4-Cyanonaphthalen-1-ylcarbamoyl)-3-hydroxypyrrolidine-2-carboxylic acid methyl ester
  • To a suspension of the Step 1 compound (0.51 g, 1.96 mmol) in CH2Cl2 (2 mL) cooled to 0° C. was added i-Pr2EtN (0.37 mL, 2.16 mmol). After stirring at 0° C. for 20 min, cis-3-hydroxyproline methyl ester (0.34 g, 1.75 mmol) in CH2Cl2 (1 mL) solution was added, along with 4 Å molecular sieves (0.5 g) and the resulting mixture stirred at rt until urea formation was complete (˜1 h). The reaction mixture was then loaded on a silica gel column, eluted with 50% EtOAc/hexane, and 5% MeOH in EtOAc/hexane (1:1) to afford 436 mg of the title compound as a white foam. HPLC: 99% at 1.65 min (retention time) (Conditions: Phenom. Luna C18 (4.6×50 mm); Eluted with 0% to 100% B; 4 min gradient (A=90% H2O−10% CH3CN−0.1% TFA and B=10% H2O−90% CH3CN−0.1% TFA), Flow rate at 4 mL/min., UV detection at 220 nm). Chiral HPLC: retention time=8.78 min (99%); Conditions: (CHIRALPAK® OD column 4.6×250 mm; 25% isopropanol in hexane over 30 minutes at flow rate 1.0 mL/min, UV detection at 220 nm); MS (ES): m/z 340 [M+1]+
  • Example 3 1-(5-Chloro-6-cyano-pyridin-3-ylcarbamoyl)-3-hydroxypyrrolidine-2-carboxylic acid methyl ester
  • Figure US20080108691A1-20080508-C00020
  • Step 1. 5-Amino-3-chloropyridine-2-carbonitrile
  • Figure US20080108691A1-20080508-C00021
  • To a solution of 3-chloro-5-nitro-pyridine-2-carbonitrile (0.1 g, 0.549 mmol) in 50% EtOAc and 40% H2O and 10% HOAc was added iron powder (0.1 g). The reaction mixture was stirred at 60° C. for 5 min until the amine was formed. The mixture was washed with H2O and the EtOAc was removed under reduced pressure. The crude product was purified by chromatography, eluted with 30% EtOAc in hexane and 2% MeOH in EtOAc/hexane to afford the title compound (0.66 g) as a white solid.
  • Step 2. 3-Chloro-5-isocyanatopyridine-2-carbonitrile
  • Figure US20080108691A1-20080508-C00022
  • To a stirring suspension of the Step 1 compound (0.066 g, 0.43 mmol) and Et3N (0.09 mL, 0.86 mmol) in THF (4 mL) cooled to 0° C. was added a solution of phosgene (20%) in toluene (0.085 mL, 0.86 mmol). After addition, the mixture was stirred at rt for 10 min. The mixture was then concentrated under reduced pressure, the resulting solid residue dried in vacuo for 1 h to afford compound 3B (0.065 g) as a light yellow solid.
  • Step 3. 1-(5-Chloro-6-cyanopyridin-3-ylcarbamoyl)-3-hydroxypyrrolidine-2-carboxylic acid methyl ester
  • A solution of cis-3-hydroxyproline methyl ester, HCl salt (0.14 g, 0.55 mmol) in MeOH (10 mL) was added WA21J resin (0.5 g). The resulting suspension was stirred at rt for 30 min, and then filtered. The collected resin was rinsed with MeOH (2×) and combined filtrate concentrated carefully under reduced pressure to give the corresponding free base (0.074 g) as a colorless oil. To a suspension of cis-3-hydroxyproline (0.074 g, 0.51 mmol) in CH2Cl2 (2 mL) was added the Step 2 compound (0.065 g, 0.36 mmol) in toluene (1 mL) solution, along with 4 Å molecular sieves (0.5 g) and the resulting mixture stirred at rt until urea formation was complete (˜1 d). The reaction mixture was loaded on a silica gel column, eluted with 50% EtOAc/hexane, and 5% MeOH in EtOAc/hexane (1:1) to afford 30 mg of the title compound as a white solid. HPLC: 99% at 1.51 min (retention time) (Conditions: Phenom. Luna C18 (4.6×50 mm); Eluted with 0% to 100% B; 4 min gradient (A=90% H2O−10% CH3CN−0.1% TFA and B=10% H2O−90% CH3CN−0.1% TFA), Flow rate at 4 mL/min., UV detection at 220 nm). Chiral HPLC: retention time=8.45 min (99%); Conditions: (CHIRALPAK® OD column 4.6×250 mm; 25% isopropanol in hexane over 30 minutes at flow rate 1.0 mL/min, UV detection at 220 nm); MS (ES): m/z 325 [M+1]+
  • Example 4 1-[2-(4-Cyanonaphthalen-1-yl)acetyl]-3-hydroxypyrrolidine-2-carboxylic acid methyl ester
  • Figure US20080108691A1-20080508-C00023
  • Step 1. (4-Cyanonaphthalen-1-yl)acetic acid methyl ester
  • Figure US20080108691A1-20080508-C00024
  • A solution of 4-methyl-1-cyanonaphthalene (4.35 g, 25.9 mmol) in THF (100 mL) was cooled to −78° C. and treated with commercially available LDA (2 M, 60 mL). The reaction mixture was then warmed to 0° C. and maintained at 0° C. for 30 min. The resulting red-colored solution was cooled to −78° C. and methyl chloroformate (10 mL, 130 mmol) was added. The reaction mixture was stirred while warming to rt until the displacement was complete (4 h) and then quenched with sat'd NH4Cl. The THF layer was evaporated and extracted with EtOAc (3×20 mL). The EtOAc was dried over Na2SO4 and concentrated in vacuo to afford an oil.
  • Step 2. (4-Cyanonaphthalen-1-yl)acetic acid
  • Figure US20080108691A1-20080508-C00025
  • A solution of the Step 1 compound (6.20 g, 26.7 mmol) in MeOH (100 mL) was treated with saturated aqueous LiOH (15 mL), and the reaction mixture was stirred at rt overnight (˜16 h). The resulting mixture was concentrated in vacuo to dryness and partitioned between a mixed solvent (50% Et2O:50% EtOAc) and H2O. The H2O layer was acidified to pH 2-3 and extracted with EtOAc (3×20 mL). The EtOAc was dried over Na2SO4 and concentrated in vacuo to give a yellow solid (2.1 g) in 38% yield.
  • Step 3. 1-[2-(4-Cyanonaphthalen-1-yl)acetyl]-3-hydroxypyrrolidine-2-carboxylic acid methyl ester
  • To a solution of the Step 2 compound (1.37 g, 5.3 mmol) in DMF (30 mL) was added HOBT (1.94 g, 14.4 mmol), EDAC (1.85 g, 9.6 mmol), followed by TEA (2 mL, 14.4 mmol), and the reaction was stirred at rt overnight (˜16 h). The resulting mixture was partitioned between EtOAc (100 mL) and H2O (4×50 mL). The EtOAc layer was dried over Na2SO4 and was evaporated in vacuo. The crude product was cleaned up by silica gel chromatography using 10% MeOH in CH2Cl2 to afford 1.01 g of the title compound as an off-white solid. HPLC: 97.5% at 2.99 min (retention time) (Conditions: YMC S5 ODS (4.6×50 mm) Ballistic; Eluted with 0% to 100% B; 4 min gradient (A=90% H2O-10% CH3OH−0.2% H3PO4 and B=10% H2O-90% CH3OH−0.2% H3PO4), Flow rate at 4 mL/min., UV detection at 220 nm). MS (ES): m/z 339 [M+1]+

Claims (8)

1. A compound of the formula I
Figure US20080108691A1-20080508-C00026
or pharmaceutically acceptable salt thereof wherein
R1 is selected from the group consisting of hydrogen, alkyl or substituted alkyl, alkenyl or substituted alkenyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl and CH2OR4;
R2 is selected from the group consisting of hydrogen, alkyl or substituted alkyl, alkenyl or substituted alkenyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo, heteroaryl or substituted heteroaryl and CH2OR4;
R3 is selected from the group consisting of hydrogen, alkyl or substituted alkyl, CH2OR4, OR2, SR2, halo, NHR2, NHCOR4, NHCO2R4, NHCONR4R4′ and NHSO2R4;
R4 and R4′ for each occurrence are each independently selected from the group consisting of hydrogen, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heterocyclo or substituted heterocyclo and heteroaryl or substituted heteroaryl;
G is a mono- or polycyclic ring system selected from the group consisting of aryl, heterocyclo and heteroaryl, wherein said ring system may optionally substituted with one or more substituents selected from the group consisting of hydrogen, halo, CN, CF3, OR4, CO2R4, NR4R4′, CONR4R4′, CH2OR4, SR4, SOR4, SO2R4, NO2, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
X is a linking group selected from the group consisting of NR4 and CHR4;
Y is selected from the group consisting of O, NR4, NOR4, S and CH2;
z is —O— or NR4; and
n is 2;
with the following provisos:
(a) when Y is NOR4, R4 is not hydrogen;
(b) the following do not occur simultaneously: R1 is methyl; X is NH; Y is O or S;
and Z is O; and
(c) the following do not occur simultaneously:
R1 is methyl;
X is NH;
Z is O;
Y is NR4;
R4 is selected from the group consisting of hydrogen, alkyl or substituted alkyl, alkenyl or substituted alkenyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl; and
G has the following structure:
Figure US20080108691A1-20080508-C00027
wherein
R13 is selected from the group consisting of hydrogen, cyano (—CN), nitro (—NO2), halo, heterocyclo, OR14, CO2R15, CONHR15, COR15, S(O)pR15, SO2NR15R15′, NHCOR15 and NHSO2R15;
R14 in each functional group is independently selected from the group consisting of hydrogen, alkyl or substituted alkyl, CHF2, CF3 and COR15;
R15 and R15′ in each functional group are each independently selected from the group consisting of hydrogen, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, heterocycloalkyl or substituted heterocycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl and —CN;
A and B are each independently selected from the group consisting of hydrogen, halo, cyano(—CN), nitro(—NO2), alkyl or substituted alkyl and OR14; and
p is an integer from 0 to 2.
2. A compound or salt thereof as defined in claim 1 wherein G is selected from:
Figure US20080108691A1-20080508-C00028
wherein
R8, R9, R10 and R11 are each independently selected from the group consisting of hydrogen (H), NO2, CN, CF3, OR4, CO2R4, NR4′, CONR4R4′, CH2OR4, alkyl or substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, cycloalkyl or substituted cycloalkyl, arylalkyl or substituted arylalkyl, aryl or substituted aryl and heteroaryl or substituted heteroaryl;
A to F is each independently selected from N or CR1;
J, K, L, P and Q are each independently selected from NR12, O, S, SO, SO2 or CR12R12′;
R12 and R12′ in each functional group are each independently selected from a bond or R1; and
m is an integer of 0 or 1.
3. A compound or salt thereof as defined in claim 2 wherein R8 is CN.
4. A pharmaceutical composition comprising the compound or salt as defined in claim 1 and a pharmaceutically acceptable carrier therefore.
5. A pharmaceutical composition as defined in claim 4 further comprising a growth promoting agent.
6. A pharmaceutical composition comprising a compound or salt as defined in claim 1 and at least one additional therapeutic agent selected from the group consisting of parathyroid hormone, bisphosphonates, estrogen, testosterone, progesterone, selective estrogen receptor modulators, growth hormone secretagogues, growth hormone, progesterone receptor modulators, anti-diabetic agents, anti-hypertensive agents, anti-inflammatory agents, anti-osteoporosis agents, anti-obesity agents, cardiac glycosides, cholesterol lowering agents, anti-depressants, anti-anxiety agents, anabolic agents, and thyroid mimetics.
7. A method for treating or delaying the progression or onset of muscular atrophy, lipodistrophy, long-term critical illness, sarcopenia, frailty or age-related functional decline, reduced muscle strength and function, reduced bone density or growth, the catabolic side effects of glucocorticoids, chronic fatigue syndrome, bone fracture repair, acute fatigue syndrome and muscle loss following elective surgery, cachexia, chronic catabolic state, eating disorders, side effects of chemotherapy, wasting, depression, nervousness, irritability, stress, growth retardation, reduced cognitive function, male contraception, hypogonadism, Syndrome X, diabetic complications or obesity, which comprises administering to a mammalian species in need of treatment a therapeutically effective amount of a compound or salt as defined in claim 1.
8. A method according to claim 8 further comprising administering, concurrently or sequentially, a therapeutically effective amount of at least one additional therapeutic agent selected from the group consisting of other compounds or salts of formula I, parathyroid hormone, bisphosphonates, estrogen, testosterone, progesterone, selective estrogen receptor modulators, growth hormone secretagogues, growth hormone, progesterone receptor modulators, anti-diabetic agents, anti-hypertensive agents, anti-inflammatory agents, anti-osteoporosis agents, anti-obesity agents, cardiac glycosides, cholesterol lowering agents, anti-depressants, anti-anxiety agents, anabolic agents and thyroid mimetics.
US11/931,498 2002-11-15 2007-10-31 Open-chain prolyl urea-related modulators of androgen receptor function Abandoned US20080108691A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/931,498 US20080108691A1 (en) 2002-11-15 2007-10-31 Open-chain prolyl urea-related modulators of androgen receptor function

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US42669402P 2002-11-15 2002-11-15
US10/712,456 US7632858B2 (en) 2002-11-15 2003-11-13 Open chain prolyl urea-related modulators of androgen receptor function
US11/931,498 US20080108691A1 (en) 2002-11-15 2007-10-31 Open-chain prolyl urea-related modulators of androgen receptor function

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/712,456 Division US7632858B2 (en) 2002-11-15 2003-11-13 Open chain prolyl urea-related modulators of androgen receptor function

Publications (1)

Publication Number Publication Date
US20080108691A1 true US20080108691A1 (en) 2008-05-08

Family

ID=32326402

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/712,456 Active 2025-12-05 US7632858B2 (en) 2002-11-15 2003-11-13 Open chain prolyl urea-related modulators of androgen receptor function
US11/931,498 Abandoned US20080108691A1 (en) 2002-11-15 2007-10-31 Open-chain prolyl urea-related modulators of androgen receptor function

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/712,456 Active 2025-12-05 US7632858B2 (en) 2002-11-15 2003-11-13 Open chain prolyl urea-related modulators of androgen receptor function

Country Status (4)

Country Link
US (2) US7632858B2 (en)
EP (1) EP1567487A4 (en)
AU (1) AU2003302084A1 (en)
WO (1) WO2004045518A2 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050059652A1 (en) * 2002-11-15 2005-03-17 Hamann Lawrence G. Open chain prolyl urea-related modulators of androgen receptor function
WO2017035401A1 (en) * 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
WO2017035405A1 (en) * 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US9796741B2 (en) 2014-02-25 2017-10-24 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of medical disorders
US10011612B2 (en) 2015-08-26 2018-07-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10071448B1 (en) 2010-10-11 2018-09-11 Blendtec, Inc. Method for making a mixing blade for blending apparatus
US10092584B2 (en) 2015-08-26 2018-10-09 Achillion Pharmaceuticals, Inc. Compounds for the treatment of medical disorders
US10138225B2 (en) 2015-08-26 2018-11-27 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of medical disorders
US10385097B2 (en) 2015-08-26 2019-08-20 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
US10660876B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of medical disorders
US10662175B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US10807952B2 (en) 2015-08-26 2020-10-20 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune inflammatory disorders
US11001600B2 (en) 2015-08-26 2021-05-11 Achillion Pharmaceuticals, Inc. Disubstituted compounds for treatment of medical disorders
US11053253B2 (en) 2017-03-01 2021-07-06 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11807627B2 (en) 2018-09-25 2023-11-07 Achillon Pharmaceuticals, Inc. Morphic forms of complement factor D inhibitors
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders
US11814363B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Morphic forms of danicopan

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7026484B2 (en) 2001-02-23 2006-04-11 Ligand Pharmaceuticals Incorporated Tricyclic androgen receptor modulator compounds and methods
US7405234B2 (en) * 2002-05-17 2008-07-29 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
AR044519A1 (en) 2003-05-02 2005-09-14 Novartis Ag DERIVATIVES OF PIRIDIN-TIAZOL AMINA AND PIRIMIDIN-TIAZOL AMINA
US7816372B2 (en) 2003-08-22 2010-10-19 Ligand Pharmaceuticals Incorporated 6-cycloamino-2-quinolinone derivatives as androgen receptor modulator compounds
US20050182105A1 (en) * 2004-02-04 2005-08-18 Nirschl Alexandra A. Method of using 3-cyano-4-arylpyridine derivatives as modulators of androgen receptor function
US7820702B2 (en) 2004-02-04 2010-10-26 Bristol-Myers Squibb Company Sulfonylpyrrolidine modulators of androgen receptor function and method
US7388027B2 (en) * 2004-03-04 2008-06-17 Bristol-Myers Squibb Company Bicyclic compounds as modulators of androgen receptor function and method
US7625923B2 (en) 2004-03-04 2009-12-01 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US7696241B2 (en) * 2004-03-04 2010-04-13 Bristol-Myers Squibb Company Bicyclic compounds as modulators of androgen receptor function and method
EP1954133A4 (en) * 2005-11-30 2010-11-24 Glaxosmithkline Llc Pyrrolidineanilines
JP5473334B2 (en) 2005-12-23 2014-04-16 ジーランド ファーマ アクティーゼルスカブ Modified lysine mimetic compounds
DK2468724T3 (en) 2006-12-21 2016-02-22 Zealand Pharma As Synthesis of pyrrolidine compounds
UY31545A1 (en) 2007-12-20 2009-08-03 NEW DERIVATIVES OF 2-CARBOXAMIDE CIANOAMINOUREA, ITS SALTS AND PHARMACEUTICALLY ACCEPTABLE PROFARMS, PREPARATION PROCESSES AND APPLICATIONS
US8268872B2 (en) 2008-02-22 2012-09-18 Radius Health, Inc. Selective androgen receptor modulators
CA2716320C (en) 2008-02-22 2014-01-28 Radius Health, Inc. Selective androgen receptor modulators
UA104147C2 (en) 2008-09-10 2014-01-10 Новартис Аг Pyrrolidine dicarboxylic acid derivative and use thereof in the treatment of proliferative diseases
US8293753B2 (en) 2009-07-02 2012-10-23 Novartis Ag Substituted 2-carboxamide cycloamino ureas
CA2766853A1 (en) 2009-07-02 2011-01-06 Novartis Ag 2-carboxamide cycloamino ureas useful as pi3k inhibitors
AU2011212813B2 (en) 2010-02-04 2014-10-23 Radius Health, Inc. Selective androgen receptor modulators
PT2568806T (en) 2010-05-12 2016-08-05 Radius Health Inc Therapeutic regimens
US8642632B2 (en) 2010-07-02 2014-02-04 Radius Health, Inc. Selective androgen receptor modulators
JP5965909B2 (en) 2010-09-28 2016-08-10 ラジウス ヘルス,インコーポレイテッド Selective androgen receptor modulator
US9085555B2 (en) 2011-01-04 2015-07-21 Novartis Ag Complement pathway modulators and uses thereof
WO2012109238A2 (en) 2011-02-07 2012-08-16 President And Fellows Of Harvard College Methods for increasing immune responses using agents that directly bind to and activate ire-1
WO2013134774A1 (en) 2012-03-09 2013-09-12 Cornell University Modulation of breast cancer growth by modulation of xbp1 activity
ES2712190T3 (en) 2012-06-28 2019-05-09 Novartis Ag Modulators of the complement pathway and its uses
WO2014002054A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
EP2867229B1 (en) 2012-06-28 2017-07-26 Novartis AG Pyrrolidine derivatives and their use as complement pathway modulators
CN105121429B (en) 2012-06-28 2017-12-12 诺华股份有限公司 Complement pathway conditioning agent and its purposes
CN104603126B (en) 2012-06-28 2017-05-31 诺华股份有限公司 Pyrrolidin derivatives and its purposes as complement pathway conditioning agent
ES2687983T3 (en) 2012-07-12 2018-10-30 Novartis Ag Modulators of the complement path and uses thereof
WO2015048331A1 (en) 2013-09-25 2015-04-02 Cornell University Compounds for inducing anti-tumor immunity and methods thereof
EP4035664A3 (en) 2014-03-28 2022-11-30 Duke University Treating cancer using selective estrogen receptor modulators
US9421264B2 (en) 2014-03-28 2016-08-23 Duke University Method of treating cancer using selective estrogen receptor modulators
EP3319968A1 (en) 2015-07-06 2018-05-16 Rodin Therapeutics, Inc. Heterobicyclic n-aminophenyl-amides as inhibitors of histone deacetylase
JP6936796B2 (en) 2015-07-06 2021-09-22 ロダン・セラピューティクス,インコーポレーテッド Histone deacetylase heterohalo inhibitor
ES2913470T3 (en) 2016-06-22 2022-06-02 Ellipses Pharma Ltd AR+ breast cancer treatment methods
KR20190036520A (en) 2016-06-27 2019-04-04 아칠리온 파르마세우티칼스 인코포레이티드 Quinazoline and indole compounds for the treatment of medical disorders
KR102322802B1 (en) 2017-01-05 2021-11-04 래디어스 파마슈티컬스, 인코포레이티드 Polymorphic form of RAD1901-2HCL
HRP20220223T1 (en) 2017-01-11 2022-04-29 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
DK3618847T3 (en) 2017-05-05 2021-05-25 Boston Medical Ct Corp GAP junction modulators of intercellular communication and their use in the treatment of diabetic eye disease
RS63343B1 (en) 2017-08-07 2022-07-29 Alkermes Inc Bicyclic inhibitors of histone deacetylase
AU2019297421A1 (en) 2018-07-04 2021-01-28 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD 1901-2HCL

Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3239345A (en) * 1965-02-15 1966-03-08 Estrogenic compounds and animal growth promoters
US3948933A (en) * 1973-11-01 1976-04-06 Gruppo Lepetit S.P.A. Pyrrolo[1,2-c]imidazolediones
US4036979A (en) * 1974-01-25 1977-07-19 American Cyanamid Company Compositions containing 4,5,6,7-tetrahydrobenz[b]thien-4-yl-ureas or derivatives and methods of enhancing growth rate
US4411890A (en) * 1981-04-14 1983-10-25 Beckman Instruments, Inc. Synthetic peptides having pituitary growth hormone releasing activity
US4959361A (en) * 1987-12-18 1990-09-25 Hoffmann-La Roche Inc. Triazolo(4,3-A)(1,4)benzodiazepines and thieno (3,2-F)(1,2,4)triazolo(4,3-A)(1,4)diazepine compounds which have useful activity as platelet activating factor (PAF) antagonists
US5114945A (en) * 1988-02-23 1992-05-19 Glaxo Group Limited Spiropiperidine derivatives
US5179080A (en) * 1989-08-31 1993-01-12 Clinical Homecare, Corp. Formulations containing growth hormone and nutritional supplements, and methods of treating malnutrition in chronic lung disease
US5403817A (en) * 1990-07-27 1995-04-04 Sandoz Ltd. Arylaminocarbonyl compounds
US5482921A (en) * 1990-12-18 1996-01-09 Sandoz Ltd. Hydantoin compounds
US5488064A (en) * 1994-05-02 1996-01-30 Bristol-Myers Squibb Company Benzo 1,3 dioxole derivatives
US5491134A (en) * 1994-09-16 1996-02-13 Bristol-Myers Squibb Company Sulfonic, phosphonic or phosphiniic acid β3 agonist derivatives
US5541204A (en) * 1994-12-02 1996-07-30 Bristol-Myers Squibb Company Aryloxypropanolamine β 3 adrenergic agonists
US5556909A (en) * 1994-12-02 1996-09-17 Sanofi Winthrop Inc. Substituted 2-arylcarbonyloxymethyl-1,2,5-thiadiazolidin-3-one 1,1-dioxide derivatives and compositions and method of use thereof
US5605877A (en) * 1992-09-10 1997-02-25 E. I. Du Pont De Nemours And Company Bicyclic imides as herbicides
US5612359A (en) * 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
US5688810A (en) * 1994-12-22 1997-11-18 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5770615A (en) * 1996-04-04 1998-06-23 Bristol-Myers Squibb Company Catecholamine surrogates useful as β3 agonists
US5776983A (en) * 1993-12-21 1998-07-07 Bristol-Myers Squibb Company Catecholamine surrogates useful as β3 agonists
US5811374A (en) * 1991-02-07 1998-09-22 Bayer Aktiengesellschaft 3-aryl-pyrrolidine-2,4-dione derivatives
US6011029A (en) * 1996-02-26 2000-01-04 Bristol-Myers Squibb Company Inhibitors of farnesyl protein transferase
US6040321A (en) * 1997-11-12 2000-03-21 Bristol-Myers Squibb Company Aminothiazole inhibitors of cyclin dependent kinases
US6043265A (en) * 1997-01-30 2000-03-28 Bristol-Myers Squibb Co. Isoxazolyl endothelin antagonists
US6184231B1 (en) * 1998-12-04 2001-02-06 Bristol-Myers Squibb 3-substituted-4-arylquinolin-2-one derivatives as potassium channel modulators
US6310095B1 (en) * 1995-11-06 2001-10-30 University Of Pittsburgh Inhibitors of protein isoprenyl transferases
US6365615B1 (en) * 1999-07-21 2002-04-02 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
US20020133004A1 (en) * 1999-09-03 2002-09-19 Ajinomoto Co. Inc Process for producing new oxazepine derivatives
US6531612B2 (en) * 1999-12-24 2003-03-11 Hoffman-La Roche Inc. Nitrile derivatives that inhibit cathepsin K
US6566367B2 (en) * 2000-12-12 2003-05-20 Pfizer Inc. Spiro[isobenzofuran-1,4′-piperidin]-3-ones and 3H-spiroisobenzofuran-1,4′-piperidines
US6670386B2 (en) * 2001-07-31 2003-12-30 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US20040019063A1 (en) * 2002-05-17 2004-01-29 Chongqing Sun Bicyclic modulators of androgen receptor function
US6710064B2 (en) * 2000-12-01 2004-03-23 Bristol-Myers Squibb Co. Hydantoin compounds useful as anti-inflammatory agents
US20040181064A1 (en) * 2002-05-17 2004-09-16 Chong-Qing Sun Bicyclic modulators of androgen receptor function
US6794823B2 (en) * 1997-03-31 2004-09-21 Mitsubishi Denki Kabushiki Kaisha Planar display panel controller
US20050059652A1 (en) * 2002-11-15 2005-03-17 Hamann Lawrence G. Open chain prolyl urea-related modulators of androgen receptor function
US6897225B1 (en) * 1999-10-20 2005-05-24 Tanabe Seiyaku Co., Ltd. Inhibitors of αLβ2 mediated cell adhesion
US20050182105A1 (en) * 2004-02-04 2005-08-18 Nirschl Alexandra A. Method of using 3-cyano-4-arylpyridine derivatives as modulators of androgen receptor function
US20050187267A1 (en) * 2004-02-04 2005-08-25 Hamann Lawrence G. Sulfonylpyrrolidine modulators of androgen receptor function and method
US20050197367A1 (en) * 2004-03-04 2005-09-08 Li James J. Novel bicyclic compounds as modulators of androgen receptor function and method
US20050197359A1 (en) * 2004-03-04 2005-09-08 Alexandra Nirschl Bicyclic modulators of androgen receptor function
US6974823B2 (en) * 1999-12-21 2005-12-13 Gpi Nil Holdindgs, Inc. Hydantoin derivative compounds, pharmaceutical compositions, and methods of using same
US20070004717A1 (en) * 2004-03-04 2007-01-04 Li James J Novel bicyclic compounds as modulators of androgen receptor function and method
US7256208B2 (en) * 2003-11-13 2007-08-14 Bristol-Myers Squibb Company Monocyclic N-Aryl hydantoin modulators of androgen receptor function

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS4920973B1 (en) 1970-05-28 1974-05-29
CA1076114A (en) 1975-02-10 1980-04-22 Mitsubishi Chemical Industries Limited 1,5-alkylene-3-aryl hydantoin derivatives
JPS5933593B2 (en) 1976-01-01 1984-08-16 三菱化学株式会社 Derivatives of hydantoin or thiohydantoin
DE3643748A1 (en) 1986-12-20 1988-06-30 Hoechst Ag BICYLIC IMIDES, METHODS FOR THEIR PRODUCTION AND THEIR USE IN PLANT PROTECTION
AU637316B2 (en) 1988-01-28 1993-05-27 Eastman Kodak Company Polypeptide compounds having growth hormone releasing activity
WO1989007111A1 (en) 1988-01-28 1989-08-10 Eastman Kodak Company Polypeptide compounds having growth hormone releasing activity
DE3809390A1 (en) 1988-03-16 1989-09-28 Schering Ag Perhydro-imidazopyridines and -pyrroloimidazoles, processes for their preparation, and their use as herbicides
US5144945A (en) * 1989-04-20 1992-09-08 Nippon Sanso Kabushiki Kaisha Portable oxygen-enriching air inhaler
US5663146A (en) 1991-08-22 1997-09-02 Administrators Of The Tulane Educational Fund Polypeptide analogues having growth hormone releasing activity
DE4138042C2 (en) 1991-11-19 1993-10-14 Biotechnolog Forschung Gmbh Epothilones, their production processes and agents containing these compounds
IL107719A0 (en) 1992-12-21 1994-02-27 Du Pont Imidazolones their manufacture and their use as herbicides
EP1041066A1 (en) 1994-12-22 2000-10-04 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
DE59609305D1 (en) 1995-11-17 2002-07-11 Biotechnolog Forschung Gmbh Epothilone derivatives and their production
US6236946B1 (en) 1995-12-13 2001-05-22 Thomas S. Scanlan Nuclear receptor ligands and ligand binding domains
ES2168650T3 (en) 1996-06-27 2002-06-16 Ligand Pharm Inc COMPOUNDS AND MODULATING METHODS OF ANDROGEN RECEPTORS.
WO1998022461A1 (en) 1996-11-18 1998-05-28 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilone c, d, e and f, production process, and their use as cytostatic as well as phytosanitary agents
CA2273083C (en) 1996-12-03 2012-09-18 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6380394B1 (en) 1996-12-13 2002-04-30 The Scripps Research Institute Epothilone analogs
US6441186B1 (en) 1996-12-13 2002-08-27 The Scripps Research Institute Epothilone analogs
HU228851B1 (en) 1997-02-25 2013-06-28 Biotechnolog Forschung Gmbh Process for preparing epothilones with a modified side chain and compounds prepared by this process
GB9801231D0 (en) 1997-06-05 1998-03-18 Merck & Co Inc A method of treating cancer
GB9713739D0 (en) 1997-06-27 1997-09-03 Karobio Ab Thyroid receptor ligands
WO1999002224A1 (en) 1997-07-07 1999-01-21 Lewis Robert D Statistical analysis and feedback system for sports employing a projectile
US6605599B1 (en) 1997-07-08 2003-08-12 Bristol-Myers Squibb Company Epothilone derivatives
EP1001951B1 (en) 1997-07-16 2002-09-25 Schering Aktiengesellschaft Thiazole derivatives, method for their production and use
ATE368036T1 (en) 1997-08-09 2007-08-15 Bayer Schering Pharma Ag NEW EPOTHILONE DERIVATIVES, PROCESS FOR THEIR PRODUCTION AND THEIR PHARMACEUTICAL USE
US6365749B1 (en) 1997-12-04 2002-04-02 Bristol-Myers Squibb Company Process for the preparation of ring-opened epothilone intermediates which are useful for the preparation of epothilone analogs
AU738576B2 (en) 1997-12-04 2001-09-20 Bristol-Myers Squibb Company A process for the reduction of oxiranyl epothilones to olefinic epothilones
US6203870B1 (en) 1998-02-24 2001-03-20 Plastipak Packaging, Inc. Liner and preform
US6498257B1 (en) 1998-04-21 2002-12-24 Bristol-Myers Squibb Company 2,3-olefinic epothilone derivatives
US6399638B1 (en) 1998-04-21 2002-06-04 Bristol-Myers Squibb Company 12,13-modified epothilone derivatives
AU743199B2 (en) 1998-06-02 2002-01-24 Bristol-Myers Squibb Company Neurotrophic difluoroamide agents
DE19826988A1 (en) 1998-06-18 1999-12-23 Biotechnolog Forschung Gmbh Epothilone minor components
AU4775299A (en) 1998-06-22 2000-01-10 Nicolaou, Kyriacos Costa Desmethyl epothilones
US6228872B1 (en) * 1998-11-12 2001-05-08 Bristol-Myers Squibb Company Neurotrophic diamide and carbamate agents
US6297380B1 (en) 1998-11-23 2001-10-02 Pfizer Inc. Process and intermediates for growth hormone secretagogues
CA2366264A1 (en) * 1999-04-02 2000-10-12 Bristol-Myers Squibb Company Novel amide derivatives as inhibitors of matrix metalloproteinases, tnf-.alpha., and aggrecanase
US6548529B1 (en) * 1999-04-05 2003-04-15 Bristol-Myers Squibb Company Heterocyclic containing biphenyl aP2 inhibitors and method
BR0014651A (en) * 1999-10-20 2002-06-18 Tanabe Seiyaku Co Beta2-mediated cell adhesion inhibitors
DOP2000000107A (en) * 1999-12-01 2002-09-16 Agouron Pharmaceutical Inc COMPOUNDS, COMPOSITIONS AND METHODS TO STIMULATE THE GROWTH AND LENGTH OF NEURONS
WO2001054503A1 (en) * 2000-01-28 2001-08-02 Akkadix Corporation Methods for killing nematodes and nematode eggs using 4-phenoxy-6-aminopyrimidine derivatives
EP1125925A1 (en) * 2000-02-15 2001-08-22 Applied Research Systems ARS Holding N.V. Amine derivatives for the treatment of apoptosis
US6395767B2 (en) * 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
AU2001239896A1 (en) * 2000-03-16 2001-10-03 Sepracor, Inc. Peptidomimetic ligands for cellular receptors and ion channels
UA74362C2 (en) * 2000-03-27 2005-12-15 Еплайд Рісьорч Системз Ерс Холдінг Н.В. Pharnaceutically active derivatives of pyrrolidine
JP2002047272A (en) 2000-07-26 2002-02-12 Dai Ichi Seiyaku Co Ltd Polyamine amide derivative
WO2002018335A1 (en) * 2000-08-28 2002-03-07 Yamanouchi Pharmaceutical Co., Ltd. Cyclic amine derivatives
US6573287B2 (en) * 2001-04-12 2003-06-03 Bristo-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase IV and method
US7265145B2 (en) * 2003-05-28 2007-09-04 Bristol-Myers Squibb Company Substituted piperidines and pyrrolidines as calcium sensing receptor modulators and method

Patent Citations (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3239345A (en) * 1965-02-15 1966-03-08 Estrogenic compounds and animal growth promoters
US3948933A (en) * 1973-11-01 1976-04-06 Gruppo Lepetit S.P.A. Pyrrolo[1,2-c]imidazolediones
US4036979A (en) * 1974-01-25 1977-07-19 American Cyanamid Company Compositions containing 4,5,6,7-tetrahydrobenz[b]thien-4-yl-ureas or derivatives and methods of enhancing growth rate
US4411890A (en) * 1981-04-14 1983-10-25 Beckman Instruments, Inc. Synthetic peptides having pituitary growth hormone releasing activity
US4959361A (en) * 1987-12-18 1990-09-25 Hoffmann-La Roche Inc. Triazolo(4,3-A)(1,4)benzodiazepines and thieno (3,2-F)(1,2,4)triazolo(4,3-A)(1,4)diazepine compounds which have useful activity as platelet activating factor (PAF) antagonists
US5114945A (en) * 1988-02-23 1992-05-19 Glaxo Group Limited Spiropiperidine derivatives
US5179080A (en) * 1989-08-31 1993-01-12 Clinical Homecare, Corp. Formulations containing growth hormone and nutritional supplements, and methods of treating malnutrition in chronic lung disease
US5403817A (en) * 1990-07-27 1995-04-04 Sandoz Ltd. Arylaminocarbonyl compounds
US5482921A (en) * 1990-12-18 1996-01-09 Sandoz Ltd. Hydantoin compounds
US5811374A (en) * 1991-02-07 1998-09-22 Bayer Aktiengesellschaft 3-aryl-pyrrolidine-2,4-dione derivatives
US5605877A (en) * 1992-09-10 1997-02-25 E. I. Du Pont De Nemours And Company Bicyclic imides as herbicides
US5776983A (en) * 1993-12-21 1998-07-07 Bristol-Myers Squibb Company Catecholamine surrogates useful as β3 agonists
US5488064A (en) * 1994-05-02 1996-01-30 Bristol-Myers Squibb Company Benzo 1,3 dioxole derivatives
US5612359A (en) * 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
US5491134A (en) * 1994-09-16 1996-02-13 Bristol-Myers Squibb Company Sulfonic, phosphonic or phosphiniic acid β3 agonist derivatives
US5556909A (en) * 1994-12-02 1996-09-17 Sanofi Winthrop Inc. Substituted 2-arylcarbonyloxymethyl-1,2,5-thiadiazolidin-3-one 1,1-dioxide derivatives and compositions and method of use thereof
US5541204A (en) * 1994-12-02 1996-07-30 Bristol-Myers Squibb Company Aryloxypropanolamine β 3 adrenergic agonists
US5696133A (en) * 1994-12-22 1997-12-09 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5696130A (en) * 1994-12-22 1997-12-09 Ligand Pharmaceuticals Incorporated Tricyclic steroid receptor modulator compounds and methods
US5696127A (en) * 1994-12-22 1997-12-09 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5688808A (en) * 1994-12-22 1997-11-18 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5693647A (en) * 1994-12-22 1997-12-02 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5693646A (en) * 1994-12-22 1997-12-02 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US5688810A (en) * 1994-12-22 1997-11-18 Ligand Pharmaceuticals Incorporated Steroid receptor modulator compounds and methods
US6310095B1 (en) * 1995-11-06 2001-10-30 University Of Pittsburgh Inhibitors of protein isoprenyl transferases
US6011029A (en) * 1996-02-26 2000-01-04 Bristol-Myers Squibb Company Inhibitors of farnesyl protein transferase
US5770615A (en) * 1996-04-04 1998-06-23 Bristol-Myers Squibb Company Catecholamine surrogates useful as β3 agonists
US6043265A (en) * 1997-01-30 2000-03-28 Bristol-Myers Squibb Co. Isoxazolyl endothelin antagonists
US6794823B2 (en) * 1997-03-31 2004-09-21 Mitsubishi Denki Kabushiki Kaisha Planar display panel controller
US6040321A (en) * 1997-11-12 2000-03-21 Bristol-Myers Squibb Company Aminothiazole inhibitors of cyclin dependent kinases
US6184231B1 (en) * 1998-12-04 2001-02-06 Bristol-Myers Squibb 3-substituted-4-arylquinolin-2-one derivatives as potassium channel modulators
US6365615B1 (en) * 1999-07-21 2002-04-02 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
US20020133004A1 (en) * 1999-09-03 2002-09-19 Ajinomoto Co. Inc Process for producing new oxazepine derivatives
US6897225B1 (en) * 1999-10-20 2005-05-24 Tanabe Seiyaku Co., Ltd. Inhibitors of αLβ2 mediated cell adhesion
US6974823B2 (en) * 1999-12-21 2005-12-13 Gpi Nil Holdindgs, Inc. Hydantoin derivative compounds, pharmaceutical compositions, and methods of using same
US6531612B2 (en) * 1999-12-24 2003-03-11 Hoffman-La Roche Inc. Nitrile derivatives that inhibit cathepsin K
US6710064B2 (en) * 2000-12-01 2004-03-23 Bristol-Myers Squibb Co. Hydantoin compounds useful as anti-inflammatory agents
US6566367B2 (en) * 2000-12-12 2003-05-20 Pfizer Inc. Spiro[isobenzofuran-1,4′-piperidin]-3-ones and 3H-spiroisobenzofuran-1,4′-piperidines
US6992102B2 (en) * 2001-07-31 2006-01-31 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US6670386B2 (en) * 2001-07-31 2003-12-30 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US20040181064A1 (en) * 2002-05-17 2004-09-16 Chong-Qing Sun Bicyclic modulators of androgen receptor function
US20040019063A1 (en) * 2002-05-17 2004-01-29 Chongqing Sun Bicyclic modulators of androgen receptor function
US20050059652A1 (en) * 2002-11-15 2005-03-17 Hamann Lawrence G. Open chain prolyl urea-related modulators of androgen receptor function
US7256208B2 (en) * 2003-11-13 2007-08-14 Bristol-Myers Squibb Company Monocyclic N-Aryl hydantoin modulators of androgen receptor function
US20050182105A1 (en) * 2004-02-04 2005-08-18 Nirschl Alexandra A. Method of using 3-cyano-4-arylpyridine derivatives as modulators of androgen receptor function
US20050187267A1 (en) * 2004-02-04 2005-08-25 Hamann Lawrence G. Sulfonylpyrrolidine modulators of androgen receptor function and method
US20050197367A1 (en) * 2004-03-04 2005-09-08 Li James J. Novel bicyclic compounds as modulators of androgen receptor function and method
US20050197359A1 (en) * 2004-03-04 2005-09-08 Alexandra Nirschl Bicyclic modulators of androgen receptor function
US20070004717A1 (en) * 2004-03-04 2007-01-04 Li James J Novel bicyclic compounds as modulators of androgen receptor function and method

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7632858B2 (en) 2002-11-15 2009-12-15 Bristol-Myers Squibb Company Open chain prolyl urea-related modulators of androgen receptor function
US20050059652A1 (en) * 2002-11-15 2005-03-17 Hamann Lawrence G. Open chain prolyl urea-related modulators of androgen receptor function
US10071448B1 (en) 2010-10-11 2018-09-11 Blendtec, Inc. Method for making a mixing blade for blending apparatus
US9796741B2 (en) 2014-02-25 2017-10-24 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10301336B2 (en) 2014-02-25 2019-05-28 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of complement mediated disorders
US9828396B2 (en) 2014-02-25 2017-11-28 Achillion Pharmaceuticals, Inc. Alkyne compounds for treatment of complement mediated disorders
US10464956B2 (en) 2014-02-25 2019-11-05 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10428094B2 (en) 2014-02-25 2019-10-01 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of complement mediated disorders
US10550140B2 (en) 2014-02-25 2020-02-04 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of complement mediated disorders
US10081645B2 (en) 2014-02-25 2018-09-25 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10689409B2 (en) 2014-02-25 2020-06-23 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
US10100072B2 (en) 2014-02-25 2018-10-16 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of complement mediated disorders
US10106563B2 (en) 2014-02-25 2018-10-23 Achillion Pharmaecuticals, Inc. Ether compounds for treatment of complement mediated disorders
US10370394B2 (en) 2014-02-25 2019-08-06 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US10189869B2 (en) 2014-02-25 2019-01-29 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
US10253053B2 (en) 2014-02-25 2019-04-09 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10092584B2 (en) 2015-08-26 2018-10-09 Achillion Pharmaceuticals, Inc. Compounds for the treatment of medical disorders
US10919884B2 (en) 2015-08-26 2021-02-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US10138225B2 (en) 2015-08-26 2018-11-27 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of medical disorders
US10385097B2 (en) 2015-08-26 2019-08-20 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
US10011612B2 (en) 2015-08-26 2018-07-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of medical disorders
WO2017035405A1 (en) * 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US10660876B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of medical disorders
US10662175B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
WO2017035401A1 (en) * 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US10807952B2 (en) 2015-08-26 2020-10-20 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune inflammatory disorders
US10822352B2 (en) 2015-08-26 2020-11-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10906887B2 (en) 2015-08-26 2021-02-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US10287301B2 (en) 2015-08-26 2019-05-14 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US11001600B2 (en) 2015-08-26 2021-05-11 Achillion Pharmaceuticals, Inc. Disubstituted compounds for treatment of medical disorders
US11926617B2 (en) 2015-08-26 2024-03-12 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US11649223B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US11407738B2 (en) 2015-08-26 2022-08-09 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US11649229B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11708351B2 (en) 2017-03-01 2023-07-25 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11718626B2 (en) 2017-03-01 2023-08-08 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
US11053253B2 (en) 2017-03-01 2021-07-06 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders
US11814363B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Morphic forms of danicopan
US11807627B2 (en) 2018-09-25 2023-11-07 Achillon Pharmaceuticals, Inc. Morphic forms of complement factor D inhibitors

Also Published As

Publication number Publication date
US20050059652A1 (en) 2005-03-17
EP1567487A4 (en) 2005-11-16
WO2004045518A2 (en) 2004-06-03
EP1567487A2 (en) 2005-08-31
AU2003302084A1 (en) 2004-06-15
US7632858B2 (en) 2009-12-15
WO2004045518A3 (en) 2004-10-07
AU2003302084A8 (en) 2004-06-15

Similar Documents

Publication Publication Date Title
US7632858B2 (en) Open chain prolyl urea-related modulators of androgen receptor function
US7388027B2 (en) Bicyclic compounds as modulators of androgen receptor function and method
US7989640B2 (en) Sulfonylpyrrolidine modulators of androgen receptor function and method
US7256208B2 (en) Monocyclic N-Aryl hydantoin modulators of androgen receptor function
US7732480B2 (en) Bicyclic compounds as modulators of androgen receptor function and method
US6992102B2 (en) Bicyclic modulators of androgen receptor function
US7625923B2 (en) Bicyclic modulators of androgen receptor function
US20050182105A1 (en) Method of using 3-cyano-4-arylpyridine derivatives as modulators of androgen receptor function
US7772267B2 (en) Bicyclic modulators of androgen receptor function
US20040019063A1 (en) Bicyclic modulators of androgen receptor function
US20050124625A1 (en) Piperazine derivatives and their use as modulators of nuclear hormone receptor function
US20040077606A1 (en) Fused cyclic modulators of nuclear hormone receptor function
US20030114420A1 (en) Fused cyclic modulators of nuclear hormone receptor function
CA2413683A1 (en) Fused cyclic compounds as modulators of nuclear hormone receptor function

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION